1
|
Asadi M, Ghaffari AD, Mohammadhasani F. In silico analysis and structural vaccinology prediction of Toxoplasma gondii ROP41 gene via immunoinformatics methods as a vaccine candidate. Curr Res Transl Med 2025; 73:103475. [PMID: 39461097 DOI: 10.1016/j.retram.2024.103475] [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: 07/23/2024] [Revised: 09/10/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
INTRODUCTION Toxoplasma gondii (T. gondii) infects all warm-blooded animals, including humans. Currently, no effective treatments exist to prevent the generation of chronic tissue cysts in infected hosts. Therefore, developing a vaccine to protect to deal with toxoplasmosis is a promising strategy, as a single immunization could provide lifelong protective immunity. Rhoptry proteins (ROPs) play a vital role for the parasite's survival within host cells and perform critical functions during different phases of parasite invasion. Little is known about ROP41 gene. Nevertheless, Understanding the characteristics of ROP41 will enhance diagnostic and vaccine research. MATERIALS AND METHODS The current article provides a comprehensive analysis of the essential components of the ROP41 protein, including its transmembrane domain, physico-chemical properties, subcellular location, tertiary and secondary structures, and potential T- and B-cell epitopes. These features were determined by many bioinformatics approaches to identify possible epitopes for developing a highly effective vaccine. RESULTS ROP41 protein showed 36 possible post-translational modification regions. The ROP41 protein secondary structure contains 17.35 % extended strand, 33.47 % alpha-helix, and 49.18 % random coil. Also, ROP41 showed many possible B- and T-cell epitopes. According to the Ramachandran plot, 90.78 % of amino acid residues had been placed in favored, 3.28 % in outlier, and 5.94 % in allowed areas. Also, the allergenicity and antigenicity evaluation indicated that ROP41 is non-allergenic and immunogenic. CONCLUSION The current study offered critical basic and conceptual information on ROP41 to increase a successful vaccine in opposition to continual and acute toxoplasmosis for in addition in vivo assessments. Further research is necessary for the development of vaccines utilizing ROP41 alone or combined with various antigens.
Collapse
Affiliation(s)
- Masoumeh Asadi
- Student Research Committee, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Ali Dalir Ghaffari
- Department of Parasitology and Mycology, Faculty of Medicine, Shahed University, Tehran, Iran.
| | | |
Collapse
|
2
|
Zhang Z, Chen G, Li N, Li Y, Sirimanapong W, Chen J, Xia L. Development and evaluation of immunological effects of a DNA vaccine encoding phosphoketolase family protein against Nocardia seriolae in hybrid snakehead. FISH & SHELLFISH IMMUNOLOGY 2024; 152:109773. [PMID: 39019124 DOI: 10.1016/j.fsi.2024.109773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 07/05/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024]
Abstract
Fish nocardiosis is a chronic disease mainly caused by Nocardia seriolae, which occurs in a variety of economically cultured freshwater and marine fish. Studies have shown that DNA vaccine is an effective treatment to protect fish from bacterial infection. In our previous experiment, an in vivo-induced gene of N. seriolae, encoding phosphoketolase (PK) family protein, was identified by in vivo-induced antigen technology. In the present study, the antigenic gene encoding PK family protein was analyzed by bioinformatics and further inserted into the eukaryotic expression vector pcDNA3.1-myc-his-A for DNA vaccine development. The immunological effects of pcDNA-PK DNA vaccine were assessed in hybrid snakehead (Channa maculata ♀ × Channa argus ♂), showing induction in several serum enzyme activity parameters (including LZM, SOD, ACP and AKP), increasing in specific-antibody IgM levels, as well as up-regulation in six immune-related genes (CD4, CD8α, TNFα, IL-1β, MHCIα and MHCIIα). Moreover, an immune-protection with a relative survival rate was provided at 53.82 % following artificial challenge with N. seriolae in vaccinated fish in comparison to the control group. In summary, these results indicate that pcDNA-PK DNA vaccine could boost strong immune responses in hybrid snakehead and show preferably protective efficacy against N. seriolae, which may be applied in aquaculture to control fish nocardiosis.
Collapse
Affiliation(s)
- Ziwen Zhang
- Fisheries College of Guangdong Ocean University, Shenzhen Institute of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong, China
| | - Guoquan Chen
- Fisheries College of Guangdong Ocean University, Shenzhen Institute of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong, China
| | - Na Li
- Fisheries College of Guangdong Ocean University, Shenzhen Institute of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong, China
| | - Yuhao Li
- Fisheries College of Guangdong Ocean University, Shenzhen Institute of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong, China
| | - Wanna Sirimanapong
- Veterinary Aquatic Animal Research & Health Care Unit, Department of Clinical Sciences and Public Health, Faculty of Veterinary Science, Mahidol University, Nakhon-pathom, Thailand
| | - Jianlin Chen
- Fisheries College of Guangdong Ocean University, Shenzhen Institute of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong, China.
| | - Liqun Xia
- Fisheries College of Guangdong Ocean University, Shenzhen Institute of Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Guangdong, China.
| |
Collapse
|
3
|
Jiang S, Wu S, Zhao G, He Y, Bao L, Liu J, Qin C, Hou J, Ding Y, Cheng A, Jiang B, Wu J, Yan J, Humeau L, Patella A, Weiner DB, Broderick K, Wang B. Comparison of Wild Type DNA Sequence of Spike Protein from SARS-CoV-2 with Optimized Sequence on The Induction of Protective Responses Against SARS-Cov-2 Challenge in Mouse Model. Hum Vaccin Immunother 2022; 18:2016201. [PMID: 35061975 PMCID: PMC8986195 DOI: 10.1080/21645515.2021.2016201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Genetic optimization of Nucleic Acid immunogens is important for potentially improving their immune potency. A COVID-19 DNA vaccine is in phase III clinical trial which is based on a promising highly developable technology platform. Here, we show optimization in mice generating a pGX-9501 DNA vaccine encoding full-length spike protein, which results in induction of potent humoral and cellular immune responses, including neutralizing antibodies, that block hACE2-RBD binding of live CoV2 virus in vitro. Optimization resulted in improved induction of cellular immunity by pGX-9501 as demonstrated by increased IFN-γ expression in both CD8+ and CD4 + T cells and this was associated with more robust antiviral CTL responses compared to unoptimized constructs. Vaccination with pGX-9501 induced subsequent protection against virus challenge in a rigorous hACE2 transgenic mouse model. Overall, pGX-9501 is a promising optimized COVID-19 DNA vaccine candidate inducing humoral and cellular immunity contributing to the vaccine's protective effects.
Collapse
Affiliation(s)
- Sheng Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College (SHMC), Fudan University, Shanghai, China,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Shuting Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College (SHMC), Fudan University, Shanghai, China
| | - Gan Zhao
- Biomedical Research Institute of Advaccine (BRIA), Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou City, Jiangsu
| | - Yue He
- Biomedical Research Institute of Advaccine (BRIA), Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou City, Jiangsu
| | - Linlin Bao
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China
| | - Jiangning Liu
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China
| | - Chuan Qin
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Beijing Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College (PUMC), Beijing, China
| | - Jiawang Hou
- Biomedical Research Institute of Advaccine (BRIA), Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou City, Jiangsu
| | - Yuan Ding
- Biomedical Research Institute of Advaccine (BRIA), Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou City, Jiangsu
| | - Alex Cheng
- Biomedical Research Institute of Advaccine (BRIA), Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou City, Jiangsu
| | - Brian Jiang
- Biomedical Research Institute of Advaccine (BRIA), Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou City, Jiangsu
| | - John Wu
- Biomedical Research Institute of Advaccine (BRIA), Advaccine Biopharmaceutics (Suzhou) Co. LTD, Suzhou City, Jiangsu
| | - Jian Yan
- Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | | | - Ami Patella
- Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | | | | | - Bin Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College (SHMC), Fudan University, Shanghai, China,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China,CONTACT Bin Wang School of Basic Medical Sciences, Fudan University, 131 Dong’an Road, 409 Fuxing Building, Shanghai200032, China
| |
Collapse
|
4
|
Abstract
Leishmaniasis is a zoonotic and vector-borne infectious disease that is caused by the genus Leishmania belonging to the trypanosomatid family. The protozoan parasite has a digenetic life cycle involving a mammalian host and an insect vector. Leishmaniasisis is a worldwide public health problem falling under the neglected tropical disease category, with over 90 endemic countries, and approximately 1 million new cases and 20,000 deaths annually. Leishmania infection can progress toward the development of species–specific pathologic disorders, ranging in severity from self-healing cutaneous lesions to disseminating muco-cutaneous and fatal visceral manifestations. The severity and the outcome of leishmaniasis is determined by the parasite’s antigenic epitope characteristics, the vector physiology, and most importantly, the immune response and immune status of the host. This review examines the nature of host–pathogen interaction in leishmaniasis, innate and adaptive immune responses, and various strategies that have been employed for vaccine development.
Collapse
|
5
|
Abstract
Leishmaniasis is caused by protozoan Leishmania parasites that are transmitted through female sandfly bites. The disease is predominantly endemic to the tropics and semi-tropics and has been reported in more than 98 countries. Due to the side effects of anti-Leishmania drugs and the emergence of drug-resistant isolates, there is currently no encouraging prospect of introducing an effective therapy for the disease. Hence, it seems that the key to disease control management is the introduction of an effective vaccine, particularly against its cutaneous form. Advances in understanding underlying immune mechanisms are feasibale using a variety of candidate antigens, including attenuated live parasites, crude antigens, pure or recombinant Leishmania proteins, Leishmania genes encoding protective proteins, as well as immune system activators from the saliva of parasite vectors. However, there is still no vaccine against different types of human leishmaniasis. In this study, we review the works conducted or being performed in this field.
Collapse
|
6
|
Peng XL, Cheng JSY, Gong HL, Yuan MD, Zhao XH, Li Z, Wei DX. Advances in the design and development of SARS-CoV-2 vaccines. Mil Med Res 2021; 8:67. [PMID: 34911569 PMCID: PMC8674100 DOI: 10.1186/s40779-021-00360-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 11/15/2021] [Indexed: 01/18/2023] Open
Abstract
Since the end of 2019, coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has spread worldwide. The RNA genome of SARS-CoV-2, which is highly infectious and prone to rapid mutation, encodes both structural and nonstructural proteins. Vaccination is currently the only effective method to prevent COVID-19, and structural proteins are critical targets for vaccine development. Currently, many vaccines are in clinical trials or are already on the market. This review highlights ongoing advances in the design of prophylactic or therapeutic vaccines against COVID-19, including viral vector vaccines, DNA vaccines, RNA vaccines, live-attenuated vaccines, inactivated virus vaccines, recombinant protein vaccines and bionic nanoparticle vaccines. In addition to traditional inactivated virus vaccines, some novel vaccines based on viral vectors, nanoscience and synthetic biology also play important roles in combating COVID-19. However, many challenges persist in ongoing clinical trials.
Collapse
Affiliation(s)
- Xue-Liang Peng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi’an, 710069 China
| | - Ji-Si-Yu Cheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi’an, 710069 China
| | - Hai-Lun Gong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi’an, 710069 China
| | - Meng-Di Yuan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi’an, 710069 China
| | - Xiao-Hong Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi’an, 710069 China
| | - Zibiao Li
- Institute of Materials Research and Engineering, A*STAR (Agency for Science, Technology and Research), 2 Fusionopolis Way, Innovis, #08-03, Singapore, 138634 Singapore
| | - Dai-Xu Wei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi’an, 710069 China
| |
Collapse
|
7
|
Duman N, ALzaidi Z, Aynekin B, Taskin D, Demirors B, Yildirim A, Sahin IO, Bilgili F, Turanli ET, Beccari T, Bertelli M, Dundar M. COVID-19 vaccine candidates and vaccine development platforms available worldwide. J Pharm Anal 2021; 11:675-682. [PMID: 34540318 PMCID: PMC8437828 DOI: 10.1016/j.jpha.2021.09.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 09/08/2021] [Accepted: 09/13/2021] [Indexed: 12/12/2022] Open
Abstract
The pandemic caused by the worldwide spread of the coronavirus, which first appeared in 2019, has been named coronavirus disease 19 (COVID-19). More than 4.5 million deaths have been recorded due to the pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), according to the World Health Organization. COVID-19 Dashboard in September 2021. Apart from the wildtype, other variations have been successfully transmitted early in the outbreak although they were not discovered until March 2020. Modifications in the SARS-CoV-2 genetic material, such as mutation and recombination, have the ability to modify the viral life span, along with transitivity, cellular tropism, and symptom severity. Several processes are involved in introducing novel vaccines to the population, including vaccine manufacturing, preclinical studies, Food and Drug Administration permission or certification, processing, and marketing. COVID-19 vaccine candidates have been developed by a number of public and private groups employing a variety of strategies, such as RNA, DNA, protein, and viral vectored vaccines. This comprehensive review, which included the most subsequent evidence on unique features of SARS-CoV-2 and the associated morbidity and mortality, was carried out using a systematic search of recent online databases in order to generate useful knowledge about the COVID-19 updated versions and their consequences on the disease symptoms and vaccine development. The ongoing vaccine studies all over the world against the COVID-19 epidemic have been reviewed. The effects of different vaccine platforms and new variants on vaccine studies were discussed. The effect of vaccines on existing and novel variants was evaluated.
Collapse
Affiliation(s)
- Nilgun Duman
- Department of Medical Genetics, Dragos Hospital Istanbul, Bezmialem Vakif University, Istanbul, 34854, Turkey
| | - Zahraa ALzaidi
- Department of Biotechnology, Faculty of Applied Science, Cukurova University, Adana, 01380, Turkey
| | - Busra Aynekin
- Department of Medical Genetics, Medical Faculty, Erciyes University, Kayseri, 38030, Turkey
| | - Duygu Taskin
- Department of Medical Genetics, Medical Faculty, Erciyes University, Kayseri, 38030, Turkey
| | - Busra Demirors
- Department of Medical Genetics, Medical Faculty, Erciyes University, Kayseri, 38030, Turkey
| | - Abdulbaki Yildirim
- Department of Medical Genetics, Medical Faculty, Erciyes University, Kayseri, 38030, Turkey
| | - Izem Olcay Sahin
- Department of Medical Genetics, Medical Faculty, Erciyes University, Kayseri, 38030, Turkey
| | - Faik Bilgili
- Department of Economics, Faculty of Economics and Administrative Sciences, Erciyes University, Kayseri, 38030, Turkey
| | - Eda Tahir Turanli
- Department of Molecular Biology and Genetics, Faculty of Science, Acibadem University, 34684, Istanbul, Turkey
| | - Tommaso Beccari
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, 06123, Italy
| | | | - Munis Dundar
- Department of Medical Genetics, Medical Faculty, Erciyes University, Kayseri, 38030, Turkey
- Corresponding author.
| |
Collapse
|
8
|
Burgos-Reyes MA, Baylón-Pacheco L, Espíritu-Gordillo P, Galindo-Gómez S, Tsutsumi V, Rosales-Encina JL. Effect of Prophylactic Vaccination with the Membrane-Bound Acid Phosphatase Gene of Leishmania mexicana in the Murine Model of Localized Cutaneous Leishmaniasis. J Immunol Res 2021; 2021:6624246. [PMID: 33928168 PMCID: PMC8053065 DOI: 10.1155/2021/6624246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 03/20/2021] [Accepted: 03/30/2021] [Indexed: 11/18/2022] Open
Abstract
Leishmaniasis is a disease caused by an intracellular protozoan parasite of the genus Leishmania. Current treatments for leishmaniasis are long, toxic, and expensive and are not available in some endemic regions. Attempts to develop an effective vaccine are feasible, but no vaccine is in active clinical use. In this study, the LmxMBA gene of Leishmania mexicana was selected as a possible vaccine candidate using the reverse vaccinology approach, and the prophylactic effect generated by DNA vaccination with this gene in a murine model of cutaneous leishmaniasis was evaluated. The results showed that prophylactic vaccination with pVAX1::LmxMBA significantly reduced the size of the lesion and the parasitic load on the footpad, compared to the control groups. At a histological level, a smaller number of parasites were evident in the dermis, as well as the absence of connective tissue damage. Mice immunized with plasmid pVAX1::LmxMBA induced immunity characterized by an increase in the IgG2a/IgG1 > 1 ratio and a higher rate of lymphocyte proliferation. In this study, immunization with the plasmid promoted an improvement in the macroscopic and microscopic clinical manifestations of the experimental infection by L. mexicana, with a T helper 1 response characterized by an IgG2a/IgG1 > 1 ratio and high lymphoproliferative response. These findings support immunization with the plasmid pVAX1::LmxMBA as a preventive strategy against cutaneous infection of L. mexicana.
Collapse
Affiliation(s)
- María Angélica Burgos-Reyes
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, Mexico
| | - Lidia Baylón-Pacheco
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, Mexico
| | - Patricia Espíritu-Gordillo
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, Mexico
| | - Silvia Galindo-Gómez
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, Mexico
| | - Víctor Tsutsumi
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, Mexico
| | - José Luis Rosales-Encina
- Departamento de Infectómica y Patogénesis Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México City, Mexico
| |
Collapse
|
9
|
Lu G, Shan S, Zainab B, Ayaz Z, He J, Xie Z, Rashid U, Zhang D, Mehmood Abbasi A. Novel vaccine design based on genomics data analysis: A review. Scand J Immunol 2021; 93:e12986. [PMID: 33043473 DOI: 10.1111/sji.12986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 10/04/2020] [Accepted: 10/05/2020] [Indexed: 12/28/2022]
Abstract
Modification of pathogenic strains with the passage of time is responsible for evolution in the timeline of vaccine development for last 30 years. Recent advancements in computational vaccinology on the one hand and genome sequencing approaches on the other have generated new hopes in vaccine development. The aim of this review was to discuss the evolution of vaccines, their characteristics and limitations. In this review, we highlighted the evolution of vaccines, from first generation to the current status, pointing out how different vaccines have emerged and different approaches that are being followed up in the development of more rational vaccines against a wide range of diseases. Data were collected using Google Scholar, Web of Science, Science Direct, Web of Knowledge, Scopus and Science Hub, whereas computational tools such as NCBI, GeneMANIA and STRING were used to analyse the pathways of vaccine action. Innovative tools, such as computational tools, recombinant technologies and intra-dermal devices, are currently being investigated in order to improve the immunological response. New technologies enlightened the interactions of host proteins with pathogenic proteins for vaccine candidate development, but still there is a need of integrating transcriptomic and proteomic approaches. Although immunization with genomics data is a successful approach, its advantages must be assessed case by case and its applicability depends on the nature of the agent to be immunized, the nature of the antigen and the type of immune response required to achieve effective protection.
Collapse
Affiliation(s)
- Guangli Lu
- Institute of Business, School of Business, Henan University, Henan, China
| | - Sharui Shan
- The First Affiliated Hospital of Jinan University (Guangzhou Overseas Chinese Hospital), Guangzhou, China
- Department of Rehabilitation, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Bibi Zainab
- Department of Environmental Sciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Zainab Ayaz
- Department of Environmental Sciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Jialiang He
- School of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Zhenxing Xie
- Basic School of Medicine, Henan University, Kaifeng, China
| | - Umer Rashid
- Department of Chemistry, COMSATS University Islamabad, Islamabad, Pakistan
| | - Dalin Zhang
- Department of Biochemistry and Molecular Medicine, UC Davis Comprehensive Cancer Center, University of California Davis, Sacramento, CA, USA
| | - Arshad Mehmood Abbasi
- Department of Environmental Sciences, COMSATS University Islamabad, Islamabad, Pakistan
| |
Collapse
|
10
|
Abstract
The first proof-of-concept studies about the feasibility of genetic vaccines were published over three decades ago, opening the way for future development. The idea of nonviral antigen delivery had multiple advantages over the traditional live or inactivated pathogen-based vaccines, but a great deal of effort had to be invested to turn the idea of genetic vaccination into reality. Although early proof-of-concept studies were groundbreaking, they also showed that numerous aspects of genetic vaccines needed to be improved. Until the early 2000s, the vast majority of effort was invested into the development of DNA vaccines due to the potential issues of instability and low in vivo translatability of messenger RNA (mRNA). In recent years, numerous studies have demonstrated the outstanding abilities of mRNA to elicit potent immune responses against infectious pathogens and different types of cancer, making it a viable platform for vaccine development. Multiple mRNA vaccine platforms have been developed and evaluated in small and large animals and humans and the results seem to be promising. RNA-based vaccines have important advantages over other vaccine approaches including outstanding efficacy, safety, and the potential for rapid, inexpensive, and scalable production. There is a substantial investment by new mRNA companies into the development of mRNA therapeutics, particularly vaccines, increasing the number of basic and translational research publications and human clinical trials underway. This review gives a broad overview about genetic vaccines and mainly focuses on the past and present of mRNA vaccines along with the future directions to bring this potent vaccine platform closer to therapeutic use.
Collapse
|
11
|
Chung YH, Beiss V, Fiering SN, Steinmetz NF. COVID-19 Vaccine Frontrunners and Their Nanotechnology Design. ACS NANO 2020; 14:12522-12537. [PMID: 33034449 PMCID: PMC7553041 DOI: 10.1021/acsnano.0c07197] [Citation(s) in RCA: 227] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 10/05/2020] [Indexed: 05/18/2023]
Abstract
Humanity is experiencing a catastrophic pandemic. SARS-CoV-2 has spread globally to cause significant morbidity and mortality, and there still remain unknowns about the biology and pathology of the virus. Even with testing, tracing, and social distancing, many countries are struggling to contain SARS-CoV-2. COVID-19 will only be suppressible when herd immunity develops, either because of an effective vaccine or if the population has been infected and is resistant to reinfection. There is virtually no chance of a return to pre-COVID-19 societal behavior until there is an effective vaccine. Concerted efforts by physicians, academic laboratories, and companies around the world have improved detection and treatment and made promising early steps, developing many vaccine candidates at a pace that has been unmatched for prior diseases. As of August 11, 2020, 28 of these companies have advanced into clinical trials with Moderna, CanSino, the University of Oxford, BioNTech, Sinovac, Sinopharm, Anhui Zhifei Longcom, Inovio, Novavax, Vaxine, Zydus Cadila, Institute of Medical Biology, and the Gamaleya Research Institute having moved beyond their initial safety and immunogenicity studies. This review analyzes these frontrunners in the vaccine development space and delves into their posted results while highlighting the role of the nanotechnologies applied by all the vaccine developers.
Collapse
Affiliation(s)
- Young Hun Chung
- Department of Bioengineering, University
of California San Diego, La Jolla, California 92093, United
States
| | - Veronique Beiss
- Department of NanoEngineering, University
of California San Diego, La Jolla, California 92093, United
States
| | - Steven N. Fiering
- Geisel School of Medicine, Dartmouth
College, Hanover, New Hampshire 03755, United
States
- Norris Cotton Cancer Center,
Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire 03766,
United States
| | - Nicole F. Steinmetz
- Department of Bioengineering, University
of California San Diego, La Jolla, California 92093, United
States
- Department of NanoEngineering, University
of California San Diego, La Jolla, California 92093, United
States
- Department of Radiology, University of
California San Diego, La Jolla, California 92093, United
States
- Moores Cancer Center, University of California
San Diego, La Jolla, California 92093, United
States
- Center for Nano-ImmunoEngineering,
University of California San Diego, La Jolla, California
92093, United States
| |
Collapse
|
12
|
Singh AP, Prabhu SN, Nagaleekar VK, Dangi SK, Prakash C, Singh VP. Immunogenicity assessment of Clostridium perfringens type D epsilon toxin epitope-based chimeric construct in mice and rabbit. 3 Biotech 2020; 10:406. [PMID: 32864287 PMCID: PMC7447850 DOI: 10.1007/s13205-020-02400-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 08/17/2020] [Indexed: 12/05/2022] Open
Abstract
Epsilon toxin (Etx) belongs to family of pore-forming toxin and is produced by Clostridium perfringens type D. The Etx toxin is responsible for the pathogenesis of enterotoxaemia in sheep and goats, and occasionally in other livestock animals. The present study aimed to develop a Clostridium perfringens epsilon toxin-based chimeric epitope construct having immunodominant B-cell epitope and universal T-cell epitope and its immunogenicity was evaluated in mice and rabbit. An artificial chimeric epitope construct (CEC) was prepared by joining tandem repeats of a peptide containing amino acids (aa) 134–145 of epsilon toxin B-cell epitope and universal T-cell epitopes. The CEC was expressed in the Escherichia coli following codon optimization for efficient translational efficiency and purified by affinity chromatography. The antigenic reactivity of r-CEC proteins was confirmed by western blot with rabbit anti-r-Etox hyperimmune sera. The immunogenicity of the recombinant single CEC was examined in mice and rabbit by indirect ELISA. It was found that r-CEC yielded high titers of neutralizing antibodies (≥ 1.035 IU/ml) in immunized mice and rabbit. The potency of chimeric protein immunized serum was observed to be higher than the recommended level (0.1–0.3 IU/ml) for protection in sheep and goats. This indicated the potential ability of the chimeric protein as a vaccine candidate. This further requires studying the immune response in targeted host species (sheep and goat).
Collapse
Affiliation(s)
- Ajay Pratap Singh
- Department of Veterinary Microbiology, College of Veterinary and Animal Science, COVSc.&AH, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura, UP 281001 India
| | - Shyama N. Prabhu
- Department of Veterinary Pathology, COVSc.&AH, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya Evam Go Anusandhan Sansthan (DUVASU), Mathura, UP 281001 India
| | - Viswas K. Nagaleekar
- Division Bacteriology and Mycology, Indian Veterinary Reaserch Institute (IVRI), Izatnagar, Bareilly, Uttar Pradesh 243 122 India
| | - Saroj K. Dangi
- Division Bacteriology and Mycology, Indian Veterinary Reaserch Institute (IVRI), Izatnagar, Bareilly, Uttar Pradesh 243 122 India
| | - Chandan Prakash
- Centre for Advance Animal Research and Diagnosis, Indian Veterinary Reaserch Institute (IVRI), Izatnagar, Bareilly, Uttar Pradesh 243 122 India
| | - Vijendra Pal Singh
- National Institute of High Security Animal Disease (NISHAD), Bhopal, Madhya Pradesh 462021 India
| |
Collapse
|
13
|
Serwer P. Optimizing Anti-Viral Vaccine Responses: Input from a Non-Specialist. Antibiotics (Basel) 2020; 9:antibiotics9050255. [PMID: 32429032 PMCID: PMC7277631 DOI: 10.3390/antibiotics9050255] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/11/2022] Open
Abstract
Recently, the research community has had a real-world look at reasons for improving vaccine responses to emerging RNA viruses. Here, a vaccine non-specialist suggests how this might be done. I propose two alternative options and compare the primary alternative option with current practice. The basis of comparison is feasibility in achieving what we need: a safe, mass-produced, emerging virus-targeted vaccine on 2–4 week notice. The primary option is the following. (1) Start with a platform based on live viruses that infect bacteria, but not humans (bacteriophages, or phages). (2) Isolate phages (to be called pathogen homologs) that resemble and provide antigenic context for membrane-covered, pathogenic RNA viruses; coronavirus-phage homologs will probably be found if the search is correctly done. (3) Upon isolating a viral pathogen, evolve its phage homolog to bind antibodies neutralizing for the viral pathogen. Vaccinate with the evolved phage homolog by generating a local, non-hazardous infection with the phage host and then curing the infection by propagating the phage in the artificially infecting bacterial host. I discuss how this alternative option has the potential to provide what is needed after appropriate platforms are built.
Collapse
Affiliation(s)
- Philip Serwer
- Department of Biochemistry and Structural Biology, The University of Texas Health Center, San Antonio, TX 78229-3900, USA
| |
Collapse
|
14
|
Bai X, Song JH, Dai F, Lee JY, Hong SJ. Clonorchis sinensis secretory protein CsAg17 vaccine induces immune protection. Parasit Vectors 2020; 13:215. [PMID: 32334611 PMCID: PMC7183723 DOI: 10.1186/s13071-020-04083-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 04/13/2020] [Indexed: 01/25/2023] Open
Abstract
Background Clonorchiasis is endemic in East and Southeast Asian countries. For a preventive strategy against infectious diseases, vaccination is the most effective. Here, we evaluated the molecular characteristics and immune responses of CsAg17 protein from Clonorchis sinensis, and investigated its protective effects against C. sinensis challenge. Methods A cDNA clone encoding CsAg17 protein and containing a secretory signal peptide at the N-terminus was retrieved from the C. sinensis transcriptome bank. Recombinant CsAg17 B-cell epitope protein and cDNA vaccines were produced and their immune responses were evaluated in FVB mice. The proportional changes of CD3+/CD4+ and CD3+/CD8+ T cells were detected by flow cytometry, and immune effectors were measured by ELISA. Results The CsAg17 mRNA was transcribed at a higher level in C. sinensis adults than in metacercariae. The CsAg17 protein was distributed in the sperms, oral and ventral suckers, and mesenchymal tissues of C. sinensis adults. In mice challenged with C. sinensis metacercariae, vaccination with CsAg17 protein and cDNA resulted in a reduction to 64% and 69% in worm burden, respectively. Both CsAg17 protein and cDNA vaccines increased the proportion of CD3+/CD4+ and CD3+/CD8+ T cells and stimulated the production of Th1 type cytokines such as interleukin (IL)-2, IL-12, and interferon-γ, while maintaining minimum levels of Th2 cytokines. The levels of IgG specific to CsAg17 protein steeply increased in the two vaccinated groups from 2 weeks after immunization. The liver tissue retained good morphology in the mice vaccinated with CsAg17 protein or cDNA, whereas severe inflammation and large serous cysts were observed in the liver of the unvaccinated mice. Conclusions Vaccination with CsAg17 protein and cDNA reduced the pathological changes in the bile duct and liver, and ameliorated the worm burden via cellular and humoral immune responses. Thus, they may serve as good vaccine candidates against C. sinensis infections.![]()
Collapse
Affiliation(s)
- Xuelian Bai
- Department of Medical Environmental Biology, Chung-Ang University College of Medicine, Seoul, Republic of Korea. .,Clinical Medicine Laboratory, Binzhou Medical University Hospital, Binzhou, Shandong, People's Republic of China.
| | - Jin-Ho Song
- Department of Pharmacology, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Fuhong Dai
- Department of Medical Environmental Biology, Chung-Ang University College of Medicine, Seoul, Republic of Korea.,Department of Parasitology, School of Biology and Basic Medical Sciences, Medical College, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Ji-Yun Lee
- Department of Medical Environmental Biology, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Sung-Jong Hong
- Department of Medical Environmental Biology, Chung-Ang University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Soler Besumbes E, Fornaguera C, Monge M, García-Celma MJ, Carrión J, Solans C, Dols-Perez A. PLGA cationic nanoparticles, obtained from nano-emulsion templating, as potential DNA vaccines. Eur Polym J 2019. [DOI: 10.1016/j.eurpolymj.2019.109229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
16
|
Loh FK, Nathan S, Chow SC, Fang CM. Vaccination challenges and strategies against long-lived Toxoplasma gondii. Vaccine 2019; 37:3989-4000. [PMID: 31186188 DOI: 10.1016/j.vaccine.2019.05.083] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 04/05/2019] [Accepted: 05/21/2019] [Indexed: 01/03/2023]
Abstract
Since the discovery of Toxoplasma gondii in 1908, it is estimated that one-third of the global population has been exposed to this ubiquitous intracellular protozoan. The complex life cycle of T. gondii has enabled itself to overcome stress and transmit easily within a broad host range thus achieving a high seroprevalence worldwide. To date, toxoplasmosis remains one of the most prevalent HIV-associated opportunistic central nervous system infections. This review presents a comprehensive overview of different vaccination approaches ranging from traditional inactivated whole-T. gondii vaccines to the popular DNA vaccines. Extensive discussions are made to highlight the challenges in constructing these vaccines, selecting adjuvants as well as delivery methods, immunisation approaches and developing study models. Herein we also deliberate over the latest and promising enhancement strategies that can address the limitations in developing an effective T. gondii prophylactic vaccine.
Collapse
Affiliation(s)
- Fei-Kean Loh
- Division of Biomedical Sciences, School of Pharmacy, The University of Nottingham Malaysia Campus, 43500 Semenyih, Selangor, Malaysia
| | - Sheila Nathan
- School of Biosciences and Biotechnology, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor, Malaysia
| | - Sek-Chuen Chow
- School of Science, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor, Malaysia
| | - Chee-Mun Fang
- Division of Biomedical Sciences, School of Pharmacy, The University of Nottingham Malaysia Campus, 43500 Semenyih, Selangor, Malaysia.
| |
Collapse
|
17
|
Ding D, Zhu Q. Recent advances of PLGA micro/nanoparticles for the delivery of biomacromolecular therapeutics. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:1041-1060. [DOI: 10.1016/j.msec.2017.12.036] [Citation(s) in RCA: 162] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/18/2017] [Accepted: 12/30/2017] [Indexed: 01/06/2023]
|
18
|
Wang Y, Zhang B, Li J, Aipire A, Li Y, Zhang F. Enhanced contraception of canine zona pellucida 3 DNA vaccine via targeting DEC-205 in mice. Theriogenology 2018; 113:56-62. [PMID: 29455062 DOI: 10.1016/j.theriogenology.2018.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 01/24/2018] [Accepted: 02/04/2018] [Indexed: 12/25/2022]
Abstract
Zona pellucida 3 (ZP3) is a potential antigen for the development of contraceptive vaccines to control animal population. In this study, we designed a canine ZP3 (CZP3) DNA vaccine through targeting DEC-205 (named as pcD-scFv-CZP3c) and investigated its contraceptive effect in mice. Female BALB/c mice were intramuscularly immunized 3 times at 2 weeks intervals. After immunization, humoral and cellular immune responses were detected by ELISA and flow cytometry. The results showed that pcD-CZP3 and pcD-scFv-CZP3c induced CZP3-specific antibody (Ab) responses both in serum and vaginal secretions compared to pcDNA3.1. Additionally, compared to pcD-CZP3, pcD-scFv-CZP3c increased the levels of CZP3-specific Abs after a third immunization. Abs induced by these two DNA vaccines could bind with mice and dogs oocytes. Moreover, pcD-scFv-CZP3c enhanced the activation of CD4+ T cells characterized by the increased frequencies of CD4+CD44+ T cells. Finally, the contraceptive effect was evaluated in the immunized mice. These two DNA vaccines significantly decreased a mean litter size of mice compared to pcDNA3.1, but pcD-scFv-CZP3c group showed the smallest mean litter size. The mean litter size of pcD-scFv-CZP3 were 3.2 ± 0.742 and 4.6 ± 1.118 in two mating tests, which were significantly lower than pcDNA3.1(P < 0.001 and P < 0.05). Our results suggest that the CZP3 DNA vaccine targeted with DEC-205 may be a potential strategy for developing a contraceptive DNA vaccine.
Collapse
Affiliation(s)
- Ying Wang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Beibei Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Adila Aipire
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Yijie Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Fuchun Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China.
| |
Collapse
|
19
|
Arciola CR, Speziale P, Montanaro L. Perspectives on DNA Vaccines. Targeting Staphylococcal Adhesins to Prevent Implant Infections. Int J Artif Organs 2018; 32:635-41. [DOI: 10.1177/039139880903200913] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
DNA vaccines consist of a plasmid DNA genetically engineered to produce one or more proteins able to elicit protective immune responses against virulence factors of infectious pathogens. Once introduced into the cells of the host, a DNA vaccine induces a high production of antigens by the endogenous presence of the peptide codifying gene; improves antigen processing and presentation; may be able to simultaneously co-express multiple antigenic molecules; and, lastly, switches on both humoral and cellular immune responses. In this mini-review, we underscore the advantageous characteristics of DNA vaccines compared with traditional ones and provide summaries of some of the more recent studies on them, mainly focusing the possibility of their use in targeting the staphylococcal adhesins that play a key role in the first adhesive phase of implant infections.
Collapse
Affiliation(s)
- Carla Renata Arciola
- Research Unit on Implant Infections, Rizzoli Orthopedic Institute, Bologna - Italy
- Experimental Pathology Department, University of Bologna, Bologna - Italy
| | - Pietro Speziale
- Department of Biochemistry, University of Pavia, Pavia - Italy
| | - Lucio Montanaro
- Research Unit on Implant Infections, Rizzoli Orthopedic Institute, Bologna - Italy
- Experimental Pathology Department, University of Bologna, Bologna - Italy
| |
Collapse
|
20
|
Alam A, Imam N, farooqui A, Ali S, Malik MZ, Ishrat R. Recent trends in ZikV research: A step away from cure. Biomed Pharmacother 2017; 91:1152-1159. [DOI: 10.1016/j.biopha.2017.05.045] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/02/2017] [Accepted: 05/09/2017] [Indexed: 12/23/2022] Open
|
21
|
El-Mogy MA, Abdalla MA, Misic V, Haj-Ahmad Y. Effect of adenovirus infection on transgene expression under the adenoviral MLP/TPL and the CMVie promoter/enhancer in CHO cells. J Genet Eng Biotechnol 2017; 15:211-217. [PMID: 30647657 PMCID: PMC6296590 DOI: 10.1016/j.jgeb.2017.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 02/27/2017] [Accepted: 04/05/2017] [Indexed: 11/19/2022]
Abstract
The adenovirus major late promoter (MLP) and its translational regulator - the tripartite leader (TPL) sequence - can actively drive efficient gene expression during adenoviral infection. However, both elements have not been widely tested in transgene expression outside of the adenovirus genome context. In this study, we tested whether the combination of MLP and TPL would enhance transgene expression beyond that of the most widely used promoter in transgene expression in mammalian cells, the cytomegalovirus immediate early (CMVie) promoter/enhancer. The activity of these two regulatory elements was compared in Chinese hamster ovary (CHO) cells. Although transient expression was significantly higher under the control of the CMVie promoter/enhance compared to the MLP/TPL, this difference was greater at the level of transcription (30 folds) than translation (11 folds). Even with adenovirus infection to provide additional elements (in trans), CMVie promoter/enhancer exhibited significantly higher activity relative to MLP/TPL. Interestingly, the CMVie promoter/enhancer was 1.9 folds more active in adenovirus-infected cells than in non-infected cells. Our study shows that the MLP-TPL drives lower transgene expression than the CMVie promoter/enhancer particularly at the transcription level. The data also highlight the utility of the TPL sequence at the translation level and/or possible overwhelming of the cellular translational machinery by the high transcription activity of the CMVie promoter/enhancer. In addition, here we present data that show stimulation of the CMVie promoter/enhancer by adenovirus infection, which may prove interesting in future work to test the combination of CMVie/TPL sequence, and additional adenovirus elements, for transgene expression.
Collapse
Key Words
- Adenovirus
- CHO, Chinese hamster ovary
- CMVie promoter/enhancer
- CMVie, cytomegalovirus immediate early
- GFP, green fluorescence proteins
- IRES, internal ribosome entry site
- MLP
- MLP, major late promoter
- MOI, multiplicity of infection
- PFU, plaque forming unit
- RFU, relative fluorescence units
- TPL
- TPL, tripartite leader
- Transgene expression
- qPCR, Quantitative PCR
Collapse
Affiliation(s)
- Mohamed A. El-Mogy
- Molecular Biology Department, National Research Centre, Dokki, Giza, Egypt
- Department of Biological Sciences, Brock University, St. Catharines, ON, Canada
- Corresponding author at: Molecular Biology Department, National Research Centre, Dokki, Giza, Egypt.
| | - Moemen A.K. Abdalla
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Vanja Misic
- Department of Biological Sciences, Brock University, St. Catharines, ON, Canada
| | - Yousef Haj-Ahmad
- Department of Biological Sciences, Brock University, St. Catharines, ON, Canada
| |
Collapse
|
22
|
Zhou J, Lu G, He S. Analysis of structures and epitopes of a novel secreted protein MYR1 in Toxoplasma gondii. Folia Parasitol (Praha) 2016; 63. [DOI: 10.14411/fp.2016.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 07/05/2016] [Indexed: 11/19/2022]
|
23
|
Zhou J, Wang L, Lu G, Zhou A, Zhu M, Li Q, Wang Z, Arken M, Wang A, He S. Epitope analysis and protection by a ROP19 DNA vaccine against Toxoplasma gondii. ACTA ACUST UNITED AC 2016; 23:17. [PMID: 27055564 PMCID: PMC4824872 DOI: 10.1051/parasite/2016017] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 03/16/2016] [Indexed: 11/14/2022]
Abstract
We used bioinformatics approaches to identify B-cell and T-cell epitopes on the ROP19 protein of Toxoplasma gondii. Then, we constructed plasmids with ROP19 (pEGFP-C1-ROP19) and injected them into BALB/c mice to test the immunoprotection induced by this vaccine candidate. The results showed that immunization with pEGFP-C1-ROP19 induced effective cellular and humoral immune responses in mice; specifically, high serum levels of T. gondii-specific IgG and increased interferon-gamma production by splenocytes. Furthermore, the mice vaccinated with pROP19 had significantly fewer brain cysts (583 ± 160) than the mice injected with phosphate-buffered saline (1350 ± 243) or with the control plasmid, pEGFP-C1 (1300 ± 167). Compared with PBS-treated mice, those immunized with pROP19 had only 43% of the number of brain cysts. These results suggest that the DNA vaccine encoding ROP19 induced a significant immune response and provided protection against a challenge with T. gondii strain PRU cysts.
Collapse
Affiliation(s)
- Jian Zhou
- Department of Parasitology, Shandong University School of Medicine,250012 Jinan, Shandong Province, PR China
| | - Lin Wang
- Department of Ji Nan Children's Hospital, 250022 Jinan, Shandong Province, PR China
| | - Gang Lu
- Department of Parasitology, Shandong University School of Medicine,250012 Jinan, Shandong Province, PR China
| | - Aihua Zhou
- Department of Pediatrics, Provincial Hospital Affiliated to Shandong University, Shandong University School of Medicine, 250021 Jinan, Shandong Province, PR China
| | - Meiyan Zhu
- Department of Parasitology, Shandong University School of Medicine,250012 Jinan, Shandong Province, PR China
| | - Qihang Li
- Department of Parasitology, Shandong University School of Medicine,250012 Jinan, Shandong Province, PR China
| | - Zhilin Wang
- Department of Parasitology, Shandong University School of Medicine,250012 Jinan, Shandong Province, PR China
| | - Miradel Arken
- Department of Parasitology, Shandong University School of Medicine,250012 Jinan, Shandong Province, PR China
| | - Ao Wang
- Department of Parasitology, Shandong University School of Medicine,250012 Jinan, Shandong Province, PR China
| | - Shenyi He
- Department of Parasitology, Shandong University School of Medicine,250012 Jinan, Shandong Province, PR China
| |
Collapse
|
24
|
Zhou J, Wang L, Zhou A, Lu G, Li Q, Wang Z, Zhu M, Zhou H, Cong H, He S. Bioinformatics analysis and expression of a novel protein ROP48 in Toxoplasma gondii. Acta Parasitol 2016; 61:319-28. [PMID: 27078655 DOI: 10.1515/ap-2016-0042] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 12/10/2015] [Indexed: 01/30/2023]
Abstract
Toxoplasma gondii is an obligate intracellular apicomplexan parasite, and can infect warmblooded animals and humans all over the world. In the past years, ROP family genes encoding particular proteins of T. gondii had made a great contribution to toxoplasmosis. In this study, we used multiple bioinformatics approaches to predict the physical and chemical characteristics, transmembrane domain, epitope, and topological structure of the rhoptry protein 48 (ROP48). The results indicated that ROP48 protein was mainly located in the membrane and had several positive linear-B cell epitopes and Th-cell epitopes, which suggested that ROP48 is a potential DNA vaccine candidate against toxoplasmosis. Then the PCR product amplified from the ROP48 cDNA was inserted into a pEASY-T1 vector to build a recombinant cloning plasmid. After sequencing, ROP48 was subcloned into a eukaryotic expression plasmid pEGFP-C1 to obtain pEGFP-C1-ROP48 (pROP48). After identification by PCR and restriction enzyme digestion, the recombinant plasmid pROP48 was transfected into HEK 293-T cell and identified by RT-PCR. The results showed that the eukaryotic expression plasmid pROP48 was constructed and transfected to the cells of HEK 293-T successfully. Western blotting showed that the expressed proteins can be recognized by anti-STAg mouse sera.
Collapse
|
25
|
Chaudhari A, Pathakota GB, Annam PK. Design and Construction of Shrimp Antiviral DNA Vaccines Expressing Long and Short Hairpins for Protection by RNA Interference. Methods Mol Biol 2016; 1404:225-240. [PMID: 27076302 DOI: 10.1007/978-1-4939-3389-1_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
DNA vaccines present the aquaculture industry with an effective and economically viable method of controlling viral pathogens that drastically affect productivity. Since specific immune response is rudimentary in invertebrates, the presence of RNA interference (RNAi) pathway in shrimps provides a promising new approach to vaccination. Plasmid DNA vaccines that express short or long double stranded RNA in vivo have shown protection against viral diseases. The design, construction and considerations for preparing such vaccines are discussed.
Collapse
Affiliation(s)
- Aparna Chaudhari
- ICAR-Central Institute of Fisheries Education, Versova, Andheri West, Mumbai, 400061, India.
| | - Gireesh-Babu Pathakota
- ICAR-Central Institute of Fisheries Education, Versova, Andheri West, Mumbai, 400061, India
| | - Pavan-Kumar Annam
- ICAR-Central Institute of Fisheries Education, Versova, Andheri West, Mumbai, 400061, India
| |
Collapse
|
26
|
Lu G, Wang L, Zhou A, Han Y, Guo J, Song P, Zhou H, Cong H, Zhao Q, He S. Epitope analysis, expression and protection of SAG5A vaccine against Toxoplasma gondii. Acta Trop 2015; 146:66-72. [PMID: 25792417 DOI: 10.1016/j.actatropica.2015.03.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 03/06/2015] [Accepted: 03/09/2015] [Indexed: 01/02/2023]
Abstract
Bioinformatics approaches were used to identify B-cell epitopes and T-cell epitopes on SAG5A protein. Compared to SAG1, SAG5A with good B-cell epitopes and T-cell epitopes had a potentiality to become a more successful vaccine against Toxoplasma gondii. Thereafter, SAG5A DNA vaccine was constructed successfully and was injected into mice with peptide to evaluate the immunoprotection. Compared to the control groups, the vaccine (DNA/peptide) could induce more effective cellular and humoral immune responses in immunized mice. Furthermore, a significant reduction of brain cyst was detected in the mice vaccinated with peptide (732±160), pSAG5A (815±197), or pSAG5A/peptide (436±174) compared by the mice injected by PBS (1260±241) or pEGFP-C1 (1350±268). The number of cysts in brains was 35% reduced in the mice immunized with DNA/peptide than in the control mice treated by PBS. The results indicated that the DNA vaccine encoding SAG5A significantly induced immune responses and enhanced protection against cysts of PRU strain, especially with the help of peptide.
Collapse
|
27
|
Zhu Y, Li C, Bi Z, Chen Z, Meng C, Wang GJ, Ding C, Liu G. Protective immune responses in ducklings induced by a suicidal DNA vaccine of the sigma C gene of novel duck reovirus. Vet Immunol Immunopathol 2015; 165:88-92. [DOI: 10.1016/j.vetimm.2015.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 02/22/2015] [Accepted: 03/02/2015] [Indexed: 11/27/2022]
|
28
|
Daep CA, Muñoz-Jordán JL, Eugenin EA. Flaviviruses, an expanding threat in public health: focus on dengue, West Nile, and Japanese encephalitis virus. J Neurovirol 2014; 20:539-60. [PMID: 25287260 PMCID: PMC4331079 DOI: 10.1007/s13365-014-0285-z] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/01/2014] [Accepted: 08/26/2014] [Indexed: 10/24/2022]
Abstract
The flaviviruses dengue, West Nile, and Japanese encephalitis represent three major mosquito-borne viruses worldwide. These pathogens impact the lives of millions of individuals and potentially could affect non-endemic areas already colonized by mosquito vectors. Unintentional transport of infected vectors (Aedes and Culex spp.), traveling within endemic areas, rapid adaptation of the insects into new geographic locations, climate change, and lack of medical surveillance have greatly contributed to the increase in flaviviral infections worldwide. The mechanisms by which flaviviruses alter the immune and the central nervous system have only recently been examined despite the alarming number of infections, related deaths, and increasing global distribution. In this review, we will discuss the expansion of the geographic areas affected by flaviviruses, the potential threats to previously unaffected countries, the mechanisms of pathogenesis, and the potential therapeutic interventions to limit the devastating consequences of these viruses.
Collapse
Affiliation(s)
- Carlo Amorin Daep
- Public Health Research Institute (PHRI), Rutgers New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, USA
- Department of Microbiology and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, USA
| | - Jorge L. Muñoz-Jordán
- Centers for Disease Control and Prevention Dengue Branch, 1324 Cañada Street, San Juan, PR 00971
| | - Eliseo Alberto Eugenin
- Public Health Research Institute (PHRI), Rutgers New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, USA
- Department of Microbiology and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers the State University of New Jersey, Newark, NJ, USA
| |
Collapse
|
29
|
Zhang Y, Su WJ, Wang J, Bai XF, Huang CX, Lian JQ. A fusion DNA vaccine encoding middle version of HBV envelope protein fused to interleukin-21 did not enhance HBV-specific immune response in mice. Viral Immunol 2014; 27:430-7. [PMID: 25211639 DOI: 10.1089/vim.2014.0051] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
DNA vaccination can generate both humoral and cellular immunity, resulting in potential prophylactic and therapeutic vaccines in variety of conditions, including hepatitis B virus (HBV) infection. Fusion of cytokine gene is one of the ways to increase the immunogenicity of DNA vaccine. Interleukin (IL)-21 has been demonstrated to play an immunomodulatory role in HBV infection. Thus, we aimed to investigate the ability of IL-21 in the regulation of middle version of HBV envelop protein (MS) DNA vaccine. Fusion plasmid encoding IL-21 linked with MS was constructed. Normal and HBV transgenic mice were immunized by plasmid. pcDNA-IL-21/S2S induced a comparable level of anti-HBs antibody and HBsAg-specific CD8+ T-cell response with pcDNA-S2S. Furthermore, the level of circulating HBsAg was decreased by induction of anti-HBs antibody and HBsAg-specific CD8+ T-cell response to both pcDNA-IL-21/S2S and pcDNA-S2S vaccination in HBV transgenic mice. Thus, immunization with DNA vaccine encoding HBV MS protein induced both T- and B-cell response by targeting the specific antigen. Furthermore, it was also revealed that MS DNA vaccination could break immune tolerance in HBV transgenic mice. But IL-21 did not strengthen immune response induced by HBV DNA immunization. Our study suggested that MS-expressing plasmid may be useful for both preventive and therapeutic methods in HBV infection. However, IL-21 does not improve the immunogenicity and efficacy of MS DNA vaccination, and thus may not be used as a therapeutic marker for chronic hepatitis B.
Collapse
Affiliation(s)
- Ye Zhang
- 1 Center for Infectious Diseases, Tangdu Hospital, Fourth Military Medical University , Xi'an, China
| | | | | | | | | | | |
Collapse
|
30
|
Weng TY, Yen MC, Huang CT, Hung JJ, Chen YL, Chen WC, Wang CY, Chang JY, Lai MD. DNA vaccine elicits an efficient antitumor response by targeting the mutant Kras in a transgenic mouse lung cancer model. Gene Ther 2014; 21:888-96. [PMID: 25077772 DOI: 10.1038/gt.2014.67] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 04/09/2014] [Accepted: 06/17/2014] [Indexed: 12/22/2022]
Abstract
Mutant Kras (V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog) is observed in more than 20% of non-small-cell lung cancers; however, no effective Kras target therapy is available at present. The Kras DNA vaccine may represent as a novel immunotherapeutic agent in lung cancer. In this study, we investigated the antitumor efficacy of the Kras DNA vaccine in a genetically engineered inducible mouse lung tumor model driven by Kras(G12D). Lung tumors were induced by doxycycline, and the therapeutic effects of Kras DNA vaccine were evaluated with delivery of Kras(G12D) plasmids. Mutant Kras(G12D) DNA vaccine significantly decreased the tumor nodules. A dominant-negative mutant Kras(G12D)N17, devoid of oncogenic activity, achieved similar therapeutic effects. The T-helper 1 immune response was enhanced in mice treated with Kras DNA vaccine. Splenocytes from mice receiving Kras DNA vaccine presented an antigen-specific response by treatment with peptides of Kras but not Hras or OVA. The number of tumor-infiltrating CD8(+) T cells increased after Kras vaccination. In contrast, Kras DNA vaccine was not effective in the lung tumor in transgenic mice, which was induced by mutant L858R epidermal growth factor receptor. Overall, these results indicate that Kras DNA vaccine produces an effective antitumor response in transgenic mice, and may be useful in treating lung cancer-carrying Ras mutation.
Collapse
Affiliation(s)
- T-Y Weng
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - M-C Yen
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [3] Center for Infectious Diseases and Signal Research, National Cheng Kung University, Tainan, Taiwan, ROC
| | - C-T Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - J-J Hung
- Institute of Bioinformatics and Biosignal Transduction, College of Life Science, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Y-L Chen
- Department of Senior Citizen Services Management, Chia Nan University of Pharmacy and Science, Tainan, Taiwan, ROC
| | - W-C Chen
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - C-Y Wang
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - J-Y Chang
- National Institute of Cancer Research, National Health Research Institute, Tainan, Taiwan, ROC
| | - M-D Lai
- 1] Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [2] Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC [3] Center for Infectious Diseases and Signal Research, National Cheng Kung University, Tainan, Taiwan, ROC
| |
Collapse
|
31
|
Recent Developments in Preclinical DNA Vaccination. Vaccines (Basel) 2014; 2:89-106. [PMID: 26344468 PMCID: PMC4494203 DOI: 10.3390/vaccines2010089] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 11/22/2013] [Accepted: 11/26/2013] [Indexed: 12/21/2022] Open
Abstract
The advantages of genetic immunization of the new vaccine using plasmid DNAs are multifold. For example, it is easy to generate plasmid DNAs, increase their dose during the manufacturing process, and sterilize them. Furthermore, they can be stored for a long period of time upon stabilization, and their protein encoding sequences can be easily modified by employing various DNA-manipulation techniques. Although DNA vaccinations strongly increase Th1-mediated immune responses in animals, several problems persist. One is about their weak immunogenicity in humans. To overcome this problem, various genetic adjuvants, electroporation, and prime-boost methods have been developed preclinically, which are reviewed here.
Collapse
|
32
|
Lukashevich IS, Shirwan H. Adenovirus-Based Vectors for the Development of Prophylactic and Therapeutic Vaccines. NOVEL TECHNOLOGIES FOR VACCINE DEVELOPMENT 2014. [PMCID: PMC7121347 DOI: 10.1007/978-3-7091-1818-4_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Emerging and reemerging infectious diseases as well as cancer pose great global health impacts on the society. Vaccines have emerged as effective treatments to prevent or reduce the burdens of already developed diseases. This is achieved by means of activating various components of the immune system to generate systemic inflammatory reactions targeting infectious agents or diseased cells for control/elimination. DNA virus-based genetic vaccines gained significant attention in the past decades owing to the development of DNA manipulation technologies, which allowed engineering of recombinant viral vectors encoding sequences for foreign antigens or their immunogenic epitopes as well as various immunomodulatory molecules. Despite tremendous progress in the past 50 years, many hurdles still remain for achieving the full clinical potential of viral-vectored vaccines. This chapter will present the evolution of vaccines from “live” or “attenuated” first-generation agents to recombinant DNA and viral-vectored vaccines. Particular emphasis will be given to human adenovirus (Ad) for the development of prophylactic and therapeutic vaccines. Ad biological properties related to vaccine development will be highlighted along with their advantages and potential hurdles to be overcome. In particular, we will discuss (1) genetic modifications in the Ad capsid protein to reduce the intrinsic viral immunogenicity, (2) antigen capsid incorporation for effective presentation of foreign antigens to the immune system, (3) modification of the hexon and fiber capsid proteins for Ad liver de-targeting and selective retargeting to cancer cells, (4) Ad-based vaccines carrying “arming” transgenes with immunostimulatory functions as immune adjuvants, and (5) oncolytic Ad vectors as a new therapeutic approach against cancer. Finally, the combination of adenoviral vectors with other non-adenoviral vector systems, the prime/boost strategy of immunization, clinical trials involving Ad-based vaccines, and the perspectives for the field development will be discussed.
Collapse
Affiliation(s)
- Igor S Lukashevich
- Department of Pharmacology and Toxicolog Department of Microbiology and Immunolog, University of Louisville, Louisville, Kentucky USA
| | - Haval Shirwan
- Department of Microbiology and Immunolog, University of Louisville, Louisville, Kentucky USA
| |
Collapse
|
33
|
Induction of antigen-positive cell death by the expression of perforin, but not DTa, from a DNA vaccine enhances the immune response. Immunol Cell Biol 2013; 92:359-67. [PMID: 24323081 DOI: 10.1038/icb.2013.93] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/28/2013] [Accepted: 11/09/2013] [Indexed: 02/06/2023]
Abstract
The failure of traditional protein-based vaccines to prevent infection by viruses such as HIV or hepatitis C highlights the need for novel vaccine strategies. DNA vaccines have shown promise in small animal models, and are effective at generating anti-viral T cell-mediated immune responses; however, they have proved to be poorly immunogenic in clinical trials. We propose that the induction of necrosis will enhance the immune response to vaccine antigens encoded by DNA vaccines, as necrotic cells are known to release a range of intracellular factors that lead to dendritic cell (DC) activation and enhanced cross-presentation of antigen. Here we provide evidence that induction of cell death in DNA vaccine-targeted cells provides an adjuvant effect following intradermal vaccination of mice; however, this enhancement of the immune response is dependent on both the mechanism and timing of cell death after antigen expression. We report that a DNA vaccine encoding the cytolytic protein, perforin, resulted in DC activation, enhanced broad and multifunctional CD8 T-cell responses to the HIV-1 antigen GAG and reduced viral load following challenge with a chimeric virus, EcoHIV, compared with the canonical GAG DNA vaccine. This effect was not observed for a DNA vaccine encoding an apoptosis-inducing toxin, DTa, or when the level of perforin expression was increased to induce cell death sooner after vaccination. Thus, inducing lytic cell death following a threshold level of expression of a viral antigen can improve the immunogenicity of DNA vaccines, whereas apoptotic cell death has an inhibitory effect on the immune response.
Collapse
|
34
|
Nanotechnological Approaches for Genetic Immunization. DNA AND RNA NANOBIOTECHNOLOGIES IN MEDICINE: DIAGNOSIS AND TREATMENT OF DISEASES 2013. [PMCID: PMC7121080 DOI: 10.1007/978-3-642-36853-0_4] [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: 10/25/2022]
Abstract
Genetic immunization is one of the important findings that provide multifaceted immunological response against infectious diseases. With the advent of r-DNA technology, it is possible to construct vector with immunologically active genes against specific pathogens. Nevertheless, site-specific delivery of constructed genetic material is an important contributory factor for eliciting specific cellular and humoral immune response. Nanotechnology has demonstrated immense potential for the site-specific delivery of biomolecules. Several polymeric and lipidic nanocarriers have been utilized for the delivery of genetic materials. These systems seem to have better compatibility, low toxicity, economical and capable to delivering biomolecules to intracellular site for the better expression of desired antigens. Further, surface engineering of nanocarriers and targeting approaches have an ability to offer better presentation of antigenic material to immunological cells. This chapter gives an overview of existing and emerging nanotechnological approaches for the delivery of genetic materials.
Collapse
|
35
|
Liu L, Qiu C, Huang Y, Xu J, Shao Y. Potent T cell responses induced by single DNA vaccine boosted with recombinant vaccinia vaccine. Virol Sin 2013; 28:109-15. [PMID: 23575733 PMCID: PMC8208324 DOI: 10.1007/s12250-013-3303-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 03/06/2013] [Indexed: 01/20/2023] Open
Abstract
Plasmid DNA, an effective vaccine vector, can induce both cellular and humoral immune responses. However, plasmid DNA raises issues concerning potential genomic integration after injection. This issue should be considered in preclinical studies. Tiantan vaccinia virus (TV) has been most widely utilized in eradicating smallpox in China. This virus has also been considered as a successful vaccine vector against a few infectious diseases. Potent T cell responses through T-cell receptor (TCR) could be induced by three injections of the DNA prime vaccine followed by a single injection of recombinant vaccinia vaccine. To develop a safer immunization strategy, a single DNA prime followed by a single recombinant Tiantan vaccinia (rTV) AIDS vaccine was used to immunize mice. Our data demonstrated that one DNA prime/rTV boost regimen induced mature TCR activation with high functional avidity, preferential T cell Vβ receptor usage and high sensitivity to anti-CD3 antibody stimulation. No differences in T cell responses were observed among one, two or three DNA prime/rTV boost regimens. This study shows that one DNA prime/rTV boost regimen is sufficient to induce potent T cell responses against HIV.
Collapse
Affiliation(s)
- Lianxing Liu
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071 China
- State Key Laboratory for Infectious Diseases Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206 China
| | - Chao Qiu
- Science Research Department, Shanghai Public Health Clinical Center, Public Health Clinical Center affiliated to Fudan University, Shanghai, 201508 China
| | - Yang Huang
- Science Research Department, Shanghai Public Health Clinical Center, Public Health Clinical Center affiliated to Fudan University, Shanghai, 201508 China
| | - Jianqing Xu
- Science Research Department, Shanghai Public Health Clinical Center, Public Health Clinical Center affiliated to Fudan University, Shanghai, 201508 China
| | - Yiming Shao
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071 China
- State Key Laboratory for Infectious Diseases Prevention and Control, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206 China
| |
Collapse
|
36
|
Yan R, Sun W, Song X, Xu L, Li X. Vaccination of goats with DNA vaccine encoding Dim-1 induced partial protection against Haemonchus contortus: a preliminary experimental study. Res Vet Sci 2013; 95:189-99. [PMID: 23545480 DOI: 10.1016/j.rvsc.2013.02.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 01/05/2013] [Accepted: 02/27/2013] [Indexed: 10/27/2022]
Abstract
Disorganized muscle family member (Dim-1) belongs to immunoglobulin superfamily, and is a structural protein localized to the region of the muscle cell membrane around and between the dense bodies. Strong immunogenicity to host's immune system was induced by Dim-1 from Ascaris suum, which indicated that Dim-1 could be a potential candidate for vaccine. The homologues of Dim-1 were also detected in nematodes Brugia malayi and Trichostrongylus colubriformis. However, information on the complete coding sequence and protection potential of this molecule in Haemonchus contortus is lacking. In this study, full length of Dim-1 cDNA was cloned using a rapid amplification of cDNA ends (RACE) strategy and the DNA vaccine encoding Dim-1 open reading frame (ORF) was tested for protection against experimental H. contortus infections in goats. Fifteen goats were allocated into three trial groups. The animals of Dim-1 group were vaccinated with the DNA vaccine encoding Dim-1 on day 0 and 14, and challenged with 5000 infective H. contortus third larval stage (L3) on day 28. An unvaccinated positive control group was challenged with L3 at the same time. An unvaccinated negative control group was not challenged with L3. The results indicated Dim-1 DNA vaccines were transcribed at local injection sites and expressed in vivo post immunizations respectively. Following L3 challenge, the mean eggs per gram feces (EPG) and worm burdens of Dim-1 group were reduced by 45.7% and 51.1%, respectively. Significantly high levels of serum IgG, serum IgA, mucosal IgA, CD4(+) T lymphocytes and B lymphocytes of Dim-1 group were produced. While compared with the negative control group, increased numbers of blood eosinophils and lymphocytes and declined haemoglobin level were observed in the Dim-1 group after L3 challenge. The preliminary study suggest that recombinant H. contortus Dim-1 DNA vaccine induced partial immune response and has protective potential against goat haemonchosis.
Collapse
Affiliation(s)
- Ruofeng Yan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | | | | | | | | |
Collapse
|
37
|
Kling JC, Körner H. Different regulatory mechanisms in protozoan parasitic infections. Int J Parasitol 2013; 43:417-25. [PMID: 23470812 DOI: 10.1016/j.ijpara.2013.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 02/01/2013] [Accepted: 02/04/2013] [Indexed: 02/07/2023]
Abstract
The immune response to the protozoan pathogens, Leishmania spp., Trypanosoma spp. and Plasmodium spp., has been studied extensively with particular focus on regulation of the immune response by immunological mechanisms. More specifically, in diseases caused by parasites, immunosuppression frequently prevents immunopathology that can injure the host. However, this allows a small number of parasites to evade the immune response and remain in the host after a clinical cure. The consequences can be chronic infections, which establish a zoonotic or anthroponotic reservoir. This review will highlight some of the identified regulatory mechanisms of the immune system that govern immune responses to parasitic diseases, in particular leishmaniasis, trypanosomiasis and malaria, and discuss implications for the development of efficient vaccines against these diseases.
Collapse
Affiliation(s)
- Jessica C Kling
- Menzies Research Institute Tasmania, University of Tasmania, Hobart, Tasmania 7000, Australia
| | | |
Collapse
|
38
|
Mutiso JM, Macharia JC, Kiio MN, Ichagichu JM, Rikoi H, Gicheru MM. Development of Leishmania vaccines: predicting the future from past and present experience. J Biomed Res 2013; 27:85-102. [PMID: 23554800 PMCID: PMC3602867 DOI: 10.7555/jbr.27.20120064] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Revised: 07/14/2012] [Accepted: 08/12/2012] [Indexed: 01/13/2023] Open
Abstract
Leishmaniasis is a disease that ranges in severity from skin lesions to serious disfigurement and fatal systemic infection. Resistance to infection is associated with a T-helper-1 immune response that activates macrophages to kill the intracellular parasite in a nitric oxide-dependent manner. Conversely, disease progression is generally associated with a T-helper-2 response that activates humoral immunity. Current control is based on chemotherapeutic treatments which are expensive, toxic and associated with high relapse and resistance rates. Vaccination remains the best hope for control of all forms of the disease, and the development of a safe, effective and affordable antileishmanial vaccine is a critical global public-health priority. Extensive evidence from studies in animal models indicates that solid protection can be achieved by immunization with defined subunit vaccines or live-attenuated strains of Leishmania. However, to date, no vaccine is available despite substantial efforts by many laboratories. Major impediments in Leishmania vaccine development include: lack of adequate funding from national and international agencies, problems related to the translation of data from animal models to human disease, and the transition from the laboratory to the field. Furthermore, a thorough understanding of protective immune responses and generation and maintenance of the immunological memory, an important but least-studied aspect of antiparasitic vaccine development, during Leishmania infection is needed. This review focuses on the progress of the search for an effective vaccine against human and canine leishmaniasis.
Collapse
Affiliation(s)
- Joshua Muli Mutiso
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Karen, Nairobi 24481-00502, Kenya;
- Department of Zoological Sciences, Kenyatta University, Nairobi 43844-00100, Kenya.
| | - John Chege Macharia
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Karen, Nairobi 24481-00502, Kenya;
| | - Maria Ndunge Kiio
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Karen, Nairobi 24481-00502, Kenya;
| | - James Maina Ichagichu
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Karen, Nairobi 24481-00502, Kenya;
| | - Hitler Rikoi
- Department of Tropical and Infectious Diseases, Institute of Primate Research, Karen, Nairobi 24481-00502, Kenya;
| | | |
Collapse
|
39
|
Abstract
Vaccination is the most successful application of immunological principles to human health. Vaccine efficacy needs to be reviewed from time to time and its safety is an overriding consideration. DNA vaccines offer simple yet effective means of inducing broad-based immunity. These vaccines work by allowing the expression of the microbial antigen inside host cells that take up the plasmid. These vaccines function by generating the desired antigen inside the cells, with the advantage that this may facilitate presentation through the major histocompatibility complex. This review article is based on a literature survey and it describes the working and designing strategies of DNA vaccines. Advantages and disadvantages for this type of vaccines have also been explained, together with applications of DNA vaccines. DNA vaccines against cancer, tuberculosis, Edwardsiella tarda, HIV, anthrax, influenza, malaria, dengue, typhoid and other diseases were explored.
Collapse
|
40
|
Cheng Y, Chen Z, Li C, Meng C, Wu R, Liu G. Protective immune responses in rabbits induced by a suicidal DNA vaccine of the VP60 gene of rabbit hemorrhagic disease virus. Antiviral Res 2013; 97:227-31. [DOI: 10.1016/j.antiviral.2012.12.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 12/14/2012] [Accepted: 12/15/2012] [Indexed: 11/29/2022]
|
41
|
Toxoplasma gondii: bioinformatics analysis, cloning and expression of a novel protein TgIMP1. Exp Parasitol 2012; 132:458-64. [PMID: 23026454 DOI: 10.1016/j.exppara.2012.09.015] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 06/08/2012] [Accepted: 09/20/2012] [Indexed: 11/20/2022]
Abstract
Toxoplasma gondii is an obligate intracellular protozoan parasite, infecting a large variety of animals and human beings. In recent years, the study of DNA vaccine against T. gondii has made a great progress; however, few vaccines have completely controlled toxoplasmosis. Thus people started to look for more effective antigenic proteins. Here we report a novel T. gondii protein termed immune mapped protein 1 (TgIMP1). We used multiple bioinformatics approaches to predict the physical and chemical characters, signal peptide, transmembrane domain, epitope, topological structure and function of the protein, and we theoretically determined that the TgIMP1 has multiple epitopes, and with immunogenicity, suggesting that the TgIMP1 may be a vaccine candidate against toxoplasmosis. Then the gene coding TgIMP1 was obtained by PCR and connected with cloning vector. Recombinant plasmid was identified by PCR, double digestion and sequencing analysis. Then the TgIMP1 gene was directly inserted into the eukaryotic expression vector pBudCE4.1, so that the recombinant eukaryotic expression plasmid pBudCE4.1-TgIMP1 was constructed. After identification by PCR and restriction enzyme digestion, the recombinant plasmid pBudCE4.1-TgIMP1 was transfected into cells of HFF, and then identified by RT-PCR. The results showed that the eukaryotic expression plasmid pBudCE4.1-TgIMP1 was constructed and was transfected to the HFF cells successfully.
Collapse
|
42
|
Multicomponent DNA vaccine-encoding Toxoplasma gondii GRA1 and SAG1 primes: anti-Toxoplasma immune response in mice. Parasitol Res 2012; 111:2001-9. [PMID: 22837100 PMCID: PMC3480591 DOI: 10.1007/s00436-012-3047-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 07/10/2012] [Indexed: 12/20/2022]
Abstract
A multicomponent DNA vaccine, encoding Toxoplasma gondii GRA1 and SAG1, was constructed and tested for its ability to confer protection. BALB/c mice were challenged with tachyzoites of the virulent T. gondii RH strain at 4 weeks following the last immunization, and immune responses and survival times were observed. The results show that vaccination by the multicomponent vaccine prolonged survival of mice challenged with the T. gondii RH strain (from average 4.50 ± 0.22 to 7.60 ± 0.74 days); induced high levels of IgG antibody (from 0.252 ± 0.080 to 0.790 ± 0.083), IFN-gamma (from 598.74 ± 67.50 to 853.77 ± 66.74 pg/ml), and IL-2 (from 89.44 ± 10.66 to 192.24 ± 19.90 pg/ml); changed the CD4(+)/CD8(+) lymphocyte ratio (from 1.81 ± 0.14 to 1.09 ± 0.19); and stimulated NK cell-killing activity (from 46.81 ± 3.96 to 64.15 ± 7.71 %). These findings demonstrate that a multicomponent DNA vaccine, encoding GRA1 and SAG1, primes a strong humoral and cellular immune response and enhances protection against T. gondii challenge. The new, combined DNA vaccine provides another means to combat T. gondii infection.
Collapse
|
43
|
Fu Y, Chen Z, Li C, Liu G. Protective immune responses in ducklings induced by a suicidal DNA vaccine of the VP1 gene of duck hepatitis virus type 1. Vet Microbiol 2012; 160:314-8. [PMID: 22819169 DOI: 10.1016/j.vetmic.2012.06.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Revised: 05/10/2012] [Accepted: 06/19/2012] [Indexed: 10/28/2022]
Abstract
A suicidal DNA vaccine based on a Semliki Forest virus (SFV) replicon was evaluated for the development of a vaccine against duck hepatitis virus type 1 (DHV-1). The VP1 gene of DHV-1 was cloned and inserted into pSCA1, an SFV DNA-based replicon vector. The resultant plasmid, pSCA/VP1, was transfected into BHK-21 cells and the antigenicity of the expressed protein was confirmed using an indirect immunofluorescence and western blot assay. Immunogenicity was studied in ducklings. Ducklings were injected intramuscularly two times with pSCA/VP1 at 14 days intervals. Anti-DHV-1 antibodies were detected by ELISA, the lymphocyte proliferation response was also tested by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide method and neutralizing antibodies were measured by microneutralization tests. Our results showed that DHV-1-specific antibodies, neutralizing antibodies and lymphocyte proliferation were well induced in ducklings. Furthermore, all the ducklings were protected against challenge with wild DHV-1. In conclusion, we demonstrate that the suicidal DNA vaccine is a promising vaccine candidate facilitating the prevention of duck hepatitis caused by DHV-1.
Collapse
Affiliation(s)
- Yuzhi Fu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | | | | | | |
Collapse
|
44
|
Okwor I, Mou Z, Liu D, Uzonna J. Protective immunity and vaccination against cutaneous leishmaniasis. Front Immunol 2012; 3:128. [PMID: 22661975 PMCID: PMC3361738 DOI: 10.3389/fimmu.2012.00128] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 05/03/2012] [Indexed: 11/21/2022] Open
Abstract
Although a great deal of knowledge has been gained from studies on the immunobiology of leishmaniasis, there is still no universally acceptable, safe, and effective vaccine against the disease. This strongly suggests that we still do not completely understand the factors that control and/or regulate the development and sustenance of anti-Leishmania immunity, particularly those associated with secondary (memory) immunity. Such an understanding is critically important for designing safe, effective, and universally acceptable vaccine against the disease. Here we review the literature on the correlate of protective anti-Leishmania immunity and vaccination strategies against leishmaniasis with a bias emphasis on experimental cutaneous leishmaniasis.
Collapse
Affiliation(s)
- Ifeoma Okwor
- Department of Medical Microbiology, University of Manitoba Winnipeg, MB, Canada
| | | | | | | |
Collapse
|
45
|
Cui X, Lei T, Yang D, Hao P, Li B, Liu Q. Toxoplasma gondii immune mapped protein-1 (TgIMP1) is a novel vaccine candidate against toxoplasmosis. Vaccine 2012; 30:2282-7. [DOI: 10.1016/j.vaccine.2012.01.073] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 12/28/2011] [Accepted: 01/23/2012] [Indexed: 01/19/2023]
|
46
|
Ding J, Qian W, Liu Q, Liu Q. Multi-epitope recombinant vaccine induces immunoprotection against mixed infection of Eimeria spp. Parasitol Res 2011; 110:2297-306. [PMID: 22200955 DOI: 10.1007/s00436-011-2764-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 12/05/2011] [Indexed: 10/14/2022]
Abstract
Immunity to Eimeria is species-specific, and chickens with immunity to one species of Eimeria remain susceptible to other Eimeria species. This presents a major challenge in the development of effective vaccines against multiple Eimeria species. In this study, we cloned the antigenic epitope of a tachyzoite surface protein gene of Eimeria tenella, a tachyzoite surface protein gene of Eimeria acervulina and the gametocyte protein gene of Eimeria maxima, and constructed prokaryotic and eukaryotic plasmids carrying the multi-epitope antigenic gene. Immunization of chickens with the multivalent DNA and protein conferred partial protection against infection by the three Eimeria species, as shown by increased CD4+ T lymphocytes in the intestinal mucosa, decreased oocyst excretion and intestinal lesions, and increased body weight gain compared with non-immunized controls. The DNA prime-protein boost immunization schedule induced greater cellular immunity and protection from Eimeria infection than immunization with DNA or protein alone. Our findings demonstrated that DNA prime-protein boost immunization with a multivalent vaccine could stimulate protective immunity against challenge infection of multiple Eimeria species. This work provides a promising step towards DNA-protein vaccination against multiple species of pathogens.
Collapse
Affiliation(s)
- Jun Ding
- Key Laboratory of Zoonosis of Ministry of Agriculture and National animal Protozoa Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | | | | | | |
Collapse
|
47
|
Sun W, Song X, Yan R, Xu L, Li X. Vaccination of goats with a glutathione peroxidase DNA vaccine induced partial protection against Haemonchus contortus infection. Vet Parasitol 2011; 182:239-47. [DOI: 10.1016/j.vetpar.2011.05.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 05/10/2011] [Accepted: 05/18/2011] [Indexed: 11/15/2022]
|
48
|
Vaccine candidates for leishmaniasis: A review. Int Immunopharmacol 2011; 11:1464-88. [DOI: 10.1016/j.intimp.2011.05.008] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 04/13/2011] [Accepted: 05/09/2011] [Indexed: 01/08/2023]
|
49
|
Jahangiri A, Rasooli I, Gargari SLM, Owlia P, Rahbar MR, Amani J, Khalili S. An in silico DNA vaccine against Listeria monocytogenes. Vaccine 2011; 29:6948-58. [PMID: 21791233 DOI: 10.1016/j.vaccine.2011.07.040] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2011] [Revised: 07/09/2011] [Accepted: 07/11/2011] [Indexed: 01/14/2023]
Abstract
Listeria monocytogenes causes listeriosis with mortality rate >20%. Listeriolysin-O (LLO), a pore-forming hemolysin, belongs to the family of cholesterol-dependent toxins (CDTX) and plays roles in the pathogenicity. In this study bioinformatic analyses were carried out on LLO sequence as a major immunodominant listerial antigen toward designing a DNA vaccine stimulating cytotoxic T-lymphocytes (CTLs). Mouse and human constructs were designed based on predicted T cell epitopes and MHC class I binders, which were then tandemly fused together. LLO-derived construct codons and a variety of critical gene expression efficiency parameters were optimized. Post-translational modifications such as glycosylation, phosphorylation were analysed. The constructs corresponded to LLO sequences of L. monocytogenes in BLAST search. Neither human nor mouse construct was allergen. Secretory pathway was location of the human construct that enhances immune induction and contribute to the efficacy of the vaccine candidate. mRNAs from optimized DNA sequences of both human and mouse constructs are more stable than the native and are suitable for initiation of translation. The constructs contain several sites for phosphorylation that could improve its degradation and subsequent entry into the MHC class I pathway. Addition of GPI anchor, myristoylation and ubiquitin signals or proline (P), glutamic acid (E), serine (S), threonine (T) (PEST)-like motifs at the N-terminal of constructs increase efficacy of the DNA vaccine. Close physical contact between the favorable immunogen and the suitable CpG oligodeoxynucleotides (CpG ODN) promotes immune response. Vectors for checking the expression of constructs in mammalian cells and for harboring the foreign genes as DNA vaccine are suggested.
Collapse
Affiliation(s)
- Abolfazl Jahangiri
- Department of Biology, Shahed University, Tehran-Qom Express Way, Opposite Imam Khomeini's Shrine, Tehran-3319118651, Iran
| | | | | | | | | | | | | |
Collapse
|
50
|
Toxoplasma gondii: Protective immunity against experimental toxoplasmosis induced by a DNA vaccine encoding the perforin-like protein 1. Exp Parasitol 2011; 128:38-43. [DOI: 10.1016/j.exppara.2011.02.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Revised: 01/10/2011] [Accepted: 02/01/2011] [Indexed: 01/28/2023]
|