1
|
Jearanaiwitayakul T, Warit S, Lekjinda K, Seesen M, Limthongkul J, Midoeng P, Sunintaboon P, Ubol S. The Adjuvant Activity of BCG Cell Wall Cytoskeleton on a Dengue Virus-2 Subunit Vaccine. Vaccines (Basel) 2023; 11:1344. [PMID: 37631912 PMCID: PMC10459381 DOI: 10.3390/vaccines11081344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/29/2023] Open
Abstract
The uneven immunogenicity of the attenuated tetravalent dengue vaccine has made it difficult to achieve balanced protection against all four serotypes of the dengue virus (DENV). To overcome this problem, non-replicative vaccines have come into focus, as their immunogenicity is adjustable. This approach is excellent for multivalent vaccines but commonly faces the issue of low immunogenicity. In this present study, we developed a non-replicating dengue vaccine composed of UV-inactivated dengue virus-2 (UV-DENV-2) and DENV-2 NS1-279 protein encapsidated within nanoparticles. This vaccine candidate was administered in the presence of BCG cell wall cytoskeleton (BCG-CWS) as an adjuvant. We revealed, here, that encapsidated immunogens with BCG-CWS exerted potent activities on both B and T cells and elicited Th-1/Th-2 responses in mice. This was evidenced by BCG-CWS significantly augmenting antibody-mediated complement-fixing activity, strongly stimulating the antigen-specific polyfunctional T cell responses, and activating mixed Th-1/Th-2 responses specific to DENV-2- and NS1-279 antigens. In conclusion, BCG-CWS potently adjuvanted the inactivated DENV-2 and DENV subunit immunogens. The mechanism of adjuvanticity remains unclear. This study revealed the potential use of BCG-CWS in vaccine development.
Collapse
Affiliation(s)
- Tuksin Jearanaiwitayakul
- Department of Clinical Pathology, Faculty of Medicine, Vajira Hospital, Navamindradhiraj University, Bangkok 10300, Thailand;
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (M.S.); (J.L.)
| | - Saradee Warit
- Tuberculosis Research Laboratory, Medical Molecular Biology Research Unit, BIOTEC, National Science and Technology Development Agency, Thailand Science Park, Pathum Thani 12120, Thailand;
| | - Kritsadayut Lekjinda
- Department of Chemistry, Faculty of Science, Mahidol University, Salaya 73170, Thailand; (K.L.); (P.S.)
| | - Mathurin Seesen
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (M.S.); (J.L.)
| | - Jitra Limthongkul
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (M.S.); (J.L.)
| | - Panuwat Midoeng
- Division of Pathology, Army Institute of Pathology, Phramongkutklao Hospital, Bangkok 10400, Thailand;
| | - Panya Sunintaboon
- Department of Chemistry, Faculty of Science, Mahidol University, Salaya 73170, Thailand; (K.L.); (P.S.)
| | - Sukathida Ubol
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; (M.S.); (J.L.)
| |
Collapse
|
2
|
Seesen M, Jearanaiwitayakul T, Limthongkul J, Midoeng P, Sunintaboon P, Ubol S. A bivalent form of nanoparticle-based dengue vaccine stimulated responses that potently eliminate both DENV-2 particles and DENV-2-infected cells. Vaccine 2023; 41:1638-1648. [PMID: 36740559 DOI: 10.1016/j.vaccine.2023.01.062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/03/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
Dengue is the most prevalent mosquito-borne viral disease and continues to be a global public health concern. Although a licensed dengue vaccine is available, its efficacy and safety profile are not satisfactory. Hence, there remains a need for a safe and effective dengue vaccine. We are currently developing a bivalent dengue vaccine candidate. This vaccine candidate is composed of a C-terminus truncated non-structural protein 1 (NS11-279) and envelope domain III (EDIII) of DENV-2 encapsidated in the nanocarriers, N, N, N-trimethyl chitosan nanoparticles (TMC NPs). The immunogenicity of this bivalent vaccine candidate was investigated in the present study using BALB/c mice. In this work, we demonstrate that NS1 + EDIII TMC NP-immunized mice strongly elicited antigen-specific antibody responses (anti-NS1 and anti-EDIII IgG) and T-cell responses (NS1- and EDIII-specific-CD4+ and CD8+ T cells). Importantly, the antibody response induced by NS1 + EDIII TMC NPs provided antiviral activities against DENV-2, including serotype-specific neutralization and antibody-mediated complement-dependent cytotoxicity. Moreover, the significant upregulation of Th1- and Th2-associated cytokines, as well as the increased levels of antigen-specific IgG2a and IgG1, indicated a balanced Th1/Th2 response. Collectively, our findings suggest that NS1 + EDIII TMC NPs induced protective responses that can not only neutralize infectious DENV-2 but also eliminate DENV-2-infected cells.
Collapse
Affiliation(s)
- Mathurin Seesen
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Tuksin Jearanaiwitayakul
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand; Department of Clinical Pathology, Faculty of Medicine Vajira Hospital, Navamindradhiraj University, Bangkok 10300, Thailand
| | - Jitra Limthongkul
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Panuwat Midoeng
- Division of Pathology, Army Institute of Pathology, Phramongkutklao Hospital, Bangkok, Thailand
| | - Panya Sunintaboon
- Department of Chemistry, Faculty of Science, Mahidol University, Salaya, Nakornpatom, Thailand
| | - Sukathida Ubol
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
3
|
Seesen M, Jearanaiwitayakul T, Limthongkul J, Sunintaboon P, Ubol S. Mice immunized with trimethyl chitosan nanoparticles containing DENV-2 envelope domain III elicit neutralizing antibodies with undetectable antibody-dependent enhancement activity. J Gen Virol 2022; 103. [PMID: 35833704 DOI: 10.1099/jgv.0.001768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Dengue is a disease that poses a significant global public health concern. Although a tetravalent live-attenuated dengue vaccine has been licensed, its efficacy is still debated due to evidence of vaccine breakthrough infection. To avoid this issue, dengue vaccines should stimulate a high degree of serotype-specific response. Thus, envelope domain III (EDIII), which contains serotype-specific neutralizing epitopes, is an attractive target for dengue vaccine development. In this study, we investigated how EDIII encapsidated in N, N, N-trimethyl chitosan chloride nanoparticles (TMC NPs) stimulates a serotype-specific response and whether this response exerts a potential in vitro breakthrough infection. The immune response to DENV-2 elicited by EDIII TMC NP-immunized mice was monitored. We demonstrated that immunization with EDIII TMC NPs resulted in a high level of anti-EDIII antibody production. These antibodies included IgG, IgG1, and IgG2a subtypes. Importantly, antibodies from the immunized mice exerted efficient neutralizing activity with undetectable antibody dependent enhancement (ADE) activity. We also found that EDIII TMC NPs activated functional EDIII-specific CD4+ and CD8+ T cell responses. In conclusion, EDIII TMC NPs stimulated humoral immunity with a strong neutralizing antibody response, as well as a cellular immune response against DENV-2.
Collapse
Affiliation(s)
- Mathurin Seesen
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | | - Jitra Limthongkul
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Panya Sunintaboon
- Department of Chemistry, Faculty of Science, Mahidol University, Salaya, Nakornpatom 73170, Thailand
| | - Sukathida Ubol
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
4
|
Jearanaiwitayakul T, Sunintaboon P, Chawengkittikul R, Limthongkul J, Midoeng P, Chaisuwirat P, Warit S, Ubol S. Whole inactivated dengue virus-loaded trimethyl chitosan nanoparticle-based vaccine: immunogenic properties in ex vivo and in vivo models. Hum Vaccin Immunother 2021; 17:2793-2807. [PMID: 33861177 DOI: 10.1080/21645515.2021.1884473] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Dengue virus (DENV) is a mosquito-borne virus that poses an incomparable public health problem, particularly in tropical and subtropical areas. Vaccination remains the most rational measure for controlling DENV infection. In this study, an ultraviolet irradiation (UV)-inactivated DENV-2 carried by N,N,N-trimethyl chitosan nanoparticles (UV-inactivated DENV2 TMC NPs) was investigated as a potential non-replicating dengue vaccine candidate. Using a human ex vivo model, the human monocyte-derived dendritic cells (MoDCs), we showed that TMC served as both a vaccine vehicle and a potent adjuvant. TMC NPs not only efficiently enhanced UV-inactivated DENV2 internalization into MoDCs but also greatly increased the breadth of UV-inactivated DENV2 immunogenicity to drive the maturation of MoDCs. Moreover, UV-inactivated DENV2 TMC NPs were highly immunogenic in mice, inducing greater levels of antibodies (total IgG, IgG1, IgG2a and neutralizing antibodies) and T cells (activated CD4⁺ and CD8⁺ T cells) against DENV-2 compared to soluble DENV-2 immunogens. Notably, the neutralizing activity of sera from mice immunized with UV-inactivated DENV2 TMC NPs was significantly augmented in the presence of complement activation, leading to the strong elimination of both DENV-2 particles and infected cells. We further showed that the immunogenicity of an inactivated dengue-based vaccine was significantly improved in a concentration-dependent manner. These positive results warrant further investigations of this platform of vaccine delivery for tetravalent vaccines or monovalent vaccines in sequential immunizations.
Collapse
Affiliation(s)
| | - Panya Sunintaboon
- Department of Chemistry, Faculty of Science, Mahidol University, Salaya, Thailand
| | | | - Jitra Limthongkul
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Panuwat Midoeng
- Army Institute of Pathology, Phramongkutklao Hospital, Bangkok, Thailand
| | | | - Saradee Warit
- Tuberculosis Research Laboratory, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Sukathida Ubol
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
5
|
Duarte JL, Filippo LDD, Araujo VHS, Oliveira AEMDFM, de Araújo JTC, Silva FBDR, Pinto MC, Chorilli M. Nanotechnology as a tool for detection and treatment of arbovirus infections. Acta Trop 2021; 216:105848. [PMID: 33524384 DOI: 10.1016/j.actatropica.2021.105848] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/21/2021] [Accepted: 01/23/2021] [Indexed: 12/19/2022]
Abstract
Arboviruses are medically important viruses that cause high rates of infection all over the world. In addition, the severity of the symptoms and the inadequate diagnostic methods represent a challenge far beyond eradicating the vector. The lack of specific treatments for arbovirus infections reflects the imminent need for new research for safe and efficient medicines to treat these infections. Nanotechnology is an innovative approach currently used as a platform for developing new treatments, thus improving the biopharmaceutical properties of drugs. It can also be applied to the development of diagnostic devices, improving their detection capacity. The purpose of this paper is to review recent research on the use of nanotechnology for developing new treatments and detection devices for arbovirus infections. Interestingly, it was found that only a few studies report on the use of nanotechnology to treat arbovirus infections and that most of these reports focus on the fabrication of diagnostic tools. Also, some papers report on the use of nanotechnology for the development of vaccines, which in association with mosquito eradication programs could effectively reduce the high rates of infections by these viruses.
Collapse
Affiliation(s)
- Jonatas Lobato Duarte
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Leonardo Delello Di Filippo
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Victor Hugo Sousa Araujo
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Anna Eliza Maciel de Faria Mota Oliveira
- Federal University of Amapá - UNIFAP, Department of Health and biological sciences, Rodovia Juscelino Kubitschek, Km 02, Jardim Marco Zero, Macapá-AP, 68903-361, Brazil
| | - Jennifer Thayanne Cavalcante de Araújo
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Flávia Benini da Rocha Silva
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Mara Cristina Pinto
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil
| | - Marlus Chorilli
- São Paulo State University - UNESP, School of Pharmaceutical Sciences, Rodovia Araraquara Jaú, Km 01 - s/n - Campos Ville - Araraquara/SP -, 14800-903, Brazil.
| |
Collapse
|
6
|
Jearanaiwitayakul T, Sunintaboon P, Chawengkittikul R, Limthongkul J, Midoeng P, Warit S, Ubol S. Nanodelivery system enhances the immunogenicity of dengue-2 nonstructural protein 1, DENV-2 NS1. Vaccine 2020; 38:6814-6825. [PMID: 32829977 DOI: 10.1016/j.vaccine.2020.08.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 06/19/2020] [Accepted: 08/09/2020] [Indexed: 12/11/2022]
Abstract
Nonstructural protein 1 (NS1) of dengue virus (DENV) is currently recognized as a dengue vaccine candidate. Unfortunately, most of non-replicating immunogens typically stimulate unsatisfactory immune responses, thus, the additional adjuvant is required. In this study, C-terminal truncated DENV-2 NS1 loaded in N,N,N, trimethyl chitosan nanoparticles (NS11-279TMC NPs) was prepared through the ionic gelation method. The immunogenicity of NS11-279TMC NPs was investigated using human ex vivo as well as the murine model. Through a human ex vivo model, it was demonstrated in this study that not only can TMC particles effectively deliver NS11-279 protein into monocyte-derived dendritic cells (MoDCs), but also potently stimulate those cells, resulting in increased expression of maturation marker (CD83), costimulating molecules (CD80, CD86 and HLA-DR) and markedly secreted various types of innate immune cytokines/chemokines. Moreover, mice administered with NS11-279TMC NPs strongly elicited both antibody and T cell responses, produced higher levels of IgG, IgG1, IgG2a and potently activated CD8+ T cells, as compared to mice administered with soluble NS11-279. Importantly, we further demonstrated that anti-NS11-279 antibody induced by this platform of NS11-279 effectively eliminated DENV-2 infected cells through antibody dependent complement-mediated cytotoxicity. Significantly, anti-DENV2 NS11-279 antibody exerted cross-antiviral activity against DENV-1 and -4 but not against DENV-3 infected cells. These findings demonstrate that TMC exerts a desirable adjuvant for enhancing delivery and antigenicity of NS1 based dengue vaccine.
Collapse
Affiliation(s)
| | - Panya Sunintaboon
- Department of Chemistry, Faculty of Science, Mahidol University, Salaya, Nakornpatom 73170, Thailand.
| | | | - Jitra Limthongkul
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| | - Panuwat Midoeng
- Division of Pathology, Army Institute of Pathology, Phramongkutklao Hospital, Bangkok, Thailand.
| | - Saradee Warit
- Tuberculosis Research Laboratory, Medical Molecular Biology Research Unit, BIOTEC, National Science and Technology Development Agency, Thailand Science Park, Pathum Thani, Thailand..
| | - Sukathida Ubol
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| |
Collapse
|
7
|
Rungrojcharoenkit K, Sunintaboon P, Ellison D, Macareo L, Midoeng P, Chaisuwirat P, Fernandez S, Ubol S. Development of an adjuvanted nanoparticle vaccine against influenza virus, an in vitro study. PLoS One 2020; 15:e0237218. [PMID: 32760143 PMCID: PMC7410248 DOI: 10.1371/journal.pone.0237218] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 07/22/2020] [Indexed: 12/02/2022] Open
Abstract
Influenza is an infectious respiratory illness caused by influenza viruses. Despite yearly updates, the efficacy of influenza vaccines is significantly curtailed by the virus antigenic drift and antigenic shift. These constant changes to the influenza virus make-up also challenge the development of a universal flu vaccine, which requires conserved antigenic regions shared by influenza viruses of different subtypes. We propose that it is possible to bypass these challenges by the development of an influenza vaccine based on conserved proteins delivered in an adjuvanted nanoparticle system. In this study, we generated influenza nanoparticle constructs using trimethyl chitosan nanoparticles (TMC nPs) as the carrier of recombinant influenza hemagglutinin subunit 2 (HA2) and nucleoprotein (NP). The purified HA2 and NP recombinant proteins were encapsulated into TMC nPs to form HA2-TMC nPs and NP-TMC nPs, respectively. Primary human intranasal epithelium cells (HNEpCs) were used as an in vitro model to measure immunity responses. HA2-TMC nPs, NP-TMC nPs, and HA2-NP-TMC nPs (influenza nanoparticle constructs) showed no toxicity in HNEpCs. The loading efficiency of HA2 and NP into the TMC nPs was 97.9% and 98.5%, respectively. HA2-TMC nPs and NP-TMC nPs more efficiently delivered HA2 and NP proteins to HNEpCs than soluble HA2 and NP proteins alone. The induction of various cytokines and chemokines was more evident in influenza nanoparticle construct-treated HNEpCs than in soluble protein-treated HNEpCs. In addition, soluble factors secreted by influenza nanoparticle construct-treated HNEpCs significantly induced MoDCs maturation markers (CD80, CD83, CD86 and HLA-DR), as compared to soluble factors secreted by protein-treated HNEpCs. HNEpCs treated with the influenza nanoparticle constructs significantly reduced influenza virus replication in an in vitro challenge assay. The results indicate that TMC nPs can be used as influenza vaccine adjuvants and carriers capable of delivering HA2 and NP proteins to HNEpCs.
Collapse
Affiliation(s)
- Kamonthip Rungrojcharoenkit
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Panya Sunintaboon
- Department of Chemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Damon Ellison
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Louis Macareo
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Panuwat Midoeng
- Division of Pathology, Army Institute of Pathology, Phramongkutklao Hospital, Bangkok, Thailand
| | - Preamrudee Chaisuwirat
- Division of Pathology, Army Institute of Pathology, Phramongkutklao Hospital, Bangkok, Thailand
| | - Stefan Fernandez
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
- * E-mail: (SF); (SU)
| | - Sukathida Ubol
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- * E-mail: (SF); (SU)
| |
Collapse
|
8
|
Liu G, Xu X, Jiang L, Ji H, Zhu F, Jin B, Han J, Dong X, Yang F, Li B. Targeted Antitumor Mechanism of C-PC/CMC-CD55sp Nanospheres in HeLa Cervical Cancer Cells. Front Pharmacol 2020; 11:906. [PMID: 32636744 PMCID: PMC7319041 DOI: 10.3389/fphar.2020.00906] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 06/03/2020] [Indexed: 12/20/2022] Open
Abstract
In vitro studies had shown that C-Phycocyanin (C-PC) inhibited cervical cancer HeLa cells growth. We constructed C-PC/CMC-CD55sp nanospheres using C-PC, Carboxymethyl Chitosan (CMC), and CD55 ligand peptide (CD55sp) to allow for targeted antitumor effects against HeLa cells in vitro and in vivo. The characteristics of the nanospheres were determined using FTIR, electron microscopy, and laser particle size analysis. Flow cytometry, laser confocal microscopy and small animal imaging system showed the targeting of C-PC/CMC-CD55sp nanospheres on HeLa cells. Subsequently, the proliferation and apoptosis were analyzed by Cell Counting Kit-8 (CCK-8), flow cytometry, TUNEL assay and electron microscopy. The expression of the apoptosis-related protein was determined using western blot. The stainings of Hematoxylin and Eosin (HE) were employed to evaluate the cell condition of tumor tissue sections. The cytokines in the blood in tumor-bearing nude mice was determined using ELISA. These results showed that C-PC/CMC-CD55sp nanospheres were successfully constructed and targeted HeLa cells. The constructed nanospheres were more effective than C-PC alone in inhibiting the proliferation and inducing apoptosis in HeLa cells. We also found that C-PC/CMC-CD55sp nanospheres had a significant inhibitory effect on the expression of antiapoptotic protein Bcl-2 and a promotion on the transformation of caspase 3 to cleaved caspase 3. C-PC/CMC-CD55sp nanospheres played an important role in tumor suppression, reduced the expression TGF-β, and increased IL-6 and TNF-α. This study demonstrates that the constructed new C-PC/CMC-CD55sp nanospheres exerted targeted antitumor effects in vivo and in vitro which provided a novel idea for application of C-PC, and provided experimental basis for comprehensive targeted treatment of tumors.
Collapse
Affiliation(s)
- Guoxiang Liu
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Xiaohui Xu
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Liangqian Jiang
- Department of Medical Genetics, Linyi People's Hospital, Linyi, China
| | - Huanhuan Ji
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Feng Zhu
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Bingnan Jin
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Jingjing Han
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Xiaolei Dong
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Fanghao Yang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Bing Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China.,Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Chitosan-based particulate systems for drug and vaccine delivery in the treatment and prevention of neglected tropical diseases. Drug Deliv Transl Res 2020; 10:1644-1674. [PMID: 32588282 DOI: 10.1007/s13346-020-00806-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neglected tropical diseases (NTDs) are a diverse group of infections which are difficult to prevent or control, affecting impoverished communities that are unique to tropical or subtropical regions. In spite of the low number of drugs that are currently used for the treatment of these diseases, progress on new drug discovery and development for NTDs is still very limited. Therefore, strategies on the development of new delivery systems for current drugs have been the main focus of formulators to provide improved efficacy and safety. In recent years, particulate delivery systems at micro- and nanosize, including polymeric micro- and nanoparticles, liposomes, solid lipid nanoparticles, metallic nanoparticles, and nanoemulsions, have been widely investigated in the treatment and control of NTDs. Among these polymers used for the preparation of such systems is chitosan, which is a marine biopolymer obtained from the shells of crustaceans. Chitosan has been investigated as a delivery system due to the versatility of its physicochemical properties as well as bioadhesive and penetration-enhancing properties. Furthermore, chitosan can be also used to improve treatment due to its bioactive properties such as antimicrobial, tissue regeneration, etc. In this review, after giving a brief introduction to neglected diseases and particulate systems developed for the treatment and control of NTDs, the chitosan-based systems will be described in more detail and the recent studies on these systems will be reviewed. Graphical abstract.
Collapse
|
10
|
Dengue virus envelope protein domain III induces pro-inflammatory signature and triggers activation of inflammasome. Cytokine 2019; 123:154780. [DOI: 10.1016/j.cyto.2019.154780] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 06/30/2019] [Accepted: 07/09/2019] [Indexed: 01/03/2023]
|
11
|
Chesnut M, Muñoz LS, Harris G, Freeman D, Gama L, Pardo CA, Pamies D. In vitro and in silico Models to Study Mosquito-Borne Flavivirus Neuropathogenesis, Prevention, and Treatment. Front Cell Infect Microbiol 2019; 9:223. [PMID: 31338335 PMCID: PMC6629778 DOI: 10.3389/fcimb.2019.00223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/11/2019] [Indexed: 01/07/2023] Open
Abstract
Mosquito-borne flaviviruses can cause disease in the nervous system, resulting in a significant burden of morbidity and mortality. Disease models are necessary to understand neuropathogenesis and identify potential therapeutics and vaccines. Non-human primates have been used extensively but present major challenges. Advances have also been made toward the development of humanized mouse models, but these models still do not fully represent human pathophysiology. Recent developments in stem cell technology and cell culture techniques have allowed the development of more physiologically relevant human cell-based models. In silico modeling has also allowed researchers to identify and predict transmission patterns and discover potential vaccine and therapeutic candidates. This review summarizes the research on in vitro and in silico models used to study three mosquito-borne flaviviruses that cause neurological disease in humans: West Nile, Dengue, and Zika. We also propose a roadmap for 21st century research on mosquito-borne flavivirus neuropathogenesis, prevention, and treatment.
Collapse
Affiliation(s)
- Megan Chesnut
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Laura S. Muñoz
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Neuroviruses Emerging in the Americas Study, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Georgina Harris
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Dana Freeman
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Lucio Gama
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Carlos A. Pardo
- Division of Neuroimmunology, Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Neuroviruses Emerging in the Americas Study, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David Pamies
- Center for Alternatives to Animal Testing, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
12
|
Limthongkul J, Mapratiep N, Apichirapokey S, Suksatu A, Midoeng P, Ubol S. Insect anionic septapeptides suppress DENV replication by activating antiviral cytokines and miRNAs in primary human monocytes. Antiviral Res 2019; 168:1-8. [PMID: 31075349 DOI: 10.1016/j.antiviral.2019.04.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 04/10/2019] [Accepted: 04/24/2019] [Indexed: 01/06/2023]
Abstract
Dengue viruses (DENVs) have threatened 2/3 of the world population for decades. Thus, combating DENV infection with either antiviral therapy or protective vaccination is an urgent goal. In the present study, we investigated the anti-DENV activity of insect cell-derived anionic septapeptides from C6/36 mosquito cell cultures persistently infected with DENV. These molecules were previously shown to protect C6/36 and Vero cells against DENV infection. We found that treatment with these septapeptides strongly and rapidly downregulated the multiplication of DENV-1 16007, DENV-3 16562, and DENV-4 1036 but not that of DENV-2 16681 in primary human monocytes. This inhibitory effect was likely mediated through various routes including the increased production of antiviral cytokines (IFN-I), activation of mononuclear cell migration, and upregulation of the expression of antiviral miRNAs (has-miR-30e*, has-miR-133a, and has-miR-223) and inflammation-related miRNAs (has-miR-146a and has-miR-147). In conclusion, anionic septapeptides exerted anti-DENV activity in human monocytes through the upregulation of innate immune responses and the activation of several previously reported antiviral and inflammation-related miRNAs.
Collapse
Affiliation(s)
- Jitra Limthongkul
- Department of Microbiology, Faculty of Science, Mahidol University, 272 Rama 6 Road, Bangkok, 10400, Thailand.
| | - Nithipong Mapratiep
- Department of Microbiology, Faculty of Science, Mahidol University, 272 Rama 6 Road, Bangkok, 10400, Thailand.
| | - Suttikarn Apichirapokey
- Department of Microbiology, Faculty of Science, Mahidol University, 272 Rama 6 Road, Bangkok, 10400, Thailand.
| | - Ampa Suksatu
- Department of Microbiology, Faculty of Science, Mahidol University, 272 Rama 6 Road, Bangkok, 10400, Thailand.
| | - Panuwat Midoeng
- Army Institute of Pathology, Phramongkutklao Hospital, Bangkok, Thailand.
| | - Sukathida Ubol
- Department of Microbiology, Faculty of Science, Mahidol University, 272 Rama 6 Road, Bangkok, 10400, Thailand.
| |
Collapse
|