1
|
Amiryaghoubi N, Fathi M, Safary A, Javadzadeh Y, Omidi Y. In situ forming alginate/gelatin hydrogel scaffold through Schiff base reaction embedded with curcumin-loaded chitosan microspheres for bone tissue regeneration. Int J Biol Macromol 2024; 256:128335. [PMID: 38007028 DOI: 10.1016/j.ijbiomac.2023.128335] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/21/2023] [Accepted: 11/13/2023] [Indexed: 11/27/2023]
Abstract
In this study, we developed a biocompatible composite hydrogel that incorporates microspheres. This was achieved using a Schiff base reaction, which combines the amino and aldehyde groups present in gelatin (Gel) and oxidized alginate (OAlg). We suggest this hydrogel as a promising scaffold for bone tissue regeneration. To further boost its osteogenic capabilities and mechanical resilience, we synthesized curcumin (Cur)-loaded chitosan microspheres (CMs) and integrated them into the Gel-OAlg matrix. This formed a robust composite gel framework. We conducted comprehensive evaluations of various properties, including gelation time, morphology, compressive strength, rheological behavior, texture, swelling rate, in vitro degradation, and release patterns. A remarkable observation was that the inclusion of 30 mg/mL Cur-CMs significantly enhanced the hydrogel's mechanical and bioactive features. Over three weeks, the Gel-OAlg/Cur-CMs (30) composite showed a cumulative curcumin release of 35.57%. This was notably lower than that observed in standalone CMs and Gel-OAlg hydrogels. Additionally, the Gel-OAlg/Cur-CMs (30) hydrogel presented a reduced swelling rate and weight loss relative to hydrogels devoid of Cur-CMs. On the cellular front, the Gel-OAlg/Cur-CMs (30) hydrogel showcased superior biocompatibility. It also displayed increased calcium deposition, alkaline phosphatase (ALP) activity, and elevated osteogenic gene expression in human bone marrow mesenchymal stem cells (hBMSCs). These results solidify its potential as a scaffold for bone tissue regeneration.
Collapse
Affiliation(s)
- Nazanin Amiryaghoubi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Marziyeh Fathi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Azam Safary
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yousef Javadzadeh
- Biotechnology Research Center and Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran.
| | - Yadollah Omidi
- Department of Pharmaceutical Sciences, Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA.
| |
Collapse
|
2
|
Ma Y, Zhao Y, Chen R, Sun W, Zhang Y, Qiao H, Chang Y, Kang S, Zhang Y. Mucosal immunity of mannose-modified chitosan microspheres loaded with the nontyepable Haemophilus influenzae outer membrane protein P6 in BALB/c mice. PLoS One 2022; 17:e0269153. [PMID: 35687548 PMCID: PMC9187061 DOI: 10.1371/journal.pone.0269153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 05/16/2022] [Indexed: 11/18/2022] Open
Abstract
Nontypeable Haemophilus influenzae (NTHi) is a common opportunistic pathogen that colonizes the nasopharynx. NTHi infections result in enormous global morbidity in two clinical settings: otitis media in children and acute exacerbation of chronic obstructive pulmonary disease (COPD) in adults. Thus, there is an urgent need to design and develop effective vaccines to prevent morbidity and reduce antibiotic use. The NTHi outer membrane protein P6, a potential vaccine candidate, is highly conserved and effectively induces protective immunity. Here, to enhance mucosal immune responses, P6-loaded mannose-modified chitosan (MC) microspheres (P6-MCMs) were developed for mucosal delivery. MC (18.75%) was synthesized by the reductive amination reaction method using sodium cyanoborohydride (NaBH3CN), and P6-MCMs with an average size of 590.4±16.2 nm were successfully prepared via the tripolyphosphate (TPP) ionotropic gelation process. After intranasal immunization with P6-MCMs, evaluation of humoral immune responses indicated that P6-MCMs enhance both systemic and mucosal immune responses. Evaluation of cellular immune responses indicated that P6-MCMs enhance cellular immunity and trigger a mixed Th1/Th2-type immune response. Importantly, P6-MCMs also trigger a Th17-type immune response. They are effective in promoting lymphocyte proliferation and differentiation without toxicity in vitro. The results also demonstrate that P6-MCMs can effectively induce MHC class I- and II-restricted cross-presentation, promoting CD4+-mediated Th immune responses and CD8+-mediated cytotoxic T lymphocyte (CTL) immune responses. Evaluation of protective immunity indicated that immunization with P6-MCMs can reduce inflammation in the nasal mucosa and the lung and prevent NTHi infection. In conclusion, MCMs are a promising adjuvant-delivery system for vaccines against NTHi.
Collapse
Affiliation(s)
- Yushuai Ma
- Institute of Pathogen Biology and Immunology, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Ying Zhao
- Institute of Pathogen Biology and Immunology, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Rui Chen
- Institute of Pathogen Biology and Immunology, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Wanru Sun
- Institute of Pathogen Biology and Immunology, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Yanxia Zhang
- Department of Microbiology, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Haixia Qiao
- Department of Microbiology, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Yueli Chang
- Department of Microbiology, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Shaoping Kang
- Department of Immunology, Hebei North University, Zhangjiakou, Hebei Province, China
| | - Yutuo Zhang
- Department of Microbiology, Hebei North University, Zhangjiakou, Hebei Province, China
- * E-mail:
| |
Collapse
|
3
|
Wu XQ, Zhang WN, Hao MZ, Liu XP, Xiao J, Wang TF, Dong YZ, Zhao J. How Chinese Herbal Medicine Prevents Epidemics: From Ancient Pestilences to COVID-19 Pandemic. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1017-1044. [PMID: 34107860 DOI: 10.1142/s0192415x2150049x] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The ongoing coronavirus disease 2019 (COVID-19) pandemic calls for effective control and prevention. Chinese medicine (CM) has developed systematic theories and approaches for infectious disease prevention over 2000 years. Here, we review and analyze Chinese herbal medicines (CHM) used in infectious disease prevention from ancient pestilences to modern epidemics and pandemics to share cumulative preventive medical experience. A total of 829 formulas, including 329 herbs from 189 ancient books, 131 formulas with 152 herbs, and 13 Chinese patent medicines (CPM) from 30 official Chinese prevention programs used in ancient epidemics, SARS, influenza and COVID-19 prevention, were reviewed and analyzed. Preventive CHM mainly has four functions and can be taken orally or applied externally. CHM that kill pathogens (Realgar [Xionghuang], Cyrtomium Fortunei J. Sm[Guanzhong]) were commonly used externally for disinfection in ancient prevention while CHM tonifying Qi (Astragali Radix [Huangq], Glycyrrhizae Radix et Rhizoma [Gancao]) are used for modern prevention. Taking CHM that expel pathogens (Realgar [Xionghuang], Lonicerae Japonicae Flos[Jinyinhua]) and CHM eliminating dampness (Atractylodis Rhizoma [Cangzhu], Pogostemonis Herba[Guanghuoxiang]) have been commonly used from ancient times to COVID-19. Damp toxins are a common characteristic of infectious diseases such as SARS and COVID-19. Thus, taking CHM expelling damp toxins and tonifying Qi are the main methods for SARS and COVID-19 prevention. CHM with different approaches have been widely used in infectious disease prevention from ancient times to the present. Multiple CM prevention methods may provide new perspectives for future pandemics.
Collapse
Affiliation(s)
- Xia-Qiu Wu
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, P. R. China
| | - Wei-Na Zhang
- Institute of Information on Traditional Chinese Medicine, Beijing 100700, P. R. China
| | - Ming-Zhao Hao
- Institute of Acupuncture and Moxibustion, Beijing 100700, P. R. China
| | - Xi-Ping Liu
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, P. R. China
| | - Jing Xiao
- College of Public Health, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, P. R. China
| | - Teng-Fei Wang
- Institute of Information on Traditional Chinese Medicine, Beijing 100700, P. R. China
| | - Yi-Zhi Dong
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P. R. China
| | - Jing Zhao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, P. R. China
| |
Collapse
|
4
|
The Application of Mucoadhesive Chitosan Nanoparticles in Nasal Drug Delivery. Mar Drugs 2020; 18:md18120605. [PMID: 33260406 PMCID: PMC7759871 DOI: 10.3390/md18120605] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/21/2020] [Accepted: 11/26/2020] [Indexed: 12/15/2022] Open
Abstract
Mucosal delivery of antigens can induce both humoral and cellular immune responses. Particularly, the nasal cavity is a strongly inductive site for mucosal immunity among several administration routes, as it is generally the first point of contact for inhaled antigens. However, the delivery of antigens to the nasal cavity has some disadvantages such as rapid clearance and disposition of inhaled materials. For these reasons, remarkable efforts have been made to develop antigen delivery systems which suit the nasal route. The use of nanoparticles as delivery vehicles enables protection of the antigen from degradation and sustains the release of the loaded antigen, eventually resulting in improved vaccine and/or drug efficacy. Chitosan, which exhibits low toxicity, biodegradability, good cost performance, and strong mucoadhesive properties, is a useful material for nanoparticles. The present review provides an overview of the mucosal immune response induced by nanoparticles, recent advances in the use of nanoparticles, and nasal delivery systems with chitosan nanoparticles.
Collapse
|
5
|
Elicitation of Th1/Th2 related responses in mice by chitosan nanoparticles loaded with Brucella abortus malate dehydrogenase, outer membrane proteins 10 and 19. Int J Med Microbiol 2019; 310:151362. [PMID: 31676233 DOI: 10.1016/j.ijmm.2019.151362] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 09/04/2019] [Accepted: 10/10/2019] [Indexed: 11/22/2022] Open
Abstract
Brucella spp. is the causative agent of brucellosis, one of the worldwide diseases. The pathogen infects humans and animals mainly through the digestive or respiratory tract. Therefore, induction of mucosal immunity is required as the first line of defense. In this study, three Brucella abortus recombinant proteins, malate dehydrogenase (rMdh), outer membrane proteins (rOmp) 10 and 19 were loaded in mucoadhesive chitosan nanoparticles (CNs) and induction of mucosal and systemic immunity were investigated after intranasal immunization of BALB/c mice. These antigens were also coimmunized as cocktail (rCocktail) to evaluate multiple antigen specific vaccine candidates. At 6-weeks post-immunization (wpi), antigen specific total IgG was increased in all of the immunized groups, predominantly IgG1. In addition, spleenocyte from rMdh-, rOmp19-, and rCocktail-immunized groups significantly produced IFN-γ and IL-4 suggesting the induction of a mixed Th1-Th2 response. For mucosal immunity, anti-Mdh IgA from nasal washes and fecal excretions, and anti-Omps IgA from sera, nasal washes, genital secretions and fecal excretions were significantly increased in single antigen immunized groups. In the rCocktail-immunized group, anti-Mdh IgA were significantly increased while anti-Omps IgA was not. Collectively, this study indicates that comprise of B. abortus antigen-loaded CNs elicited the antigen-specific IgA with a Th2-polarized immune responses and combination of the highly immunogenic antigens elicited IgG specific to each type of antigen.
Collapse
|
6
|
Soh SH, Shim S, Im YB, Park HT, Cho CS, Yoo HS. Induction of Th2-related immune responses and production of systemic IgA in mice intranasally immunized with Brucella abortus malate dehydrogenase loaded chitosan nanoparticles. Vaccine 2019; 37:1554-1564. [PMID: 30792035 DOI: 10.1016/j.vaccine.2019.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/02/2019] [Accepted: 02/07/2019] [Indexed: 12/28/2022]
Abstract
The aim of this study was to investigate the induction of mucosal immune responses by an important Brucella abortus antigen, malate dehydrogenase (Mdh), loaded in mucoadhesive chitosan nanoparticles (CNs) and immunized intranasally in a BALB/c mouse model. The production of cytokines was investigated in human leukemic monocyte cells (THP-1 cells) after stimulation with the nanoparticles. Mdh-loaded CNs (CNs-Mdh) induced higher interleukin (IL)-6 production than unloaded antigens and TF loaded CNs (CNs-TF). Using ELISpot to quantify cytokines and antibody-secreting cells in the intranasally immunized mice, IL-4 and IgG-secreting cells were found to be significantly increased at 4 weeks and 6 weeks post-immunization in the CNs-Mdh immunized group, respectively. Increases in Mdh-specific IgG, IgG1, and IgG2a antibodies were confirmed at 6 weeks after immunization, indicating a predominant IgG1 response. Analysis of the mucosal immune response in the intranasally immunized mice revealed, Mdh-specific IgA and total IgA in the nasal washes, genital secretions, fecal extracts and sera that were remarkably increased in the CNs-Mdh-immunized group compared to the CNs-TF-immunized group except total IgA of nasal wash. Therefore, the results indicated that the intranasal immunization of CNs-loaded B. abortus Mdh antigen effectively induced antigen-specific mucosal immune responses through the elicitation of Th2-related immune responses.
Collapse
Affiliation(s)
- Sang Hee Soh
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.
| | - Soojin Shim
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.
| | - Young Bin Im
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.
| | - Hong-Tae Park
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.
| | - Chong-Su Cho
- Department of Agricultural Biotechnology & Research Institute for Agriculture & Life Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| | - Han Sang Yoo
- Department of Infectious Diseases, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
7
|
Wu C, Liu X, Sun R, Qin Y, Liu Z, Yang S, Tang T, Zhu Z, Yu D, Liu F. Targeting Anion Exchange of Osteoclast, a New Strategy for Preventing Wear Particles Induced- Osteolysis. Front Pharmacol 2018; 9:1291. [PMID: 30459624 PMCID: PMC6232501 DOI: 10.3389/fphar.2018.01291] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/22/2018] [Indexed: 12/30/2022] Open
Abstract
Joint replacement is essential for the treatment of serious joint disease. However, prosthetic failure remains an important clinical issue, with periprosthesis osteolysis (PO), caused by osteoclastic bone resorption induced by wear particles, being the leading cause of failure. Nuclear factor of activated T cells c1 (NFATc1) appears to play an important role in wear particle-induced osteoclastogenesis, with bicarbonate/chloride exchanger, solute carrier family 4, anion exchanger, member 2, (SLC4A2) being upregulated during osteoclastogenesis in an NFATc1-dependent manner. Anion exchange mediated by SLC4A2 in osteoclasts could affect the bone resorption activity by regulating pHi. This study investigated the role and mechanism of SLC4A2 in wear particle-induced osteoclast differentiation and function in vitro. The use of 4, 4'-diisothiocyano-2,2'-stilbenedisulfonic acid (DIDS), an anion exchange inhibitor, suppressed wear particle-induced PO in vivo. Furthermore, controlled release of DIDS from chitosan microspheres can strengthen the PO therapy effect. Therefore, anion exchange mediated by osteoclastic SLC4A2 may be a potential therapeutic target for the treatment of aseptic loosening of artificial joints.
Collapse
Affiliation(s)
- Chuanlong Wu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuqiang Liu
- Department of Orthopaedics, The First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Ruixin Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunhao Qin
- Department of Orthopaedics, Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiqing Liu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengbing Yang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Tang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenan Zhu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Degang Yu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengxiang Liu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedics, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Xu J, Li S, Wang X, Liu J, Shan P, Zhou Y, Zhao J, Wang Z, Xu C, Chen M, Chen Z, Zhao K, Qu D. Systemic and mucosal humoral immune responses induced by the JY-adjuvanted nasal spray H7N9 vaccine in mice. Emerg Microbes Infect 2018; 7:140. [PMID: 30076293 PMCID: PMC6076272 DOI: 10.1038/s41426-018-0133-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 06/05/2018] [Accepted: 06/13/2018] [Indexed: 12/20/2022]
Abstract
Since the first case of human avian influenza A (H7N9) virus infection in 2013, five H7N9 epidemics have occurred in China, all of which caused severe diseases, including pneumonia and acute respiratory distress syndrome, and the fatality rates of these epidemics were as high as 30-40%. To control the prevalence of H7N9 influenza, an effective vaccine is urgently needed. In the present study, we used chitosan and recombinant human interleukin-2 as an adjuvant (JY) to promote the systemic and mucosal immune responses induced by the H7N9 vaccine. Mice were immunized intranasally with the inactivated split influenza A (H7N9) virus (A/Shanghai/02/2013) vaccine with or without JY. The hemagglutination inhibition (HI) titers of mice immunized with the JY-adjuvanted vaccine were significantly higher than those of mice immunized with the vaccine without adjuvant (21.11 ± 9.58 vs. 5.04 ± 3, P < 0.05). The JY-adjuvanted H7N9 nasal spray vaccine induced higher HI titers (8 ± 0.82 vs. 6.7 ± 0.67, P = 0.0035) than those did the poly (I:C)-adjuvanted H7N9 vaccine or the LTB-adjuvanted H7N9 vaccine (8 ± 0.82 vs. 6.9 ± 0.88, P = 0.0186). The optimal immunization regimen for the nasal spray H7N9 vaccine was determined to be a 21-day interval between the primary immunization and booster, with a dose of 4.5 μg hemagglutinin per mouse. The immunogenicities of the nasal spray H7N9 vaccine and intramuscular vaccine (containing only the inactivated split virus) were compared in mice. Two doses of the nasal spray H7N9 vaccine induced higher titers of HI (6.7 ± 0.67 vs. 5.3 ± 1.16, P = 0.004) and anti-HA IgG in sera (19.26 ± 0.67 vs. 13.97 ± 0.82, P < 0.0001) and of anti-HA sIgA (7.13 ± 2.54 vs. 0, P = 0.0000) in bronchoalveolar lavage fluid (BALF) than one dose of intramuscular H7N9 vaccine 3 weeks after the last immunization. However, when we immunized the mice with two doses of both vaccines separately, the nasal spray H7N9 vaccine induced higher titers of anti-HA IgG (19.26 ± 0.67 vs. 17.56 ± 0.57, P < 0.0001) and anti-HA sIgA (7.13 ± 2.54 vs. 4.02 ± 0.33, P = 0.0026) than did the intramuscular H7N9 vaccine, and there was no difference in HI titer between the two groups (P = 0.3745). This finding indicates that the JY-adjuvanted nasal spray H7N9 vaccine induced not only the systemic immune response but also a local mucosal response, which may improve the efficacy of H7N9 influenza prevention through respiratory tract transmission.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antibodies, Viral/immunology
- Female
- Humans
- Immunity, Humoral
- Immunity, Mucosal
- Influenza A Virus, H7N9 Subtype/genetics
- Influenza A Virus, H7N9 Subtype/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Mice
- Mice, Inbred BALB C
- Nasal Sprays
- Vaccination
Collapse
Affiliation(s)
- Jing Xu
- Key Laboratory of Medical Molecular Virology of MOE and MOH, School of Basic Medical Sciences, Fudan University, Shanghai, China
- China National Vaccine and Serum Institute, Beijing, China
| | - Shuxiang Li
- China National Vaccine and Serum Institute, Beijing, China
| | - Xinyi Wang
- China National Vaccine and Serum Institute, Beijing, China
| | - Jing Liu
- China National Vaccine and Serum Institute, Beijing, China
| | - Pu Shan
- China National Vaccine and Serum Institute, Beijing, China
| | - Ya Zhou
- Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jing Zhao
- Beijing JDK Bio-Tech Institute, Beijing, China
| | - Zhibiao Wang
- China National Vaccine and Serum Institute, Beijing, China
| | - Cui Xu
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Meili Chen
- Beijing Bio-Institute Biological Products Co., Ltd., Beijing, China
| | - Ze Chen
- Shanghai Institute of Biological Products Co., Ltd., Shanghai, China
| | - Kai Zhao
- China National Vaccine and Serum Institute, Beijing, China.
| | - Di Qu
- Key Laboratory of Medical Molecular Virology of MOE and MOH, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Singh B, Maharjan S, Cho KH, Cui L, Park IK, Choi YJ, Cho CS. Chitosan-based particulate systems for the delivery of mucosal vaccines against infectious diseases. Int J Biol Macromol 2018; 110:54-64. [DOI: 10.1016/j.ijbiomac.2017.10.101] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 09/23/2017] [Accepted: 10/11/2017] [Indexed: 12/22/2022]
|
10
|
Intrinsic parameters for the synthesis and tuned properties of amphiphilic chitosan drug delivery nanocarriers. J Control Release 2017. [DOI: 10.1016/j.jconrel.2017.06.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
11
|
Kavaliauskis A, Arnemo M, Speth M, Lagos L, Rishovd AL, Estepa A, Griffiths G, Gjøen T. Protective effect of a recombinant VHSV-G vaccine using poly(I:C) loaded nanoparticles as an adjuvant in zebrafish (Danio rerio) infection model. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 61:248-257. [PMID: 27084059 DOI: 10.1016/j.dci.2016.04.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 04/09/2016] [Accepted: 04/09/2016] [Indexed: 06/05/2023]
Abstract
There is a constant need to increase the efficiency of vaccines in the aquaculture industry. Although several nano-based vaccine formulations have been reported, to the best of our knowledge so far only one of them have been implemented in the industry. Here we report on chitosan-poly(I:C) nanoparticles (NPs) that could be used as a non-specific adjuvant in antiviral vaccines in aquaculture. We have characterized the physical parameters of the NPs, studied the in vivo and in vitro bio-distribution of fluorescent NPs and verified NP uptake by zebrafish leucocytes. We used the zebrafish model to test the protective efficiency of the recombinant glycoprotein G (rgpG) of VHSV compared to inactivated whole virus (iV) against VHSV using NPs as an adjuvant in both formulations. In parallel we tested free poly(I:C) and rgpG (pICrgpG), and free chitosan and rgpG (CSrgpG) vaccine formulations. While the iV group (with NP adjuvant) provided the highest overall survival, all vaccine formulations with poly(I:C) provided a significant protection against VHSV; possibly through an early induction of an anti-viral state. Our results suggest that chitosan-poly(I:C) NPs are a promising adjuvant candidate for future vaccine formulations.
Collapse
Affiliation(s)
- Arturas Kavaliauskis
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316 Oslo, Norway
| | - Marianne Arnemo
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316 Oslo, Norway
| | - Martin Speth
- Department of Biosciences, University of Oslo, P.O. Box 1066 Blindern, 0371 Oslo, Norway
| | - Leidy Lagos
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316 Oslo, Norway
| | - Anne-Lise Rishovd
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316 Oslo, Norway
| | | | - Gareth Griffiths
- Department of Biosciences, University of Oslo, P.O. Box 1066 Blindern, 0371 Oslo, Norway
| | - Tor Gjøen
- Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, P.O. Box 1068 Blindern, 0316 Oslo, Norway.
| |
Collapse
|
12
|
Natarajan SB, Kim YS, Hwang JW, Park PJ. Immunomodulatory properties of shellfish derivatives associated with human health. RSC Adv 2016. [DOI: 10.1039/c5ra26375a] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Some vital components of marine shellfish are documented as an important source for both nutritional and pharmacological applications.
Collapse
Affiliation(s)
| | - Yon-Suk Kim
- Department of Biotechnology
- Konkuk University
- Chungju 380-701
- Republic of Korea
- Nokyong Research Centre
| | - Jin-Woo Hwang
- Department of Biotechnology
- Konkuk University
- Chungju 380-701
- Republic of Korea
- Nokyong Research Centre
| | - Pyo-Jam Park
- Department of Biotechnology
- Konkuk University
- Chungju 380-701
- Republic of Korea
- Nokyong Research Centre
| |
Collapse
|
13
|
Wang C, Liu L, Zhang Z, Yan Z, Yu C, Shao M, Jiang X, Chi S, Wei K, Zhu R. Immunological and protective effects of Bordetella bronchiseptica subunit vaccines based on the recombinant N-terminal domain of dermonecrotic toxin. Int Immunopharmacol 2015; 28:952-9. [PMID: 26337750 DOI: 10.1016/j.intimp.2015.08.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 08/11/2015] [Accepted: 08/13/2015] [Indexed: 01/21/2023]
Abstract
Dermonecrotic toxin (DNT) produced by Bordetella bronchiseptica (B. bronchiseptica) can cause clinical turbinate atrophy in swine and induce dermonecrotic lesions in model mice. We know that the N-terminal of DNT molecule contains the receptor-binding domain, which facilitates binding to the target cells. However, we do not know whether this domain has sufficient immunogenicity to resist B. bronchiseptica damage and thereby to develop a subunit vaccine for the swine industry. In this study, we prokaryotically expressed the recombinant N-terminal of DNT from B. bronchiseptica (named DNT-N) and prepared it for the subunit vaccine to evaluate its immunogenicity. Taishan Pinus massoniana pollen polysaccharide (TPPPS), a known immunomodulator, was used as the adjuvant to examine its immune-conditioning effects. At 49 d after inoculation, 10 mice from each group were challenged with B. bronchiseptica, and another 10 mice were intradermally challenged with native DNT, to examine the protection imparted by the vaccines. The immune parameters (T-lymphocyte counts, cytokine secretions, serum antibody titers, and survival rates) and skin lesions were determined. The results showed that pure DNT-N vaccine significantly induced immune responses and had limited ability to resist the B. bronchiseptica and DNT challenge, whereas the mice administered with TPPPS or Freund's incomplete adjuvant vaccine could induce higher levels of the above immune parameters. Remarkably, the DNT-N vaccine combined with TPPPS adjuvant protected the mice effectively to prevent B. bronchiseptica infection. Our findings indicated that DNT-N has potential for development as an effective subunit vaccine to counteract the damage of B. bronchiseptica infection, especially when used conjointly with TPPPS.
Collapse
Affiliation(s)
- Chuanwen Wang
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Liping Liu
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Zhen Zhang
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Zhengui Yan
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Cuilian Yu
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Mingxu Shao
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Xiaodong Jiang
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Shanshan Chi
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, Shandong, China
| | - Kai Wei
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, Shandong, China.
| | - Ruiliang Zhu
- College of Animal Science and Technology, Shandong Agricultural University, Taian 271018, Shandong, China.
| |
Collapse
|
14
|
Gong Y, Tao L, Wang F, Liu W, Jing L, Liu D, Hu S, Xie Y, Zhou N. Chitosan as an adjuvant for a Helicobacter pylori therapeutic vaccine. Mol Med Rep 2015; 12:4123-4132. [PMID: 26095723 PMCID: PMC4526086 DOI: 10.3892/mmr.2015.3950] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 05/13/2015] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to delineate the therapeutic effect of a Helicobacter pylori vaccine with chitosan as an adjuvant, as well as to identify the potential mechanism against H. pylori infection when compared with an H. pylori vaccine, with cholera toxin (CT) as an adjuvant. Mice were first infected with H. pylori and, following the establishment of an effective infection model, were vaccinated using an H. pylori protein vaccine with chitosan as an adjuvant. Levels of H. pylori colonization, H. pylori‑specific antibodies and cytokines were determined by enzyme‑linked immunosorbent assay. The TLR4 and Foxp3 mRNA and protein levels were determined by reverse transcription polymerase chain reaction and immunohistochemistry, respectively. It was identified that the H. pylori elimination rate of the therapeutic vaccine with chitosan as an adjuvant (58.33%) was greater than the therapeutic vaccine with CT as an adjuvant (45.45%). The therapeutic H. pylori vaccine with chitosan as an adjuvant induced significantly greater antibody and cytokine levels when compared with the control groups. Notably, the IL‑10 and IL‑4 levels in the groups with chitosan as an adjuvant to the H. pylori vaccine were significantly greater than those in the groups with CT as an adjuvant. The mRNA expression levels of TLR4 and Foxp3 were significantly elevated in the mice that were vaccinated with chitosan as an adjuvant to the H. pylori vaccine, particularly in mice where the H. pylori infection had been eradicated. The H. pylori vaccine with chitosan as an adjuvant effectively increased the H. pylori elimination rate, the humoral immune response and the Th1/Th2 cell immune reaction; in addition, the therapeutic H. pylori vaccine regulated the Th1 and Th2 response. The significantly increased TLR4 expression and decreased CD4+CD25+Foxp3+Treg cell number contributed to the immune clearance of the H. pylori infection. Thus, the present findings demonstrate that in mice the H. pylori vaccine with chitosan as an adjuvant exerts an equivalent immunotherapeutic effect on H. pylori infection when compared with the H. pylori vaccine with CT as an adjuvant.
Collapse
Affiliation(s)
- Yanfeng Gong
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Liming Tao
- Department of Obstetrics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Fucai Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Wei Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Lei Jing
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Dongsheng Liu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Sijun Hu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Yong Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Nanjin Zhou
- Department of Biochemistry and Molecular Biology, Jiangxi Medical Science Institute, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
15
|
Zhu S, Lu P, Liu H, Chen P, Wu Y, Wang Y, Sun H, Zhang X, Xia Q, Heng BC, Zhou Y, Ouyang HW. Inhibition of Rac1 activity by controlled release of NSC23766 from chitosan microspheres effectively ameliorates osteoarthritis development in vivo. Ann Rheum Dis 2015; 74:285-93. [PMID: 24257023 PMCID: PMC4283675 DOI: 10.1136/annrheumdis-2013-203901] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 08/21/2013] [Accepted: 10/26/2013] [Indexed: 01/14/2023]
Abstract
BACKGROUND Osteoarthritis (OA) is a degenerative joint disease characterised by cartilage degradation and chondrocyte hypertrophy. A recent study showed that Rac1 promoted expression of MMP13 and chondrocyte hypertrophy within the growth plate. These findings warrant further investigations on the roles of Rac1 in OA development and therapy in animal models. OBJECTIVE To investigate the role and mechanistic pathway of Rac1 involvement in pathological changes of OA chondrocytes in vitro and OA development in vivo, as well as to develop a strategy of modulating Rac1 activity for OA treatment. MATERIAL AND METHODS OA and normal cartilage from human or mice were used for immunohistochemical study and Rac1 activity assay. Chondrocytes treated with IL1β and the untreated control were subjected to the Rac1 activity assay. Chondrocytes transfected with CA-Rac1, DN-Rac1 or GFP were cultured under conditions for inducing calcification. To evaluate the effect of Rac1 in OA development, an OA model was created by anterior cruciate ligament transection in mice. CA-Rac1, DN-Rac1 and GFP lentivirus, or NSC23766, were injected intra-articularly. Joints were subjected to histological analysis. RESULTS It was found that there is aberrant Rac1 activation in human OA cartilage. Rac1 activity could also be elevated by IL1β. Additionally, activated Rac1 promoted expression of MMP13, ADAMTS-5 and COLX by chondrocytes, partially through the β-catenin pathway. Moreover, activation of Rac1 in knee joints by CA-Rac1 lentivirus accelerated OA progression, while inhibition of Rac1 activity by DN-Rac1 lentivirus or Rac1 inhibitor NSC23766 delayed OA development. Therefore, we developed a strategy of controlled release of NSC23766 from chitosan microspheres to OA joints, which effectively protected cartilage from destruction. CONCLUSIONS These findings demonstrated that Rac1 activity is implicated in OA development. Also, controlled release of Rac1 inhibitor is a promising strategy for OA treatment.
Collapse
MESH Headings
- ADAM Proteins/genetics
- ADAMTS5 Protein
- Aminoquinolines/administration & dosage
- Aminoquinolines/pharmacology
- Animals
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Arthritis, Experimental/therapy
- Calcinosis/metabolism
- Calcinosis/pathology
- Cartilage, Articular/drug effects
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Chitosan
- Chondrocytes/metabolism
- Core Binding Factor Alpha 1 Subunit/genetics
- Delayed-Action Preparations
- Gene Expression Profiling
- Humans
- Hypertrophy
- Matrix Metalloproteinase 13/genetics
- Mice
- Microspheres
- Neuropeptides/antagonists & inhibitors
- Neuropeptides/metabolism
- Osteoarthritis, Knee/metabolism
- Osteoarthritis, Knee/pathology
- Osteoarthritis, Knee/therapy
- Pyrimidines/administration & dosage
- Pyrimidines/pharmacology
- RNA, Messenger/genetics
- rac1 GTP-Binding Protein/antagonists & inhibitors
- rac1 GTP-Binding Protein/genetics
- rac1 GTP-Binding Protein/metabolism
Collapse
Affiliation(s)
- Shouan Zhu
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ping Lu
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huanhuan Liu
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Pengfei Chen
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yan Wu
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yanyan Wang
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Heng Sun
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaolei Zhang
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Orthopaedics, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qingqing Xia
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Boon Chin Heng
- Department of Biosystems Science & Engineering, ETH-Zurich, Mattenstrasse, Switzerland
| | - Yiting Zhou
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hong Wei Ouyang
- Center for Stem Cell and Tissue Engineering, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
16
|
Ganguly K, Chaturvedi K, More UA, Nadagouda MN, Aminabhavi TM. Polysaccharide-based micro/nanohydrogels for delivering macromolecular therapeutics. J Control Release 2014; 193:162-73. [DOI: 10.1016/j.jconrel.2014.05.014] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 05/02/2014] [Accepted: 05/07/2014] [Indexed: 01/01/2023]
|
17
|
Zhu S, Chen P, Wu Y, Xiong S, Sun H, Xia Q, Shi L, Liu H, Ouyang HW. Programmed Application of Transforming Growth Factor β3 and Rac1 Inhibitor NSC23766 Committed Hyaline Cartilage Differentiation of Adipose-Derived Stem Cells for Osteochondral Defect Repair. Stem Cells Transl Med 2014; 3:1242-51. [PMID: 25154784 DOI: 10.5966/sctm.2014-0042] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Hyaline cartilage differentiation is always the challenge with application of stem cells for joint repair. Transforming growth factors (TGFs) and bone morphogenetic proteins can initiate cartilage differentiation but often lead to hypertrophy and calcification, related to abnormal Rac1 activity. In this study, we developed a strategy of programmed application of TGFβ3 and Rac1 inhibitor NSC23766 to commit the hyaline cartilage differentiation of adipose-derived stem cells (ADSCs) for joint cartilage repair. ADSCs were isolated and cultured in a micromass and pellet culture model to evaluate chondrogenic and hypertrophic differentiation. The function of Rac1 was investigated with constitutively active Rac1 mutant and dominant negative Rac1 mutant. The efficacy of ADSCs with programmed application of TGFβ3 and Rac1 inhibitor for cartilage repair was studied in a rat model of osteochondral defects. The results showed that TGFβ3 promoted ADSCs chondro-lineage differentiation and that NSC23766 prevented ADSC-derived chondrocytes from hypertrophy in vitro. The combination of ADSCs, TGFβ3, and NSC23766 promoted quality osteochondral defect repair in rats with much less chondrocytes hypertrophy and significantly higher International Cartilage Repair Society macroscopic and microscopic scores. The findings have illustrated that programmed application of TGFβ3 and Rac1 inhibitor NSC23766 can commit ADSCs to chondro-lineage differentiation and improve the efficacy of ADSCs for cartilage defect repair. These findings suggest a promising stem cell-based strategy for articular cartilage repair.
Collapse
Affiliation(s)
- Shouan Zhu
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Pengfei Chen
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Yan Wu
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Si Xiong
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Heng Sun
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Qingqing Xia
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Libing Shi
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Huanhuan Liu
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| | - Hong Wei Ouyang
- Department of Sports Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China; Zhejiang Provincial Key Laboratory for Tissue Engineering and Regenerative Medicine, School of Medicine, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
18
|
Sun Q, Fan J, Han D, Zhang J, Jiang B, Li X, Li X, Song Y. Evaluation of toxicity and adjuvant effects of peptidoglycan microspheres orally administered to mice. J Microencapsul 2014; 32:46-53. [PMID: 25090591 DOI: 10.3109/02652048.2014.944948] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In this study, peptidoglycan microspheres were evaluated for their toxicity and adjuvant effects after oral administration to mice. The liver and spleen indexes, CD cell content in peripheral blood and spleen, and immunoglobulin content in peripheral blood were measured by flow cytometry and indirect ELISA, respectively. Peptidoglycan microspheres with a loading capacity of 46.41 ± 0.83 g/100 g were prepared. In vivo tests showed that peptidoglycan microspheres revealed an immuno-enhancing profile as indicated by the slow increase of IgG content in peripheral blood compared with that of the untreated peptidoglycan group. In conclusion, peptidoglycan microspheres may be used as a new oral adjuvant in the host.
Collapse
Affiliation(s)
- Qingshen Sun
- University Key Laboratory of Microbiology, College of Life Science, Heilongjiang University , Harbin , P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Riese P, Sakthivel P, Trittel S, Guzmán CA. Intranasal formulations: promising strategy to deliver vaccines. Expert Opin Drug Deliv 2014; 11:1619-34. [PMID: 24962722 DOI: 10.1517/17425247.2014.931936] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The emergence of new diseases and the lack of efficient vaccines against numerous non-treatable pathogens require the development of novel vaccination strategies. To date, only a few mucosal vaccines have been approved for humans. This was in part due to i) the use of live attenuated vaccines, which are not suitable for certain groups of individuals, ii) safety concerns derived from implementation in humans of some mucosal vaccines, iii) the poor stability, absorption and immunogenicity of antigens delivered by the mucosal route and iv) the limited number of available technologies to overcome the bottlenecks associated with mucosal antigen delivery. Recent advances make feasible the development of efficacious mucosal vaccines with adequate safety profile. Thus, currently intranasal vaccines represent an attractive and valid alternative to conventional vaccines. AREAS COVERED The present review is focused on the potentials and limitations of market-approved intranasal vaccines and promising candidates undergoing clinical investigations. Furthermore, emerging strategies to overcome main bottlenecks including efficient breaching of the mucosal barrier and safety concerns by implementation of new adjuvants and delivery systems are discussed. EXPERT OPINION The rational design of intranasal vaccines requires an in-depth understanding of the anatomic, physicochemical and barrier properties of the nasal mucosa, as well as the molecular mechanisms governing the activation of the local innate and adaptive immune system. This would provide the critical knowledge to establish effective approaches to deliver vaccine antigens across the mucosal barrier, supporting the stimulation of a long-lasting protective response at both mucosal and systemic levels. Current developments in the area of adjuvants, nanotechnologies and mucosal immunology, together with the identification of surface receptors that can be exploited for cell targeting and manipulating their physiological properties, will become instrumental for developing a new generation of more effective intranasal vaccines.
Collapse
Affiliation(s)
- Peggy Riese
- Helmholtz Centre for Infection Research, Department of Vaccinology and Applied Microbiology , Inhoffenstrasse 7, 38124 Braunschweig , Germany
| | | | | | | |
Collapse
|
20
|
Preparation and Optimization of a Live Newcastle Disease Virus Vaccine Encapsulated in Chitosan Nanoparticles. ACTA ACUST UNITED AC 2013. [DOI: 10.4028/www.scientific.net/amr.662.149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study, the researcher’s interest is focused on establishing a model with La Sota live vaccine immobilized into chitosan, which was prepared using a ionic gelation method. The formulation, preparation procedure, influence factors, physicochemical characteristics were evaluated. The results of study demonstrate that the NDV-CS-NPs have been produced with suitable size, morphous regulation, extremely spherical shape, regular and well-distributed. The NDV-CS-NPs produced by the optimal formulation were average size (371.1nm) and proper zata potential (+2.84 mV). The entrapment efficiency was (74±3.7) %. It can provide a new useful information for the development and evaluation of synthetic vaccines.
Collapse
|
21
|
|
22
|
Islam MA, Firdous J, Choi YJ, Yun CH, Cho CS. Design and application of chitosan microspheres as oral and nasal vaccine carriers: an updated review. Int J Nanomedicine 2012; 7:6077-93. [PMID: 23271909 PMCID: PMC3526152 DOI: 10.2147/ijn.s38330] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Chitosan, a natural biodegradable polymer, is of great interest in biomedical research due to its excellent properties including bioavailability, nontoxicity, high charge density, and mucoadhesivity, which creates immense potential for various pharmaceutical applications. It has gelling properties when it interacts with counterions such as sulfates or polyphosphates and when it crosslinks with glutaraldehyde. This characteristic facilitates its usefulness in the coating or entrapment of biochemicals, drugs, antigenic molecules as a vaccine candidate, and microorganisms. Therefore, chitosan together with the advance of nanotechnology can be effectively applied as a carrier system for vaccine delivery. In fact, chitosan microspheres have been studied as a promising carrier system for mucosal vaccination, especially via the oral and nasal route to induce enhanced immune responses. Moreover, the thiolated form of chitosan is of considerable interest due to its improved mucoadhesivity, permeability, stability, and controlled/extended release profile. This review describes the various methods used to design and synthesize chitosan microspheres and recent updates on their potential applications for oral and nasal delivery of vaccines. The potential use of thiolated chitosan microspheres as next-generation mucosal vaccine carriers is also discussed.
Collapse
Affiliation(s)
- Mohammad Ariful Islam
- Department of Agricultural Biotechnology, Seoul National University, Seoul, South Korea
| | | | | | | | | |
Collapse
|
23
|
Kammona O, Kiparissides C. Recent advances in nanocarrier-based mucosal delivery of biomolecules. J Control Release 2012; 161:781-94. [PMID: 22659331 DOI: 10.1016/j.jconrel.2012.05.040] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 05/21/2012] [Accepted: 05/22/2012] [Indexed: 01/20/2023]
Abstract
This review highlights the recent developments in the area of nanocarrier-based mucosal delivery of therapeutic biomolecules and antigens. Macromolecular drugs have the unique power to tackle challenging diseases but their structure, physicochemical properties, stability, pharmacodynamics, and pharmacokinetics place stringent demands on the way they are delivered into the body (e.g., inability to cross mucosal surfaces and biological membranes). Carrier-based drug delivery systems can diminish the toxicity of therapeutic biomolecules, improve their bioavailability and make possible their administration via less-invasive routes (e.g., oral, nasal, pulmonary, etc.). Thus, the development of functionalized nanocarriers and nanoparticle-based microcarriers for the delivery of macromolecular drugs is considered an important scientific challenge and at the same time a business breakthrough for the biopharmaceutical industry. In order to be translated to the clinic the nanocarriers need to be biocompatible, biodegradable, stable in biological media, non-toxic and non-immunogenic, to exhibit mucoadhesive properties, to cross mucosal barriers and to protect their sensitive payload and deliver it to its target site in a controlled manner, thus increasing significantly its bioavailability and efficacy.
Collapse
Affiliation(s)
- Olga Kammona
- Chemical Process Engineering Research Institute, Centre for Research and Technology Hellas, P.O. Box 60361, 57001 Thessaloniki, Greece
| | | |
Collapse
|
24
|
Chua BY, Al Kobaisi M, Zeng W, Mainwaring D, Jackson DC. Chitosan Microparticles and Nanoparticles as Biocompatible Delivery Vehicles for Peptide and Protein-Based Immunocontraceptive Vaccines. Mol Pharm 2011; 9:81-90. [DOI: 10.1021/mp200264m] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Brendon Y. Chua
- Department of Microbiology and
Immunology, The University of Melbourne, Royal Parade, Parkville, Australia 3010
| | - Mohammad Al Kobaisi
- School of Applied Sciences, Royal Melbourne Institute of Technology, Melbourne,
Australia 3010
| | - Weiguang Zeng
- Department of Microbiology and
Immunology, The University of Melbourne, Royal Parade, Parkville, Australia 3010
| | - David Mainwaring
- School of Applied Sciences, Royal Melbourne Institute of Technology, Melbourne,
Australia 3010
| | - David C. Jackson
- Department of Microbiology and
Immunology, The University of Melbourne, Royal Parade, Parkville, Australia 3010
| |
Collapse
|
25
|
Bueter CL, Lee CK, Rathinam VAK, Healy GJ, Taron CH, Specht CA, Levitz SM. Chitosan but not chitin activates the inflammasome by a mechanism dependent upon phagocytosis. J Biol Chem 2011; 286:35447-35455. [PMID: 21862582 DOI: 10.1074/jbc.m111.274936] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chitin is an abundant polysaccharide found in fungal cell walls, crustacean shells, and insect exoskeletons. The immunological properties of both chitin and its deacetylated derivative chitosan are of relevance because of frequent natural exposure and their use in medical applications. Depending on the preparation studied and the end point measured, these compounds have been reported to induce allergic responses, inflammatory responses, or no response at all. We prepared highly purified chitosan and chitin and examined the capacity of these glycans to stimulate murine macrophages to release the inflammasome-associated cytokine IL-1β. We found that although chitosan was a potent NLRP3 inflammasome activator, acetylation of the chitosan to chitin resulted in a near total loss of activity. The size of the chitosan particles played an important role, with small particles eliciting the greatest activity. An inverse relationship between size and stimulatory activity was demonstrated using chitosan passed through size exclusion filters as well as with chitosan-coated beads of defined size. Partial digestion of chitosan with pepsin resulted in a larger fraction of small phagocytosable particles and more potent inflammasome activity. Inhibition of phagocytosis with cytochalasin D abolished the IL-1β stimulatory activity of chitosan, offering an explanation for why the largest particles were nearly devoid of activity. Thus, the deacetylated polysaccharide chitosan potently activates the NLRP3 inflammasome in a phagocytosis-dependent manner. In contrast, chitin is relatively inert.
Collapse
Affiliation(s)
- Chelsea L Bueter
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Chrono K Lee
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Vijay A K Rathinam
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | | | | | - Charles A Specht
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Stuart M Levitz
- Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts 01605.
| |
Collapse
|
26
|
Abstract
The current vaccine market is gaining momentum in the development of alternative administration routes namely intranasal, oral, topical, pulmonary, vaginal, and rectal; the nasal route offers the most promising opportunity for vaccine administration. It can enhance convenience, safety, elicit both local and systemic immune responses; thus potentially provide protection from pathogens at the site of entry. Nasal vaccine innovation comes with both opportunities and challenges. The innovative strategies used by industry and researchers to overcome the hurdles are discussed in this article: these include live-attenuated vaccines, adjuvants, mucoadhesives, particulate delivery systems, virus-like particles, vaccine manufacture, challenges of regulatory authorities, and the nasal vaccine impact on market potential. Critical issues for effective nasal vaccination are the antigen-retention period that enables its interaction with the lymphatic system and choice of an adjuvant that is nontoxic and induces the required immune response. Co-adjuvanting by means of a mucoadhesive technology addresses some of these issues. ChiSys(®), a natural bioadhesive with proven intranasal safety profile, has already demonstrated efficacy for several nasally delivered vaccines including norovirus. With the looming threat of a pandemic, alternatives such as intranasal vaccination will ultimately facilitate greater public compliance and rapid mass global vaccination.
Collapse
Affiliation(s)
- Inderjit Jabbal-Gill
- Archimedes Development Ltd., Albert Einstein Centre, Nottingham Science & Technology Park, University Boulevard, Nottingham, UK.
| |
Collapse
|
27
|
Şenel S. Chitosan-Based Particulate Systems for Non-Invasive Vaccine Delivery. ADVANCES IN POLYMER SCIENCE 2011. [DOI: 10.1007/12_2011_120] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
28
|
Günbeyaz M, Faraji A, Özkul A, Puralı N, Şenel S. Chitosan based delivery systems for mucosal immunization against bovine herpesvirus 1 (BHV-1). Eur J Pharm Sci 2010; 41:531-45. [DOI: 10.1016/j.ejps.2010.08.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Revised: 07/20/2010] [Accepted: 08/19/2010] [Indexed: 12/27/2022]
|
29
|
Yoo MK, Kang SK, Choi JH, Park IK, Na HS, Lee HC, Kim EB, Lee NK, Nah JW, Choi YJ, Cho CS. Targeted delivery of chitosan nanoparticles to Peyer’s patch using M cell-homing peptide selected by phage display technique. Biomaterials 2010; 31:7738-47. [DOI: 10.1016/j.biomaterials.2010.06.059] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Accepted: 06/28/2010] [Indexed: 11/29/2022]
|
30
|
Canali MM, Porporatto C, Aoki MP, Bianco ID, Correa SG. Signals elicited at the intestinal epithelium upon chitosan feeding contribute to immunomodulatory activity and biocompatibility of the polysaccharide. Vaccine 2010; 28:5718-24. [PMID: 20598784 DOI: 10.1016/j.vaccine.2010.06.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2009] [Revised: 06/01/2010] [Accepted: 06/05/2010] [Indexed: 10/19/2022]
Abstract
Chitosan is a copolymer of N-acetylglucosamine and glucosamine derived from chitin with several applications in pharmaceutical and medical fields. This polysaccharide exhibits adjuvant properties in mucosal immune responses of humans, rats and mice. Characterization of signals elicited by chitosan at the intestinal epithelium could explain its immunomodulatory activity and biocompatibility. We fed normal rats with single doses of chitosan and 16h later, we purified intestinal epithelial cells (IECs) to assess immune and biochemical parameters. Following chitosan administration, mRNA expression and release of several cytokines and chemokines increased, injury markers maintained constitutive levels and MHC type II molecule expression was augmented. IEC supernatants showed higher levels of IL-10, IL-6 and TGF-beta. Arginase activity of IECs increased upon chitosan interaction in vivo and in vitro. Together, after chitosan feeding, mild activation of IECs occurs in vivo, with production of regulatory factors that could be relevant for its biocompatibility and immunomodulatory effects.
Collapse
Affiliation(s)
- M Magdalena Canali
- Immunology, Center of Research in Biochemistry and Immunology (CIBICI) (National Council of Research in Science and Technology (CONICET), Department of Clinical Biochemistry, Faculty of Chemical Sciences, National University of Cordoba, Cordoba, Argentina
| | | | | | | | | |
Collapse
|
31
|
|
32
|
Kang ML, Cho CS, Yoo HS. Application of chitosan microspheres for nasal delivery of vaccines. Biotechnol Adv 2009; 27:857-865. [DOI: 10.1016/j.biotechadv.2009.06.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Revised: 06/24/2009] [Accepted: 06/27/2009] [Indexed: 12/01/2022]
|
33
|
Arca HC, Günbeyaz M, Senel S. Chitosan-based systems for the delivery of vaccine antigens. Expert Rev Vaccines 2009; 8:937-53. [PMID: 19538118 DOI: 10.1586/erv.09.47] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This review discusses the current status of chitosan and its derivatives as adjuvants and delivery systems in vaccine delivery, and their future possibilities and challenges. After a brief introduction to adjuvants and delivery systems, chitosan will be described in detail in regard to vaccine formulation. Applications of chitosan and its derivatives will be reviewed and their proposed mechanisms in the enhancement of immune responses will be discussed.
Collapse
Affiliation(s)
- H Ciğdem Arca
- Hacettepe University, Faculty of Pharmacy, Department of Pharmaceutical Technology, Ankara, Turkey.
| | | | | |
Collapse
|
34
|
Brandhonneur N, Loizel C, Chevanne F, Wakeley P, Jestin A, Le Potier MF, Le Corre P. Mucosal or systemic administration of rE2 glycoprotein antigen loaded PLGA microspheres. Int J Pharm 2009; 373:16-23. [PMID: 19429284 DOI: 10.1016/j.ijpharm.2009.01.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Revised: 01/23/2009] [Accepted: 01/24/2009] [Indexed: 10/21/2022]
Abstract
We have evaluated the ability of recombinant E2 antigen, as a surfactant free formulation of poly (D,L-lactide-co-glycolide) (PLGA) microspheres, to elicit a systemic immune response after administration by mucosal routes (oral and nasal) in comparison to intramuscular route. The sequence encoding a truncated E2 glycoprotein of the classical swine fever virus (CSFV) was expressed in insect cells following infection with recombinant baculovirus, as a His-tagged recombinant antigen. The recombinant E2 glycoprotein (rE2) antigen was co-encapsulated with rabbit serum albumin (RSA) as a protein stabilizer. rE2/RSA loaded PLGA microspheres, with a mean diameter of 4 microm were obtained by a water in oil in water solvent extraction method (w/o/w). Rabbits were immunized with 10 microg of rE2 formulated in PLGA microspheres administrated by three different routes (oral, nasal and intramuscular). After 60 days, each rabbit in all three groups was challenge with 5 microg of rE2 glycoprotein solution by intradermal administration. Blood samples were collected weekly for 90 days and specific rE2 antigen antibodies measured. This work showed that rE2 antigen loaded microspheres was able to initiate an immune response. The intradermal challenge after nasal and oral administration had a clear boost effect on the systemic immune response. Moreover, the response after nasal administration was more intense and less variable than oral route. In conclusion, these data demonstrate a high potential of rE2 loaded PLGA microspheres for their use as a mucosal subunit vaccine.
Collapse
Affiliation(s)
- N Brandhonneur
- Laboratoire de Pharmacie Galénique, Biopharmacie et Pharmacie Clinique, UPRES EA 3892, 2 avenue du professeur Léon Bernard 35043 Rennes Cedex, France
| | | | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Borges O, Silva M, de Sousa A, Borchard G, Junginger HE, Cordeiro-da-Silva A. Alginate coated chitosan nanoparticles are an effective subcutaneous adjuvant for hepatitis B surface antigen. Int Immunopharmacol 2008; 8:1773-80. [PMID: 18801462 DOI: 10.1016/j.intimp.2008.08.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Revised: 06/10/2008] [Accepted: 08/19/2008] [Indexed: 01/20/2023]
Abstract
We recently described a delivery system that is composed of a chitosan core to which the hepatitis B surface antigen (HBsAg) was adsorbed and subsequently coated with sodium alginate. In this present work, alginate coated chitosan nanoparticles were evaluated as a subcutaneous adjuvant for HBsAg. HBsAg loaded, alginate coated or uncoated chitosan nanoparticles, associated or not with CpGODN were subcutaneously administered to mice and several immunological parameters were evaluated. A high anti-HBsAg IgG titer (2271+/-120 mIU/ml), with the majority of antibodies being of Th2 type, was observed within group I, vaccinated with HBsAg loaded onto coated nanoparticles. However, regarding cellular immune response, no significant differences were observed for antigen-specific splenocyte proliferation or for the secretion of IFN-gamma and IL-4, when compared to the control group. The co-delivery of antigen-loaded nanoparticles in the presence of the immunopotentiator, CpG ODN 1826, resulted in an increase of anti-HBsAg IgG titers that was not statistically different from the first group; however, an increase of the IgG2a/IgG1 ratio from 0.1 to 1.0 and an increase (p<0.01) of the IFN-gamma production by the splenocytes stimulated with the HBV antigen was observed. The enhancement of the immune response observed with the antigen-loaded nanoparticles demonstrated that chitosan is a promising platform for parenteral HBsAg delivery and, when co-administered with the CpG ODN, resulted in a mixed Th1/Th2 type immune response.
Collapse
Affiliation(s)
- Olga Borges
- Center for Pharmaceutical Studies, Faculty of Pharmacy, University of Coimbra, 3000-295 Coimbra, Portugal.
| | | | | | | | | | | |
Collapse
|
37
|
Immune response by nasal delivery of hepatitis B surface antigen and codelivery of a CpG ODN in alginate coated chitosan nanoparticles. Eur J Pharm Biopharm 2008; 69:405-16. [PMID: 18364251 DOI: 10.1016/j.ejpb.2008.01.019] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2007] [Revised: 12/28/2007] [Accepted: 01/15/2008] [Indexed: 11/24/2022]
Abstract
Alginate coated chitosan nanoparticles were previously developed with the aim of protecting the antigen, adsorbed on the surface of those chitosan nanoparticles, from enzymatic degradation at mucosal surfaces. In this work, this new delivery system was loaded with the recombinant hepatitis B surface antigen (HBsAg) and applied to mice by the intranasal route. Adjuvant effect of the delivery system was studied by measuring anti-HBsAg IgG in serum, anti-HBsAg sIgA in faeces extracts or nasal and vaginal secretions and interferon-gamma production in supernatants of the spleen cells. The mice were primed with 10 microg of the vaccine associated or not with nanoparticles and associated or not with 10 microg CpG oligodeoxynucleotide (ODN) followed by two sequential boosts at three week intervals. The association of HBsAg with the alginate coated chitosan nanoparticles, administered intranasally to the mice, gave rise to the humoral mucosal immune response. Humoral systemic immune response was not induced by the HBsAg loaded nanoparticles alone. The generation of Th1-biased antigen-specific systemic antibodies, however, was observed when HBsAg loaded nanoparticles were applied together with a second adjuvant, the immunopotentiator, CpG ODN. Moreover, all intranasally vaccinated groups showed higher interferon-gamma production when compared to naïve mice.
Collapse
|
38
|
Jiang HL, Kang ML, Quan JS, Kang SG, Akaike T, Yoo HS, Cho CS. The potential of mannosylated chitosan microspheres to target macrophage mannose receptors in an adjuvant-delivery system for intranasal immunization. Biomaterials 2008; 29:1931-9. [PMID: 18221992 DOI: 10.1016/j.biomaterials.2007.12.025] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Accepted: 12/15/2007] [Indexed: 12/27/2022]
Abstract
A vaccine delivery system based on mannosylated chitosan microspheres (MCMs) was studied in vitro and in vivo. Bordetella bronchiseptica antigens containing dermonecrotoxin (BBD) were loaded in MCMs or chitosan microspheres (CMs). Fluorescence confocal microscopy indicated that BBD-loaded MCMs (BBD-MCMs) bound with mannose receptors on murine macrophages (RAW264.7 cells). In vitro experiments using macrophages demonstrated that BBD-MCMs had more effective immune-stimulating activity than BBD-loaded CMs (BBD-CMs). Mice intranasally immunized with BBD-MCMs showed significantly higher BBD-specific IgA antibody responses in saliva and serum than mice immunized with BBD-CMs (p<0.05). After challenge with B. bronchiseptica via the nasal cavity, groups treated with BBD-MCMs or BBD-CMs showed similar patterns with a high survival rate even though there was no significant difference between those groups. These results suggested that mannose moieties in the MCMs enhanced immune-stimulating activities through mucosal delivery due to a specific interaction between mannose groups in the MCMs and mannose receptors on the macrophages.
Collapse
Affiliation(s)
- Hu-Lin Jiang
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
39
|
|
40
|
Kang ML, Jiang HL, Kang SG, Guo DD, Lee DY, Cho CS, Yoo HS. Pluronic® F127 enhances the effect as an adjuvant of chitosan microspheres in the intranasal delivery of Bordetella bronchiseptica antigens containing dermonecrotoxin. Vaccine 2007; 25:4602-10. [PMID: 17485148 DOI: 10.1016/j.vaccine.2007.03.038] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Revised: 02/23/2007] [Accepted: 03/21/2007] [Indexed: 11/30/2022]
Abstract
We have studied a vaccine delivery system in vitro and in vivo based on chitosan microspheres (CMs) prepared in the presence of selected immunomodulators, Pluronic block copolymer F127 (F127). The Bordetella bronchiseptica multiple antigens containing dermonecrotoxin (BBD), a virulent factor leading to atrophic rhinitis (AR) in swine was loaded in CMs/F127 or CMs alone. The microspheres, prepared using an ionic gelation process with tripolyphosphate, demonstrated release profiles that showed a greater amount of BBD being released from BBD-loaded CMs/F127 (BBD-CMs/F127). In vitro experiments using mouse alveolar macrophage cells (RAW 264.7) demonstrated that BBD-CMs/F127 have significantly higher immune-stimulating activities than controls. The highest immune-stimulating activities by the BBD-CMs/F127 using RAW 264.7 cells were mirrored in the in vivo studies following nasal administration to mice. The mice immunized with BBD-CMs/F127 showed higher BBD specific IgA antibody responses in nasal wash, saliva and serum than mice immunized with BBD-CMs alone. Protective immunity was measured by survival rate after challenge with B. bronchiseptica via the nasal cavity. The survival rate of the group treated with BBD-CMs/F127 was higher than those of other groups. These results suggested that CMs/F127 represents a novel mucosal delivery system and that F127 could enhance the delivery of BBD-CMs in the vaccination scheme.
Collapse
Affiliation(s)
- Mi Lan Kang
- Department of Infectious Disease, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Chitosan is a partially deacetylated polymer obtained from the alkaline deacetylation of chitin, which is a glucose-based, unbranched polysaccharide that occurs widely in nature as the principal component of exoskeletons of crustaceans and insects, as well as of the cell walls of some bacteria and fungi. Chitosan exhibits a variety of physicochemical and biological properties resulting in numerous applications in fields such as waste water treatment, agriculture, fabric and textiles, cosmetics, nutritional enhancement and food processing. In addition to its lack of toxicity and allergenicity, its biocompatibility, biodegradability and bioactivity make it a very attractive substance for diverse applications as a biomaterial in the pharmaceutical and medical fields. This review takes a closer look at the biomedical applications of chitosan microspheres. Based on recent research and existing products, some new and potential future approaches in this fascinating area are discussed.
Collapse
Affiliation(s)
- Jaleh Varshosaz
- Department of Pharmaceutics, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
42
|
Xie Y, Zhou NJ, Gong YF, Zhou XJ, Chen J, Hu SJ, Lu NH, Hou XH. Th immune response induced by H pylori vaccine with chitosan as adjuvant and its relation to immune protection. World J Gastroenterol 2007; 13:1547-1553. [PMID: 17461447 PMCID: PMC4146897 DOI: 10.3748/wjg.v13.i10.1547] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2006] [Revised: 12/28/2006] [Accepted: 02/27/2007] [Indexed: 02/06/2023] Open
Abstract
AIM To study the immunological protective effect of H pylori vaccine with chitosan as an adjuvant and its mechanism. METHODS Female BALB/c mice were randomly divided into seven groups and orally immunized respectively with PBS, chitosan solution, chitosan particles, H pylori antigen, H pylori antigen plus cholera toxin (CT), H pylori antigen plus chitosan solution, H pylori antigen plus chitosan particles once a week for four weeks. Four weeks after the last immunization, the mice were challenged twice by alive H pylori (1 x 10(9) CFU/mL) and sacrificed. Part of the gastric mucosa was embedded in paraffin, cut into sections and assayed with Giemsa staining. Part of the gastric mucosa was used to quantitatively culture H pylori. ELISA was used to detect cytokine level in gastric mucosa and anti- H pylori IgG1, IgG2a levels in serum. RESULTS In the groups with chitosan as an adjuvant, immunological protection was achieved in 60% mice, which was significantly higher than in groups with H pylori antigen alone and without H pylori antigen (P < 0.05 or 0.001). Before challenge, the level of IFN and IL-12 in gastric mucosa was significantly higher in the groups with chitosan as an adjuvant than in the control group and the group without adjuvant (P < 0.05 or 0.005). After challenge, the level of IFN and IL-12 was significantly higher in the groups with adjuvant than in the groups without adjuvant and antigen (P < 0.05 or 0.001). Before challenge, the level of IL-2 in gastric mucosa was not different among different groups. After challenge the level of IL-2 was significantly higher in the groups with adjuvant than in the control group (P < 0.05 or 0.001). Before challenge, the level of IL-10 in gastric mucosa was significantly higher in the groups with chitosan as an adjuvant than in other groups without adjuvant (P < 0.05 or 0.01). After challenge, the level of IL-10 was not different among different groups. Before challenge, the level of IL-4 in gastric mucosa was significantly higher in the groups with chitosan as an adjuvant than in other groups without adjuvant (P < 0.05). After challenge, the level of IL-4 was significantly higher in the groups with chitosan particles as an adjuvant than in the group with CT as an adjuvant (P < 0.05), and in the group with chitosan solution as an adjuvant, the level of IL-4 was significantly higher than that in control group, non-adjuvant group and the groups with CT (P < 0.05 or 0.001). The ratio of anti- H pylori IgG2a/IgG1 in serum was significantly lower in the groups with chitosan as an adjuvant than in the groups with CT as an adjuvant or without adjuvant (P < 0.01). CONCLUSION H pylori vaccine with chitosan as an adjuvant can protect against H pylori infection and induce both Th1 and Th2 type immune response.
Collapse
Affiliation(s)
- Yong Xie
- Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Xiang SD, Scholzen A, Minigo G, David C, Apostolopoulos V, Mottram PL, Plebanski M. Pathogen recognition and development of particulate vaccines: does size matter? Methods 2007; 40:1-9. [PMID: 16997708 DOI: 10.1016/j.ymeth.2006.05.016] [Citation(s) in RCA: 459] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2006] [Accepted: 05/05/2006] [Indexed: 01/08/2023] Open
Abstract
The use of particulate carriers holds great promise for the development of effective and affordable recombinant vaccines. Rational development requires a detailed understanding of particle up-take and processing mechanisms to target cellular pathways capable of stimulating the required immune responses safely. These mechanisms are in turn based on how the host has evolved to recognize and process pathogens. Pathogens, as well as particulate vaccines, come in a wide range of sizes and biochemical compositions. Some of these also provide 'danger signals' so that antigen 'senting cells (APC), usually dendritic cells (DC), acquire specific stimulatory activity. Herein, we provide an overview of the types of particles currently under investigation for the formulation of vaccines, discuss cellular uptake mechanisms (endocytosis, macropinocytosis, phagocytosis, clathrin-dependent and/or caveloae-mediated) for pathogens and particles of different sizes, as well as antigen possessing and presentation by APC in general, and DC in particular. Since particle size and composition can influence the immune response, inducing humoral and/or cellular immunity, activating CD8 T cells and/or CD4 T cells of T helper 1 and/or T helper 2 type, particle characteristics have a major impact on vaccine efficacy. Recently developed methods for the formulation of particulate vaccines are presented in this issue of Methods, showcasing a range of "cutting edge" particulate vaccines that employ particles ranging from nano to micro-sized. This special issue of Methods further addresses practical issues of production, affordability, reproducibility and stability of formulation, and also includes a discussion of the economic and regulatory challenges encountered in developing vaccines for veterinary use and for common Third World infectious diseases.
Collapse
Affiliation(s)
- Sue D Xiang
- Vaccine and Infectious Diseases Laboratory, Burnet Institute at Austin, Studley Road, Heidelberg, Vic. 3084, Australia
| | | | | | | | | | | | | |
Collapse
|