51
|
Cohen E, Merzendorfer H. Chitin/Chitosan: Versatile Ecological, Industrial, and Biomedical Applications. EXTRACELLULAR SUGAR-BASED BIOPOLYMERS MATRICES 2019; 12. [PMCID: PMC7115017 DOI: 10.1007/978-3-030-12919-4_14] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chitin is a linear polysaccharide of N-acetylglucosamine, which is highly abundant in nature and mainly produced by marine crustaceans. Chitosan is obtained by hydrolytic deacetylation. Both polysaccharides are renewable resources, simply and cost-effectively extracted from waste material of fish industry, mainly crab and shrimp shells. Research over the past five decades has revealed that chitosan, in particular, possesses unique and useful characteristics such as chemical versatility, polyelectrolyte properties, gel- and film-forming ability, high adsorption capacity, antimicrobial and antioxidative properties, low toxicity, and biocompatibility and biodegradability features. A plethora of chemical chitosan derivatives have been synthesized yielding improved materials with suggested or effective applications in water treatment, biosensor engineering, agriculture, food processing and storage, textile additives, cosmetics fabrication, and in veterinary and human medicine. The number of studies in this research field has exploded particularly during the last two decades. Here, we review recent advances in utilizing chitosan and chitosan derivatives in different technical, agricultural, and biomedical fields.
Collapse
Affiliation(s)
- Ephraim Cohen
- Department of Entomology, The Robert H. Smith Faculty of Agriculture Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Hans Merzendorfer
- School of Science and Technology, Institute of Biology – Molecular Biology, University of Siegen, Siegen, Germany
| |
Collapse
|
52
|
Long S, Xu Y, Zhou F, Wang B, Yang Y, Fu Y, Du N, Li X. Characteristics of temperature changes in photothermal therapy induced by combined application of indocyanine green and laser. Oncol Lett 2019; 17:3952-3959. [PMID: 30930992 DOI: 10.3892/ol.2019.10058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Accepted: 12/19/2018] [Indexed: 01/27/2023] Open
Abstract
Photothermal therapy, a type of laser application, has the ability to eradicate tumor cells by a local thermal effect and elicit a tumor specific immune response. Indocyanine green (ICG), a photosensitizer, can effectively elevate the local temperature by absorbing energy from the laser. The present study aimed to investigate the characteristics of temperature changes during photothermal therapy with an infrared thermometer in an ICG solution and in tumor-bearing mice treated with a combination of laser and ICG. Additionally, the present study observed the morphological changes of tumor tissue by hematoxylin-eosin staining following photothermal therapy. In the solution experiment, when the laser power density was 1 W/cm2 and the concentration of ICG was 0 or 0.0187 mg/ml, the temperature of the water was elevated by 3 and 28°C, respectively. In the tumor-bearing mice experiment, when the laser power density was 1 W/cm2 and the concentration of ICG was 0 and 0.1 mg/ml, the temperature of the tumor-bearing mice was elevated by 6.9 and 28.5°C, respectively. With an increase in laser power density, including 0.6, 0.8 and 1.0 W/cm2, the temperature was 23.3, 26.7 and 28.5°C, respectively. Pathological tissue sections demonstrated that a large number of tumor cells experienced necrosis, and the envelope of the tumor was destroyed. Numerous inflammatory cells, in particular lymphocytes, infiltrated into the tumor tissue following tumor tissue treatment with a combination of laser and ICG. These results indicated that a combination treatment with laser and ICG may significantly increase the temperature of the water solutions and in the tumor-bearing mice. The concentration of ICG and laser power density contributed to the temperature elevation, in particular to the concentration of ICG.
Collapse
Affiliation(s)
- Shan Long
- Department of Oncology, The Fourth Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100048, P.R. China
| | - Yuanyuan Xu
- Department of Oncology, The Fourth Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100048, P.R. China
| | - Feifan Zhou
- Biophotonics Research Laboratory Center for Interdisciplinary Biomedical Education and Research University of Central Oklahoma, Edmond, OK 73034, USA
| | - Bo Wang
- Department of Oncology, The Fourth Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100048, P.R. China
| | - Yunning Yang
- Department of Oncology, The Fourth Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100048, P.R. China
| | - Yan Fu
- Department of Oncology, The Fourth Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100048, P.R. China
| | - Nan Du
- Department of Oncology, The Fourth Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100048, P.R. China
| | - Xiaosong Li
- Department of Oncology, The Fourth Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100048, P.R. China
| |
Collapse
|
53
|
Lei Y, Zhao F, Shao J, Li Y, Li S, Chang H, Zhang Y. Application of built-in adjuvants for epitope-based vaccines. PeerJ 2019; 6:e6185. [PMID: 30656066 PMCID: PMC6336016 DOI: 10.7717/peerj.6185] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/29/2018] [Indexed: 12/21/2022] Open
Abstract
Several studies have shown that epitope vaccines exhibit substantial advantages over conventional vaccines. However, epitope vaccines are associated with limited immunity, which can be overcome by conjugating antigenic epitopes with built-in adjuvants (e.g., some carrier proteins or new biomaterials) with special properties, including immunologic specificity, good biosecurity and biocompatibility, and the ability to vastly improve the immune response of epitope vaccines. When designing epitope vaccines, the following types of built-in adjuvants are typically considered: (1) pattern recognition receptor ligands (i.e., toll-like receptors); (2) virus-like particle carrier platforms; (3) bacterial toxin proteins; and (4) novel potential delivery systems (e.g., self-assembled peptide nanoparticles, lipid core peptides, and polymeric or inorganic nanoparticles). This review primarily discusses the current and prospective applications of these built-in adjuvants (i.e., biological carriers) to provide some references for the future design of epitope-based vaccines.
Collapse
Affiliation(s)
- Yao Lei
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Furong Zhao
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Junjun Shao
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yangfan Li
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Shifang Li
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Huiyun Chang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Yongguang Zhang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
54
|
Chitosan and its derivatives: synthesis, biotechnological applications, and future challenges. Appl Microbiol Biotechnol 2019; 103:1557-1571. [DOI: 10.1007/s00253-018-9550-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 11/26/2018] [Accepted: 11/29/2018] [Indexed: 12/25/2022]
|
55
|
Anticancer Activity of Chitosan, Chitosan Derivatives, and Their Mechanism of Action. Int J Biomater 2018; 2018:2952085. [PMID: 30693034 PMCID: PMC6332982 DOI: 10.1155/2018/2952085] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 11/26/2018] [Accepted: 12/04/2018] [Indexed: 12/15/2022] Open
Abstract
Tailoring of chitosan through the involvement of its amino, acetamido, and hydroxy groups can give derivatives of enhanced solubility and remarkable anticancer activity. The general mechanism of such activity is associated with the disturbances in normal functioning of cell cycle, interference to the central dogma of biological system from DNA to RNA to protein or enzymatic synthesis, and the disruption of hormonal path to biosynthesis to inhibit the growth of cancer cells. Both chitosan and its various derivatives have been reported to selectively permeate through the cancer cell membranes and show anticancer activity through the cellular enzymatic, antiangiogenic, immunoenhancing, antioxidant defense mechanism, and apoptotic pathways. They get sequestered from noncancer cells and provide their enhanced bioavailability in cancer cells in a sustained release manner. This review presents the putative mechanisms of anticancer activity of chitosan and mechanistic approaches of structure activity relation upon the modification of chitosan through functionalization, complex formation, and graft copolymerization to give different derivatives.
Collapse
|
56
|
Liu J, Feng X, Chen Z, Yang X, Shen Z, Guo M, Deng F, Liu Y, Zhang H, Chen C. The adjuvant effect of C 60(OH) 22 nanoparticles promoting both humoral and cellular immune responses to HCV recombinant proteins. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 97:753-759. [PMID: 30678964 DOI: 10.1016/j.msec.2018.12.088] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 12/13/2018] [Accepted: 12/25/2018] [Indexed: 12/11/2022]
Abstract
Hepatitis c virus (HCV) infection is one of major causes for chronic liver diseases worldwide and could lead to death. Development of effective HCV vaccines is a powerful auxiliary method of existing treatments. Adjuvants are necessary for modern vaccines to promote immune responses. Among the various nanomaterials that have been developed, multihydroxylated fullerene (C60(OH)22) has been proved as an efficient adjuvant for human immunodeficiency virus DNA vaccine. Here, we utilized three types of HCV recombinant proteins as antigens to investigate the activity of C60(OH)22 as a protein vaccine adjuvant. The proteins were carried by C60(OH)22 in a way of surface adsorption and self-assemble encapsulation. C60(OH)22 at a relatively low dose was sufficient to promote both humoral and cellular immune responses to HCV protein antigens and reduce the usage of antigen. These results demonstrated the positive adjuvant properties of C60(OH)22 when applied to protein vaccines.
Collapse
Affiliation(s)
- Jing Liu
- The College of Life Sciences, Northwest University, Xi'an 710069, China; CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Xiaoyan Feng
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Zhiyun Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Xiqin Yang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China
| | - Ziyi Shen
- The College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Mengyu Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | | | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
| | - Heqiu Zhang
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing 100850, China.
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China.
| |
Collapse
|
57
|
Malik A, Gupta M, Gupta V, Gogoi H, Bhatnagar R. Novel application of trimethyl chitosan as an adjuvant in vaccine delivery. Int J Nanomedicine 2018; 13:7959-7970. [PMID: 30538470 PMCID: PMC6260144 DOI: 10.2147/ijn.s165876] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The application of natural carbohydrate polysaccharides for antigen delivery and its adjuvanation potential has garnered interest in the scientific community in the recent years. These biomaterials are considered favorable candidates for adjuvant development due to their desirable properties like enormous bioavailability, non-toxicity, biodegradability, stability, affordability, and immunostimulating ability. Chitosan is the one such extensively studied natural polymer which has been appreciated for its excellent applications in pharmaceuticals. Trimethyl chitosan (TMC), a derivative of chitosan, possesses these properties. In addition it has the properties of high aqueous solubility, high charge density, mucoadhesive, permeation enhancing (ability to cross tight junction), and stability over a range of ionic conditions which makes the spectrum of its applicability much broader. It has also been seen to perform analogously to alum, complete Freund’s adjuvant, incomplete Freund’s adjuvant, and cyclic guanosine monophosphate adjuvanation, which justifies its role as a potent adjuvant. Although many review articles detailing the applications of chitosan in vaccine delivery are available, a comprehensive review of the applications of TMC as an adjuvant is not available to date. This article provides a comprehensive overview of structural and chemical properties of TMC which affect its adjuvant characteristics; the efficacy of various delivery routes for TMC antigen combination; and the recent advances in the elucidation of its mechanism of action.
Collapse
Affiliation(s)
- Anshu Malik
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| | - Manish Gupta
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| | - Vatika Gupta
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| | - Himanshu Gogoi
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| | - Rakesh Bhatnagar
- Molecular Biology and Genetic Engineering Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India,
| |
Collapse
|
58
|
Immunomodulatory properties of chitosan polymers. Biomaterials 2018; 184:1-9. [DOI: 10.1016/j.biomaterials.2018.08.054] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/22/2018] [Accepted: 08/27/2018] [Indexed: 12/14/2022]
|
59
|
In vitro and in vivo anticancer efficacy potential of Quercetin loaded polymeric nanoparticles. Biomed Pharmacother 2018; 106:1513-1526. [DOI: 10.1016/j.biopha.2018.07.106] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/16/2018] [Accepted: 07/18/2018] [Indexed: 01/01/2023] Open
|
60
|
Pascual S, Planas A. Screening Assay for Directed Evolution of Chitin Deacetylases: Application to Vibrio cholerae Deacetylase Mutant Libraries for Engineered Specificity. Anal Chem 2018; 90:10654-10658. [PMID: 30134658 DOI: 10.1021/acs.analchem.8b02729] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Not only the degree of acetylation but also the pattern of acetylation of chitosans and chitooligosaccharides (COS) appear to be critical for their biological activities. Protein engineering may expand the toolbox of chitin deacetylases (CDAs) with defined specificities for the enzymatic production of partially deacetylated COS for biotech and biomedical applications. A high-throughput screening (HTS) assay for screening directed evolution libraries is reported. It is based on a fluorescence monitoring assay of the deacetylase activity on COS substrates after capturing the expressed enzyme variants fused to a chitin binding module with chitin-coated magnetic beads. The assay is applied to the screening of random libraries of a Vibrio cholera CDA for increased activity on longer COS substrates.
Collapse
Affiliation(s)
- Sergi Pascual
- Laboratory of Biochemistry , Institut Químic de Sarrià, University Ramon Llull , 08017 Barcelona , Spain
| | - Antoni Planas
- Laboratory of Biochemistry , Institut Químic de Sarrià, University Ramon Llull , 08017 Barcelona , Spain
| |
Collapse
|
61
|
Elieh Ali Komi D, Sharma L, Dela Cruz CS. Chitin and Its Effects on Inflammatory and Immune Responses. Clin Rev Allergy Immunol 2018; 54:213-223. [PMID: 28251581 DOI: 10.1007/s12016-017-8600-0] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Chitin, a potential allergy-promoting pathogen-associated molecular pattern (PAMP), is a linear polymer composed of N-acetylglucosamine residues which are linked by β-(1,4)-glycosidic bonds. Mammalians are potential hosts for chitin-containing protozoa, fungi, arthropods, and nematodes; however, mammalians themselves do not synthetize chitin and thus it is considered as a potential target for recognition by mammalian immune system. Chitin is sensed primarily in the lungs or gut where it activates a variety of innate (eosinophils, macrophages) and adaptive immune cells (IL-4/IL-13 expressing T helper type-2 lymphocytes). Chitin induces cytokine production, leukocyte recruitment, and alternative macrophage activation. Intranasal or intraperitoneal administration of chitin (varying in size, degree of acetylation and purity) to mice has been applied as a routine approach to investigate chitin's priming effects on innate and adaptive immunity. Structural chitin present in microorganisms is actively degraded by host true chitinases, including acidic mammalian chitinases and chitotriosidase into smaller fragments that can be sensed by mammalian receptors such as FIBCD1, NKR-P1, and RegIIIc. Immune recognition of chitin also involves pattern recognition receptors, mainly via TLR-2 and Dectin-1, to activate immune cells to induce cytokine production and creation of an immune network that results in inflammatory and allergic responses. In this review, we will focus on various immunological aspects of the interaction between chitin and host immune system such as sensing, interactions with immune cells, chitinases as chitin degrading enzymes, and immunologic applications of chitin.
Collapse
Affiliation(s)
- Daniel Elieh Ali Komi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, East Azerbayjan, Iran.,Department of Immunology, Tabriz University of Medical Sciences, Tabriz, East Azerbayjan, Iran
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Charles S Dela Cruz
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA. .,Department of Microbial Pathogenesis, Yale School of Medicine, Cedar Street, New Haven, CT, TACS441D, USA.
| |
Collapse
|
62
|
Nikapitiya C, Dananjaya SHS, De Silva BCJ, Heo GJ, Oh C, De Zoysa M, Lee J. Chitosan nanoparticles: A positive immune response modulator as display in zebrafish larvae against Aeromonas hydrophila infection. FISH & SHELLFISH IMMUNOLOGY 2018; 76:240-246. [PMID: 29510255 DOI: 10.1016/j.fsi.2018.03.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/26/2018] [Accepted: 03/02/2018] [Indexed: 06/08/2023]
Abstract
Chitosan nanoparticles (CNPs) were synthesized by ionic gelation method and its immunomodulatory properties were investigated in zebrafish larvae. Average particle size and zeta potential were 181.2 nm and +37.2 mv, respectively. Initially, toxicity profile was tested in zebrafish embryo at 96 h post fertilization (hpf) stage using medium molecular weight chitosan (MMW-C) and CNPs. At 5 μg/mL, the hatching rate was almost similar in both treatments, however, the survival rate was lower in MMW-C compared to CNPs exposure, suggesting that toxicity effect of CNPs in hatched larvae was minimal at 5 μg/mL compared to MMW-C. Quantitative real time PCR results showed that in CNPs exposed larvae at 5 days post fertilization (5 dpf) stage, immune related (il-1β, tnf-α, il-6, il-10, cxcl-18b, ccl34a.4, cxcl-8a, lyz-c, defβl-1, irf-1a, irf-3, MxA) and stress response (hsp-70) genes were induced. In contrast, basal or down regulated expression of antioxidant genes (gstp-1, cat, sod-1, prdx-4, txndr-1) were observed. Moreover, zebrafish larvae (at 5 dpf stage) exposed to CNPs (5 μg/mL) showed higher survival rate at 72 h post infection stage against pathogenic Aeromonas hydrophila challenge compared to controls. These results suggest that although CNPs can have toxic effects to the larvae at higher doses, CNPs exposure at 5 μg/mL could enhance the immune responses and develop the disease resistance against A. hydrophila, which could be attributed to its strong immune modulatory properties.
Collapse
Affiliation(s)
- Chamilani Nikapitiya
- Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - S H S Dananjaya
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - B C J De Silva
- Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Gang-Joon Heo
- Veterinary Medical Center and College of Veterinary Medicine, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Chulhong Oh
- Jeju International Marine Science Research and Education Center, Korea Institute of Ocean Science and Technology, Jeju Special Self-Governing Province, 63349, Republic of Korea
| | - Mahanama De Zoysa
- College of Veterinary Medicine and Research Institute of Veterinary Medicine, Chungnam National University, Yuseong-gu, Daejeon, 34134, Republic of Korea.
| | - Jehee Lee
- Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Department of Marine Life Sciences, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea.
| |
Collapse
|
63
|
Torabi N, Dobakhti F, Faghihzadeh S, Haniloo A. In vitro and in vivo effects of chitosan-praziquantel and chitosan-albendazole nanoparticles on Echinococcus granulosus Metacestodes. Parasitol Res 2018; 117:2015-2023. [PMID: 29616349 DOI: 10.1007/s00436-018-5849-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 03/22/2018] [Indexed: 12/01/2022]
Abstract
Cystic echinococcosis (CE), which is caused by the metacestode of Echinococcus granulosus, is one of the most important zoonoses affecting humans. Benzimidazoles (in particular albendazole) and praziquantel (PZQ) are effective against CE, but poor water solubility of these agents often leads to inadequate results. Here, we evaluate the effects of chitosan-albendazole (ChABZ) and chitosan-praziquantel (ChPZQ) nanoparticles as a new formulation on hydatid cysts both in vitro and in vivo. Developed microcysts in culture were treated with different concentrations of ChABZ and ChPZQ nanoparticles (either alone or in combination), and ABZ + PZQ suspension. The viability rate of microcysts was used to evaluate the drug efficacies. In addition, the prophylactic and therapeutic effects of the drugs were studied on infected DBA/2 mice. Transmission electron microscopy was used to observe the ultra-structural changes. The viability rate of microcysts and differences in cyst weights were compared by ANOVA, and the cyst numbers were compared using the Kruskal-Wallis test. The combination of ChABZ + ChPZQ nanoparticles was more effective than the ABZ + PZQ suspension in vitro (p < 0.05). In prophylaxy, a significant reduction was observed both in size and in number of the cysts in ChABZ + ChPZQ nanoparticle groups compared with the control group (p < 0.05). In the therapeutic stage, however, this treatment only reduced the cyst numbers. Degeneration of the microcysts treated with the drugs was evident in the ultra-structural imaging. Overall, the nanoparticulate drugs were more effective than their suspension counterparts, but further studies are recommended to evaluate the full potential of these nanoparticles in the treatment of human CE.
Collapse
Affiliation(s)
- Negin Torabi
- Department of Parasitology and Mycology, School of Medicine, Zanjan University of Medical Sciences (ZUMS), Mahdavi Blvd., Shahrak Karmandan, P.O. Box 45139-56111, Zanjan, Iran
| | | | | | - Ali Haniloo
- Department of Parasitology and Mycology, School of Medicine, Zanjan University of Medical Sciences (ZUMS), Mahdavi Blvd., Shahrak Karmandan, P.O. Box 45139-56111, Zanjan, Iran.
| |
Collapse
|
64
|
Chitin Deacetylases: Structures, Specificities, and Biotech Applications. Polymers (Basel) 2018; 10:polym10040352. [PMID: 30966387 PMCID: PMC6415152 DOI: 10.3390/polym10040352] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/15/2018] [Accepted: 03/19/2018] [Indexed: 12/20/2022] Open
Abstract
Depolymerization and de-N-acetylation of chitin by chitinases and deacetylases generates a series of derivatives including chitosans and chitooligosaccharides (COS), which are involved in molecular recognition events such as modulation of cell signaling and morphogenesis, immune responses, and host-pathogen interactions. Chitosans and COS are also attractive scaffolds for the development of bionanomaterials for drug/gene delivery and tissue engineering applications. Most of the biological activities associated with COS seem to be largely dependent not only on the degree of polymerization but also on the acetylation pattern, which defines the charge density and distribution of GlcNAc and GlcNH₂ moieties in chitosans and COS. Chitin de-N-acetylases (CDAs) catalyze the hydrolysis of the acetamido group in GlcNAc residues of chitin, chitosan, and COS. The deacetylation patterns are diverse, some CDAs being specific for single positions, others showing multiple attack, processivity or random actions. This review summarizes the current knowledge on substrate specificity of bacterial and fungal CDAs, focusing on the structural and molecular aspects of their modes of action. Understanding the structural determinants of specificity will not only contribute to unravelling structure-function relationships, but also to use and engineer CDAs as biocatalysts for the production of tailor-made chitosans and COS for a growing number of applications.
Collapse
|
65
|
Substrate Recognition and Specificity of Chitin Deacetylases and Related Family 4 Carbohydrate Esterases. Int J Mol Sci 2018; 19:ijms19020412. [PMID: 29385775 PMCID: PMC5855634 DOI: 10.3390/ijms19020412] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 01/22/2018] [Accepted: 01/24/2018] [Indexed: 12/27/2022] Open
Abstract
Carbohydrate esterases family 4 (CE4 enzymes) includes chitin and peptidoglycan deacetylases, acetylxylan esterases, and poly-N-acetylglucosamine deacetylases that act on structural polysaccharides, altering their physicochemical properties, and participating in diverse biological functions. Chitin and peptidoglycan deacetylases are not only involved in cell wall morphogenesis and remodeling in fungi and bacteria, but they are also used by pathogenic microorganisms to evade host defense mechanisms. Likewise, biofilm formation in bacteria requires partial deacetylation of extracellular polysaccharides mediated by poly-N-acetylglucosamine deacetylases. Such biological functions make these enzymes attractive targets for drug design against pathogenic fungi and bacteria. On the other side, acetylxylan esterases deacetylate plant cell wall complex xylans to make them accessible to hydrolases, making them attractive biocatalysts for biomass utilization. CE4 family members are metal-dependent hydrolases. They are highly specific for their particular substrates, and show diverse modes of action, exhibiting either processive, multiple attack, or patterned deacetylation mechanisms. However, the determinants of substrate specificity remain poorly understood. Here, we review the current knowledge on the structure, activity, and specificity of CE4 enzymes, focusing on chitin deacetylases and related enzymes active on N-acetylglucosamine-containing oligo and polysaccharides.
Collapse
|
66
|
Huang T, Song X, Jing J, Zhao K, Shen Y, Zhang X, Yue B. Chitosan-DNA nanoparticles enhanced the immunogenicity of multivalent DNA vaccination on mice against Trueperella pyogenes infection. J Nanobiotechnology 2018; 16:8. [PMID: 29378591 PMCID: PMC5787914 DOI: 10.1186/s12951-018-0337-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/19/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Trueperella pyogenes is a commensal and opportunistic pathogen that normally causes mastitis, liver abscesses and pneumonia of economically important livestock. To develop efficacious and potent vaccine against T. pyogenes, chimeric gene DNA vaccines were constructed and encapsulated in chitosan nanoparticles (pPCFN-CpG-CS-NPs). RESULTS The pPCFN-CpG-CS-NPs consists of the plo, cbpA, fimA, and nanH gene of T. pyogenes and CpG ODN1826. It was produced with good morphology, high stability, a mean diameter of 93.58 nm, and a zeta potential of + 5.27 mV. Additionally, chitosan encapsulation was confirmed to protect the DNA plasmid from DNase I digestion. The immunofluorescence assay indicated that the four-chimeric gene could synchronously express in HEK293T cells and maintain good bioactivity. Compared to the mice immunized with the control plasmid, in vivo immunization showed that mice immunized with the pPCFN-CpG-CS-NPs had better immune responses, and release of the plasmid DNA was prolonged. Importantly, immunization with pPCFN-CpG-CS-NPs could significantly protect mice from highly virulent T. pyogenes TP7 infection. CONCLUSIONS This study indicates that chitosan-DNA nanoparticles are potent immunization candidates against T. pyogenes infection and provides strategies for the further development of novel vaccines encapsulated in chitosan nanoparticles.
Collapse
Affiliation(s)
- Ting Huang
- Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Xuhao Song
- Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Jie Jing
- Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Kelei Zhao
- Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Yongmei Shen
- Sichuan Engineering Technology Research Center of Medical Animal, Chengdu, China
| | - Xiuyue Zhang
- Key Laboratory of Bio-resources and Eco-environment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu, China
| | - Bisong Yue
- Sichuan Key Laboratory of Conservation Biology on Endangered Wildlife, College of Life Sciences, Sichuan University, Chengdu, 610064, Sichuan, China.
| |
Collapse
|
67
|
Schubert J, Chanana M. Coating Matters: Review on Colloidal Stability of Nanoparticles with Biocompatible Coatings in Biological Media, Living Cells and Organisms. Curr Med Chem 2018; 25:4553-4586. [PMID: 29852857 PMCID: PMC7040520 DOI: 10.2174/0929867325666180601101859] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/13/2018] [Accepted: 04/18/2018] [Indexed: 12/21/2022]
Abstract
Within the last two decades, the field of nanomedicine has not developed as successfully as has widely been hoped for. The main reason for this is the immense complexity of the biological systems, including the physico-chemical properties of the biological fluids as well as the biochemistry and the physiology of living systems. The nanoparticles' physicochemical properties are also highly important. These differ profoundly from those of freshly synthesized particles when applied in biological/living systems as recent research in this field reveals. The physico-chemical properties of nanoparticles are predefined by their structural and functional design (core and coating material) and are highly affected by their interaction with the environment (temperature, pH, salt, proteins, cells). Since the coating material is the first part of the particle to come in contact with the environment, it does not only provide biocompatibility, but also defines the behavior (e.g. colloidal stability) and the fate (degradation, excretion, accumulation) of nanoparticles in the living systems. Hence, the coating matters, particularly for a nanoparticle system for biomedical applications, which has to fulfill its task in the complex environment of biological fluids, cells and organisms. In this review, we evaluate the performance of different coating materials for nanoparticles concerning their ability to provide colloidal stability in biological media and living systems.
Collapse
Affiliation(s)
- Jonas Schubert
- Address correspondence to these authors at the Department of Nanostructured Materials, Leibniz-Institut für Polymerforschung Dresden, Dresden, Germany and Department of Physical Chemistry II, University of Bayreuth, 95447 Bayreuth, Germany;E-mails: ;
| | - Munish Chanana
- Address correspondence to these authors at the Department of Nanostructured Materials, Leibniz-Institut für Polymerforschung Dresden, Dresden, Germany and Department of Physical Chemistry II, University of Bayreuth, 95447 Bayreuth, Germany;E-mails: ;
| |
Collapse
|
68
|
Biodegradable Chitosan Decreases the Immune Response to Trichinella spiralis in Mice. Molecules 2017; 22:molecules22112008. [PMID: 29156562 PMCID: PMC6150330 DOI: 10.3390/molecules22112008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/12/2017] [Accepted: 11/16/2017] [Indexed: 12/16/2022] Open
Abstract
The purpose of this study was to evaluate the potential of chitosan units released during natural degradation of the polymer to activate the immune system against T. spiralis infection. High molecular weight chitosan was injected intraperitoneally into C57BL/6 mice. Flow cytometry and cytokine concentration, measured by ELISA, were used to characterize peritoneal cell populations during T. spiralis infection. The strong chemo-attractive properties of chitosan caused considerable infiltration into the peritoneal cavity of CD11b+ cells, with reduced expression of MHC class II, CD80, CD86, Dectin-1 or CD23 receptors in comparison to T. spiralis-infected mice. After prolonged chitosan biodegradation, cell populations expressing IL-4R, MR and Dectin-1 receptors were found to coexist with elevated IL-6, IL-10, TGF-β and IgA production. IgA cross-reacted with T. spiralis antigen and chitosan. It was found that chitosan treatment attracted immune cells with low activity, which resulted in the number of nematodes increasing. The glucosamine and N-acetyl-D-glucosamine residues were recognized by wheat germ agglutinin (WGA) lectin and therefore any biodegradable chitosan units may actively downregulate the immune response to the parasite. The findings are relevant for both people and animals treated with chitosan preparations.
Collapse
|
69
|
Food contact materials and gut health: Implications for toxicity assessment and relevance of high molecular weight migrants. Food Chem Toxicol 2017; 109:1-18. [PMID: 28830834 DOI: 10.1016/j.fct.2017.08.023] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 08/17/2017] [Accepted: 08/19/2017] [Indexed: 02/08/2023]
Abstract
Gut health is determined by an intact epithelial barrier and balanced gut microbiota, both involved in the regulation of immune responses in the gut. Disruption of this system contributes to the etiology of various non-communicable diseases, including intestinal, metabolic, and autoimmune disorders. Studies suggest that some direct food additives, but also some food contaminants, such as pesticide residues and substances migrating from food contact materials (FCMs), may adversely affect the gut barrier or gut microbiota. Here, we focus on gut-related effects of FCM-relevant substances (e.g. surfactants, N-ring containing substances, nanoparticles, and antimicrobials) and show that gut health is an underappreciated target in the toxicity assessment of FCMs. Understanding FCMs' impact on gut health requires more attention to ensure safety and prevent gut-related chronic diseases. Our review further points to the existence of large population subgroups with an increased intestinal permeability; this may lead to higher uptake of compounds of not only low (<1000 Da) but also high (>1000 Da) molecular weight. We discuss the potential toxicological relevance of high molecular weight compounds in the gut and suggest that the scientific justification for the application of a molecular weight-based cut-off in risk assessment of FCMs should be reevaluated.
Collapse
|
70
|
Effect of Chitosan and Liposome Nanoparticles as Adjuvant Codelivery on the Immunoglobulin G Subclass Distribution in a Mouse Model. J Immunol Res 2017; 2017:9125048. [PMID: 28758135 PMCID: PMC5516754 DOI: 10.1155/2017/9125048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/16/2017] [Accepted: 06/04/2017] [Indexed: 12/02/2022] Open
Abstract
Background We investigate the immunogenic properties of chitosan and liposome nanoparticles as adjuvant codelivery against a commercial pneumococcal conjugate vaccine (PCV) in an animal model. Methods The chitosan and liposome nanoparticles were prepared by ionic gelation and dry methods, respectively. The PCV immunization was performed intradermally in the presence of adjuvants and booster injections which were given without an adjuvant. The Quil-A® was used as a control adjuvant. The ELISA was performed to measure the antibodies against pneumococcal type 14 polysaccharide (Pn14PS). Results The level of total antibodies against Pn14PS antigen was no different between the mouse groups with or without adjuvant codelivery. Codelivery of the PCV with chitosan nanoparticles as well as the Quil-A adjuvant elicited IgG1, IgG2a, IgG2b, and IgG3 antibodies. Meanwhile, codelivery of liposome nanoparticles elicited mainly IgG1 antibodies against the Pn14PS. Conclusions The chitosan and liposome nanoparticles as adjuvant codelivery were successfully synthesized. These nanoparticles have different shapes in particle formation, liposome nanoparticle with their unilamellar shape and chitosan nanoparticles in large shape due to the aggregation of small-size particles. Codelivery of chitosan nanoparticles has more effect on the IgG subclass antibody production than that of liposome nanoparticles in a mouse model.
Collapse
|
71
|
Dosekova E, Filip J, Bertok T, Both P, Kasak P, Tkac J. Nanotechnology in Glycomics: Applications in Diagnostics, Therapy, Imaging, and Separation Processes. Med Res Rev 2017; 37:514-626. [PMID: 27859448 PMCID: PMC5659385 DOI: 10.1002/med.21420] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 09/08/2016] [Accepted: 09/21/2016] [Indexed: 12/14/2022]
Abstract
This review comprehensively covers the most recent achievements (from 2013) in the successful integration of nanomaterials in the field of glycomics. The first part of the paper addresses the beneficial properties of nanomaterials for the construction of biosensors, bioanalytical devices, and protocols for the detection of various analytes, including viruses and whole cells, together with their key characteristics. The second part of the review focuses on the application of nanomaterials integrated with glycans for various biomedical applications, that is, vaccines against viral and bacterial infections and cancer cells, as therapeutic agents, for in vivo imaging and nuclear magnetic resonance imaging, and for selective drug delivery. The final part of the review describes various ways in which glycan enrichment can be effectively done using nanomaterials, molecularly imprinted polymers with polymer thickness controlled at the nanoscale, with a subsequent analysis of glycans by mass spectrometry. A short section describing an active glycoprofiling by microengines (microrockets) is covered as well.
Collapse
Affiliation(s)
- Erika Dosekova
- Department of Glycobiotechnology, Institute of ChemistrySlovak Academy of SciencesDubravska cesta 9845 38BratislavaSlovakia
| | - Jaroslav Filip
- Center for Advanced MaterialsQatar UniversityP.O. Box 2713DohaQatar
| | - Tomas Bertok
- Department of Glycobiotechnology, Institute of ChemistrySlovak Academy of SciencesDubravska cesta 9845 38BratislavaSlovakia
| | - Peter Both
- School of Chemistry, Manchester Institute of BiotechnologyThe University of Manchester131 Princess StreetManchesterM1 7DNUK
| | - Peter Kasak
- Center for Advanced MaterialsQatar UniversityP.O. Box 2713DohaQatar
| | - Jan Tkac
- Department of Glycobiotechnology, Institute of ChemistrySlovak Academy of SciencesDubravska cesta 9845 38BratislavaSlovakia
| |
Collapse
|
72
|
Implications of molecular diversity of chitin and its derivatives. Appl Microbiol Biotechnol 2017; 101:3513-3536. [DOI: 10.1007/s00253-017-8229-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/26/2017] [Accepted: 03/04/2017] [Indexed: 02/03/2023]
|
73
|
El-Hussein A, Lam SSK, Raker J, Chen WR, Hamblin MR. N-dihydrogalactochitosan as a potent immune activator for dendritic cells. J Biomed Mater Res A 2017; 105:963-972. [PMID: 28028922 DOI: 10.1002/jbm.a.35991] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 12/15/2016] [Accepted: 12/21/2016] [Indexed: 12/12/2022]
Abstract
Immunotherapy has become one of the fastest growing areas of cancer research. A promising in situ autologous cancer vaccine (inCVAX) uses a novel immune activator, N-dihydrogalactochitosan (GC), that possesses the ability to stimulate dendritic cells (DC). inCVAX is a combination treatment procedure involving treatment of the tumor with a thermal near-infrared laser to liberate whole cell tumor antigens, followed by injection of GC (a glucosamine polymer with galactose attached to the amino groups) into the treated tumor thereby inducing a systemic antitumor immune response. Regression of both the treated tumor and distant untreated metastases has been observed in both nonclinical and clinical settings following inCVAX. We studied the stimulatory action of GC on relatively immature DCs (DC2.4 cell line) in vitro. GC at 1 mg/mL was a potent stimulator for DC with limited toxicity, giving increased expression of major histocompatibility complex class 2, CD80, and CD11c. Confocal imaging also revealed qualitatively increased uptake of antigen (Texas red-labeled ovalbumin) by DCs after the introduction of GC. To visualize cellular uptake, GC was conjugated with FITC-fluorophore revealing its cellular internalization after 8 hours. In some cases GC was more effective than the toxic TLR4 agonist, lipopolysaccharide. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 963-972, 2017.
Collapse
Affiliation(s)
- Ahmed El-Hussein
- Massachusetts General Hospital, Wellman Center for Photomedicine, Boston, Massachusetts 02114.,Department of Dermatology, Harvard Medical School, Boston, Massachusetts 02115.,The National Institute of Laser Enhanced Science, Cairo University, Cairo, Egypt
| | - Samuel S K Lam
- Immunophotonics, Inc, 4320 Forest Park Ave, Suite #303, St. Louis, Missouri 63108
| | - Joseph Raker
- Immunophotonics, Inc, 4320 Forest Park Ave, Suite #303, St. Louis, Missouri 63108
| | - Wei R Chen
- University of Central Oklahoma, 100 N University Dr, Edmond, Oklahoma 73034
| | - Michael R Hamblin
- Massachusetts General Hospital, Wellman Center for Photomedicine, Boston, Massachusetts 02114.,Department of Dermatology, Harvard Medical School, Boston, Massachusetts 02115.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, Massachusetts 02139
| |
Collapse
|
74
|
Molecular Weight-Dependent Immunostimulative Activity of Low Molecular Weight Chitosan via Regulating NF-κB and AP-1 Signaling Pathways in RAW264.7 Macrophages. Mar Drugs 2016; 14:md14090169. [PMID: 27657093 PMCID: PMC5039540 DOI: 10.3390/md14090169] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 09/12/2016] [Accepted: 09/13/2016] [Indexed: 11/17/2022] Open
Abstract
Chitosan and its derivatives such as low molecular weight chitosans (LMWCs) have been found to possess many important biological properties, such as antioxidant and antitumor effects. In our previous study, LMWCs were found to elicit a strong immunomodulatory response in macrophages dependent on molecular weight. Herein we further investigated the molecular weight-dependent immunostimulative activity of LMWCs and elucidated its mechanism of action on RAW264.7 macrophages. LMWCs (3 kDa and 50 kDa of molecular weight) could significantly enhance the mRNA expression levels of COX-2, IL-10 and MCP-1 in a molecular weight and concentration-dependent manner. The results suggested that LMWCs elicited a significant immunomodulatory response, which was dependent on the dose and the molecular weight. Regarding the possible molecular mechanism of action, LMWCs promoted the expression of the genes of key molecules in NF-κB and AP-1 pathways, including IKKβ, TRAF6 and JNK1, and induced the phosphorylation of protein IKBα in RAW264.7 macrophage. Moreover, LMWCs increased nuclear translocation of p65 and activation of activator protein-1 (AP-1, C-Jun and C-Fos) in a molecular weight-dependent manner. Taken together, our findings suggested that LMWCs exert immunostimulative activity via activation of NF-κB and AP-1 pathways in RAW264.7 macrophages in a molecular weight-dependent manner and that 3 kDa LMWC shows great potential as a novel agent for the treatment of immune suppression diseases and in future vaccines.
Collapse
|
75
|
Corbi-Verge C, Garton M, Nim S, Kim PM. Strategies to Develop Inhibitors of Motif-Mediated Protein-Protein Interactions as Drug Leads. Annu Rev Pharmacol Toxicol 2016; 57:39-60. [PMID: 27618737 DOI: 10.1146/annurev-pharmtox-010716-104805] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Protein-protein interactions are fundamental for virtually all functions of the cell. A large fraction of these interactions involve short peptide motifs, and there has been increased interest in targeting them using peptide-based therapeutics. Peptides benefit from being specific, relatively safe, and easy to produce. They are also easy to modify using chemical synthesis and molecular biology techniques. However, significant challenges remain regarding the use of peptides as therapeutic agents. Identification of peptide motifs is difficult, and peptides typically display low cell permeability and sensitivity to enzymatic degradation. In this review, we outline the principal high-throughput methodologies for motif discovery and describe current methods for overcoming pharmacokinetic and bioavailability limitations.
Collapse
Affiliation(s)
- Carles Corbi-Verge
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada; , , ,
| | - Michael Garton
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada; , , ,
| | - Satra Nim
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada; , , ,
| | - Philip M Kim
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada; , , , .,Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 3E1, Canada.,Department of Computer Science, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| |
Collapse
|
76
|
Qi X, Lam SS, Liu D, Kim DY, Ma L, Alleruzzo L, Chen W, Hode T, Henry CJ, Kaifi J, Kimchi ET, Li G, Staveley-O'Carroll KF. Development of inCVAX, In situ Cancer Vaccine, and Its Immune Response in Mice with Hepatocellular Cancer. ACTA ACUST UNITED AC 2016; 7. [PMID: 27656328 PMCID: PMC5027967 DOI: 10.4172/2155-9899.1000438] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Manipulation of immune system toward the rejection of established cancers has become the standard of care in some patients. Here we propose the development of an in situ autologous cancer vaccine, inCVAX, for the treatment of hepatocellular cancer (HCC). inCVAX is based on the induction of local immunogenic cancer cell death combined with local dendritic cell stimulation by intratumoral injection of the immune-activator N-dihydro-galacto-chitosan (GC). In a first set of experiments, cellular and molecular studies were performed to investigate the effect of inCVAX on immune activation in a murine model of HCC that we previously developed. Once large tumors were formed in mice, the tumor is surgically exposed and a laser fiber was inserted into the center of an individual tumor mass. Using a 10 mm diffuser tip, laser irradiation of 1.5 W was applied to heat the tumor at different durations (6-10 min) to assess tolerability of photothermal application at different temperatures. The laser application was followed by immediate injection of GC, and each mouse received one laser treatment and one GC injection. ELISA was used to assess the level of cytokines; immunohistochemical staining was conducted to analyze the effect of inCVAX on immune cell tumor-filtration and expression of tumor-specific antigens (TSAs) and tumor-associated antigens (TAAs). Results indicate that survival correlated to thermal exposure. At lower temperatures the photothermal effect was sufficient to induce tumor necrosis, but without obvious complication to the mice, although at these temperatures the treatment didn’t alter the level of TSAs and TAAs, so further optimization is suggested. Nevertheless, in response to the inCVAX treatment, cytotoxic cytokine IFN-γ was significantly increased, but suppressive cytokine TGF-β was dramatically reduced. Furthermore, inCVAX prompted tumor infiltration of CD3+, CD4+, and CD8+ T cells; but modulated macrophage subsets differently. In conclusion, while the protocol needs further optimization, it would appear that inCVAX for the treatment of HCC activates an immune response in tumor-bearing mice, which in turn may have potential for the treatment of HCC.
Collapse
Affiliation(s)
- Xiaoqiang Qi
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA ; Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| | - Samuel Sk Lam
- Immunophotonics Inc., 4320 Forest Park Avenue #303, St. Louis, Missouri 63108, USA
| | - Dai Liu
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA ; Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| | - Dae Young Kim
- Veterinary Medical Diagnostic Laboratory, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Lixin Ma
- Department of Radiology, University of Missouri, Columbia, MO 65212; Harry S. Truman Memorial VA Hospital Biomolecular Imaging Center, USA
| | - Lu Alleruzzo
- Immunophotonics Inc., 4320 Forest Park Avenue #303, St. Louis, Missouri 63108, USA
| | - Wei Chen
- Veterinary Medical and Surgery, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Tomas Hode
- Immunophotonics Inc., 4320 Forest Park Avenue #303, St. Louis, Missouri 63108, USA
| | - Carolyn J Henry
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65212, USA
| | - Jussuf Kaifi
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA ; Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| | - Eric T Kimchi
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA ; Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| | - Guangfu Li
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA ; Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA
| | - Kevin F Staveley-O'Carroll
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA ; Ellis Fischel Cancer Center, University of Missouri, Columbia, MO 65212, USA ; Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
77
|
Temizoz B, Kuroda E, Ishii KJ. Vaccine adjuvants as potential cancer immunotherapeutics. Int Immunol 2016; 28:329-38. [PMID: 27006304 PMCID: PMC4922024 DOI: 10.1093/intimm/dxw015] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 03/14/2016] [Indexed: 12/11/2022] Open
Abstract
Accumulated evidence obtained from various clinical trials and animal studies suggested that cancer vaccines need better adjuvants than those that are currently licensed, which include the most commonly used alum and incomplete Freund's adjuvant, because of either a lack of potent anti-tumor immunity or the induction of undesired immunity. Several clinical trials using immunostimulatory adjuvants, particularly agonistic as well as non-agonistic ligands for TLRs, C-type lectin receptors, retinoic acid-inducible gene I-like receptors and stimulator of interferon genes, have revealed their therapeutic potential not only as vaccine adjuvants but also as anti-tumor agents. Recently, combinations of such immunostimulatory or immunomodulatory adjuvants have shown superior efficacy over their singular use, suggesting that seeking optimal combinations of the currently available or well-characterized adjuvants may provide a better chance for the development of novel adjuvants for cancer immunotherapy.
Collapse
Affiliation(s)
- Burcu Temizoz
- Laboratory of Vaccine Science, WPI Immunology Frontier Research Center (iFReC), Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi, Saito, Ibaraki-City, Osaka 567-0085, Japan
| | - Etsushi Kuroda
- Laboratory of Vaccine Science, WPI Immunology Frontier Research Center (iFReC), Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi, Saito, Ibaraki-City, Osaka 567-0085, Japan
| | - Ken J Ishii
- Laboratory of Vaccine Science, WPI Immunology Frontier Research Center (iFReC), Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan Laboratory of Adjuvant Innovation, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), 7-6-8 Asagi, Saito, Ibaraki-City, Osaka 567-0085, Japan
| |
Collapse
|
78
|
|
79
|
Lee YH, Park HI, Choi JS. Novel glycol chitosan-based polymeric gene carrier synthesized by a Michael addition reaction with low molecular weight polyethylenimine. Carbohydr Polym 2016; 137:669-677. [DOI: 10.1016/j.carbpol.2015.10.089] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 09/25/2015] [Accepted: 10/28/2015] [Indexed: 12/27/2022]
|
80
|
Immune cell impact of three differently coated lipid nanocapsules: pluronic, chitosan and polyethylene glycol. Sci Rep 2016; 6:18423. [PMID: 26728491 PMCID: PMC4700454 DOI: 10.1038/srep18423] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 11/09/2015] [Indexed: 12/23/2022] Open
Abstract
Lipid nanocapsules (NCs) represent promising tools in clinical practice for diagnosis and therapy applications. However, the NC appropriate functionalization is essential to guarantee high biocompatibility and molecule loading ability. In any medical application, the immune system-impact of differently functionalized NCs still remains to be fully understood. A comprehensive study on the action exerted on human peripheral blood mononuclear cells (PBMCs) and major immune subpopulations by three different NC coatings: pluronic, chitosan and polyethylene glycol-polylactic acid (PEG) is reported. After a deep particle characterization, the uptake was assessed by flow-cytometry and confocal microscopy, focusing then on apoptosis, necrosis and proliferation impact in T cells and monocytes. Cell functionality by cell diameter variations, different activation marker analysis and cytokine assays were performed. We demonstrated that the NCs impact on the immune cell response is strongly correlated to their coating. Pluronic-NCs were able to induce immunomodulation of innate immunity inducing monocyte activations. Immunomodulation was observed in monocytes and T lymphocytes treated with Chitosan-NCs. Conversely, PEG-NCs were completely inert. These findings are of particular value towards a pre-selection of specific NC coatings depending on biomedical purposes for pre-clinical investigations; i.e. the immune-specific action of particular NC coating can be excellent for immunotherapy applications.
Collapse
|
81
|
Skwarczynski M, Toth I. Peptide-based synthetic vaccines. Chem Sci 2015; 7:842-854. [PMID: 28791117 PMCID: PMC5529997 DOI: 10.1039/c5sc03892h] [Citation(s) in RCA: 372] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 12/14/2015] [Indexed: 01/18/2023] Open
Abstract
Classically all vaccines were produced using live or attenuated microorganisms or parts of them. However, the use of whole organisms, their components or the biological process for vaccine production has several weaknesses. The presence of immunologically redundant biological components or biological impurities in such vaccines might cause major problems. All the disadvantageous of traditional vaccines might be overcome via the development of fully synthetic peptide-based vaccines. However, once minimal antigenic epitopes only are applied for immunisation, the immune responses are poor. The use of an adjuvant can overcome this obstacle; however, it may raise new glitches. Here we briefly summarise the current stand on peptide-based vaccines, discuss epitope and adjuvant design, and multi-epitope and nanoparticle-based vaccine approaches. This mini review discusses also the disadvantages and benefits associated with peptide-based vaccines. It proposes possible methods to overcome the weaknesses of the synthetic vaccine strategy and suggests future directions for its development.
Collapse
Affiliation(s)
- Mariusz Skwarczynski
- The University of Queensland , School of Chemistry and Molecular Biosciences , St Lucia 4072 , Australia .
| | - Istvan Toth
- The University of Queensland , School of Chemistry and Molecular Biosciences , St Lucia 4072 , Australia . .,The University of Queensland , Institute for Molecular Bioscience , St Lucia 4072 , Australia.,The University of Queensland , School of Pharmacy , Brisbane , QLD 4072 , Australia
| |
Collapse
|
82
|
Poecheim J, Heuking S, Brunner L, Barnier-Quer C, Collin N, Borchard G. Nanocarriers for DNA Vaccines: Co-Delivery of TLR-9 and NLR-2 Ligands Leads to Synergistic Enhancement of Proinflammatory Cytokine Release. NANOMATERIALS 2015; 5:2317-2334. [PMID: 28347123 PMCID: PMC5304807 DOI: 10.3390/nano5042317] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/23/2015] [Accepted: 12/02/2015] [Indexed: 12/12/2022]
Abstract
Adjuvants enhance immunogenicity of vaccines through either targeted antigen delivery or stimulation of immune receptors. Three cationic nanoparticle formulations were evaluated for their potential as carriers for a DNA vaccine, and muramyl dipeptide (MDP) as immunostimulatory agent, to induce and increase immunogenicity of Mycobacterium tuberculosis antigen encoding plasmid DNA (pDNA). The formulations included (1) trimethyl chitosan (TMC) nanoparticles, (2) a squalene-in-water nanoemulsion, and (3) a mineral oil-in-water nanoemulsion. The adjuvant effect of the pDNA-nanocomplexes was evaluated by serum antibody analysis in immunized mice. All three carriers display a strong adjuvant effect, however, only TMC nanoparticles were capable to bias immune responses towards Th1. pDNA naturally contains immunostimulatory unmethylated CpG motifs that are recognized by Toll-like receptor 9 (TLR-9). In mechanistic in vitro studies, activation of TLR-9 and the ability to enhance immunogenicity by simultaneously targeting TLR-9 and NOD-like receptor 2 (NLR-2) was determined by proinflammatory cytokine release in RAW264.7 macrophages. pDNA in combination with MDP was shown to significantly increase proinflammatory cytokine release in a synergistic manner, dependent on NLR-2 activation. In summary, novel pDNA-Ag85A loaded nanoparticle formulations, which induce antigen specific immune responses in mice were developed, taking advantage of the synergistic combinations of TLR and NLR agonists to increase the adjuvanticity of the carriers used.
Collapse
Affiliation(s)
- Johanna Poecheim
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Quai Ernest Ansermet 30, 1211 Geneva, Switzerland.
| | - Simon Heuking
- Vaccine Formulation Laboratory, Department of Biochemistry, University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland.
| | - Livia Brunner
- Vaccine Formulation Laboratory, Department of Biochemistry, University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland.
| | - Christophe Barnier-Quer
- Vaccine Formulation Laboratory, Department of Biochemistry, University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland.
| | - Nicolas Collin
- Vaccine Formulation Laboratory, Department of Biochemistry, University of Lausanne, Chemin des Boveresses 155, 1066 Epalinges, Switzerland.
| | - Gerrit Borchard
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Quai Ernest Ansermet 30, 1211 Geneva, Switzerland.
| |
Collapse
|
83
|
Oberoi HS, Yorgensen YM, Morasse A, Evans JT, Burkhart DJ. PEG modified liposomes containing CRX-601 adjuvant in combination with methylglycol chitosan enhance the murine sublingual immune response to influenza vaccination. J Control Release 2015; 223:64-74. [PMID: 26551346 DOI: 10.1016/j.jconrel.2015.11.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 11/05/2015] [Indexed: 02/05/2023]
Abstract
The mucosa is the primary point of entry for pathogens making it an important vaccination site to produce a protective mucosal immune response. While the sublingual (SL) mucosa presents several barriers to vaccine penetration, its unique anatomy and physiology makes it one of the best options for mucosal vaccination. Efficient and directed delivery of adjuvants and antigens to appropriate immune mediators in the SL tissue will aid in development of effective SL vaccines against infectious diseases. Herein we demonstrate a robust immune response against influenza antigens co-delivered sublingually with engineered liposomes carrying the synthetic Toll-like receptor-4 agonist, CRX-601. Liposome modification with PEG copolymers (Pluronics), phospholipid-PEG conjugates and chitosan were evaluated for their ability to generate an immune response in a SL murine influenza vaccine model. Phospholipid-PEG conjugates were more effective than Pluronic copolymers in generating stable, surface neutral liposomes. SL vaccination with surface modified liposomes carrying CRX-601 adjuvant generated significant improvements in flu-specific responses compared with unmodified liposomes. Furthermore, the coating of modified liposomes with methylglycol chitosan produced the most effective flu-specific immune response. These results demonstrate efficient SL vaccine delivery utilizing a combination of a muco-adhesive and surface neutral liposomes to achieve a robust mucosal and systemic immune response.
Collapse
Affiliation(s)
| | | | - Audrey Morasse
- GSK Vaccines, 525 Boulevard Cartier, Laval, QC H7V 3S8, Canada
| | - Jay T Evans
- GSK Vaccines, 553 Old Corvallis Road, Hamilton, MT 59840, USA
| | | |
Collapse
|
84
|
Wu N, Wen ZS, Xiang XW, Huang YN, Gao Y, Qu YL. Immunostimulative Activity of Low Molecular Weight Chitosans in RAW264.7 Macrophages. Mar Drugs 2015; 13:6210-25. [PMID: 26437419 PMCID: PMC4626685 DOI: 10.3390/md13106210] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/09/2015] [Accepted: 09/21/2015] [Indexed: 12/26/2022] Open
Abstract
Chitosan and its derivatives such as low molecular weight chitosans (LMWCs) have been reported to exert many biological activities, such as antioxidant and antitumor effects. However, complex and molecular weight dependent effects of chitosan remain controversial and the mechanisms that mediate these complex effects are still poorly defined. This study was carried out to investigate the immunostimulative effect of different molecular weight chitosan in RAW264.7 macrophages. Our data suggested that two LMWCs (molecular weight of 3 kDa and 50 kDa) both possessed immunostimulative activity, which was dependent on dose and, at the higher doses, also on the molecular weight. LMWCs could significantly enhance the the pinocytic activity, and induce the production of tumor necrosis factor α (TNF-α), interleukin 6 (IL-6), interferon-γ (IFN-γ), nitric oxide (NO) and inducible nitric oxide synthase (iNOS) in a molecular weight and concentration-dependent manner. LMWCs were further showed to promote the expression of the genes including iNOS, TNF-α. Taken together, our findings suggested that LMWCs elicited significantly immunomodulatory response through up-regulating mRNA expression of proinflammatory cytokines and activated RAW264.7 macrophage in a molecular weight- and concentration-dependent manner.
Collapse
Affiliation(s)
- Ning Wu
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Food and Pharmacy College, Zhejiang Ocean University, Zhoushan 316000, China.
| | - Zheng-Shun Wen
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Food and Pharmacy College, Zhejiang Ocean University, Zhoushan 316000, China.
| | - Xing-Wei Xiang
- Zhejiang Marine Development Research Institute, Zhoushan 316000, China.
| | - Yan-Na Huang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, China.
| | - Yang Gao
- School of Fishery, Zhejiang Ocean University, Zhoushan, 316000, China.
| | - You-Le Qu
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, Food and Pharmacy College, Zhejiang Ocean University, Zhoushan 316000, China.
| |
Collapse
|
85
|
Salazar-González JA, González-Ortega O, Rosales-Mendoza S. Gold nanoparticles and vaccine development. Expert Rev Vaccines 2015; 14:1197-211. [DOI: 10.1586/14760584.2015.1064772] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Jorge Alberto Salazar-González
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, México
| | - Omar González-Ortega
- Laboratorio de Biofarmacéuticos Recombinantes, Facultad de Ciencias Químicas, Universidad Autónoma de San Luis Potosí, Av. Dr. Manuel Nava 6, SLP, 78210, México
| | | |
Collapse
|
86
|
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: 3.1] [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
|
87
|
Performance of PRP associated with porous chitosan as a composite scaffold for regenerative medicine. ScientificWorldJournal 2015; 2015:396131. [PMID: 25821851 PMCID: PMC4363634 DOI: 10.1155/2015/396131] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 01/08/2015] [Indexed: 12/01/2022] Open
Abstract
This study aimed to evaluate the in vitro performance of activated platelet-rich plasma associated with porous sponges of chitosan as a composite scaffold for proliferation and osteogenic differentiation of human adipose tissue-derived mesenchymal stem cells. The sponges were prepared by controlled freezing (−20, −80, or −196°C) and lyophilization of chitosan solutions (1, 2, or 3% w/v). The platelet-rich plasma was obtained from controlled centrifugation of whole blood and activated with calcium and autologous serum. The composite scaffolds were prepared by embedding the sponges with the activated platelet-rich plasma. The results showed the performance of the scaffolds was superior to that of activated platelet-rich plasma alone, in terms of delaying the release of growth factors and increased proliferation of the stem cells. The best preparation conditions of chitosan composite scaffolds that coordinated the physicochemical and mechanical properties and cell proliferation were 3% (w/v) chitosan and a −20°C freezing temperature, while −196°C favored osteogenic differentiation. Although the composite scaffolds are promising for regenerative medicine, the structures require stabilization to prevent the collapse observed after five days.
Collapse
|
88
|
InCVAX--a novel strategy for treatment of late-stage, metastatic cancers through photoimmunotherapy induced tumor-specific immunity. Cancer Lett 2015; 359:169-77. [PMID: 25633839 DOI: 10.1016/j.canlet.2015.01.029] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/20/2015] [Accepted: 01/21/2015] [Indexed: 12/31/2022]
Abstract
A novel, promising potential cancer vaccine strategy was proposed to use a two-injection procedure for solid tumors to prompt the immune system to identify and systemically eliminate primary and metastatic cancers. The two-injection procedure consists of local photothermal application on a selected tumor intended to liberate whole cell tumor antigens, followed by a local injection of an immunoadjuvant that consists of a semi-synthetic functionalized glucosamine polymer, N-dihydro-galacto-chitosan (GC), which is intended to activate antigen presenting cells and facilitate an increased uptake of tumor antigens. This strategy is thus proposed as an in situ autologous cancer vaccine (inCVAX) that may activate antigen presenting cells and expose them to tumor antigens in situ, with the intention of inducing a systemic tumor specific T-cell response. Here, the development of inCVAX for the treatment of metastatic cancers in the past decades is systematically reviewed. The antitumor immune responses of local photothermal treatment and immunological stimulation with GC are also discussed. This treatment approach is also commonly referred to as laser immunotherapy (LIT).
Collapse
|
89
|
Skwarczynski M, Toth I. Recent advances in peptide-based subunit nanovaccines. Nanomedicine (Lond) 2014; 9:2657-69. [DOI: 10.2217/nnm.14.187] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Vaccination is the most efficient way to protect humans against pathogens. Peptide-based vaccines offer several advantages over classical vaccines, which utilized whole organisms or proteins. However, peptides alone are not immunogenic and need a delivery system that can boost their recognition by the immune system. In recent years, nanotechnology-based approaches have become one of the most promising strategies in peptide vaccine delivery. This review summarizes knowledge on peptide vaccines and nanotechnology-based approaches for their delivery. The recently reported nano-sized delivery platforms for peptide antigens are reviewed, including nanoparticles composed of polymers, peptides, lipids, inorganic materials and nanotubes. The future prospects for peptide-based nanovaccines are discussed.
Collapse
Affiliation(s)
- Mariusz Skwarczynski
- School of Chemistry & Molecular Biosciences, University of Queensland, St Lucia, Australia
| | - Istvan Toth
- School of Chemistry & Molecular Biosciences, University of Queensland, St Lucia, Australia
| |
Collapse
|
90
|
Jia L, Gao X, Wang Y, Yao N, Zhang X. Structural, phenotypic and functional maturation of bone marrow dendritic cells (BMDCs) induced by Chitosan (CTS). Biologicals 2014; 42:334-8. [PMID: 25225119 DOI: 10.1016/j.biologicals.2014.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 07/15/2014] [Accepted: 07/17/2014] [Indexed: 01/23/2023] Open
Abstract
The objective of the present work was to explore the effect of CTS on structural, phenotypic and functional maturation of murine bone marrow derived dendritic cells (BMDCs). The maturity of BMDCs post treatment with CTS was evaluated using transmission electron microscopy (TEM) for structure changes, flow cytometry (FCM) for changes of key surface molecules, FITC-dextran bio-assay for phagocytosis, test of acid phosphatase activity (ACP) for biochemical changes and enzyme linked immunosorbent assay (ELISA) for cytokine level. We found that CTS downregulated the numbers of phagosomes inside the BMDCs, up-regulated the expression of MHC II, CD40, CD83, CD80 and CD86 molecules on BMDCs, decreased activity of ACP and phagocytosis by BMDCs, and induced production of higher levels of IL-12 and TNF-α. It was therefore confirmed that CTS could effectively promote the maturation of BMDCs. Our study provided more detailed evidence and rationale to support the application of CTS as an immune stimulator for enhancing host immunity and as an adjuvant in the design of DC-based vaccines.
Collapse
Affiliation(s)
- Lihui Jia
- Department of Stomatology, General Hospital of Shenyang Military Area Command, Shenyang 110084, China
| | - Xinghua Gao
- Department of Dermatology, No.1 Hospital, China Medical University, Shenyang 110001, China
| | - Yiqing Wang
- Department of Stomatology, General Hospital of Shenyang Military Area Command, Shenyang 110084, China
| | - Na Yao
- Department of Stomatology, General Hospital of Shenyang Military Area Command, Shenyang 110084, China
| | - Xiaodong Zhang
- Department of Stomatology, General Hospital of Shenyang Military Area Command, Shenyang 110084, China.
| |
Collapse
|
91
|
Abstract
Research in cancer immunotherapy has gained momentum in the last two decades, with many studies and clinical trials showing positive therapeutic outcomes. Immunotherapy can elicit not only a strong anticancer immune response which could even control metastases, but could also induce immunological memory, resulting in long-lasting protection in the prophylactic setting and protection against possible recurrence. Nanocarriers offer an attractive means for delivery of a multitude of therapeutic immunomodulators which are readily taken up by immune cells and can initiate a particular arm of an immunostimulatory cascade leading to tumor cell killing. This review focuses on recent advances in nanocarrier-mediated immunotherapy for the treatment of cancer. Both in vitro and in vivo studies as well as clinical progress are discussed in various sections. Description of the specific role of nanoparticle technology in immunotherapy highlights the way particles can be tailor-made in terms of size, structure, payload, and surface properties for active targeting to antigen-presenting cells and/or enhanced accumulation in the solid tumor.
Collapse
Affiliation(s)
- Manu Smriti Singh
- Laboratory of Pharmaceutical Technology and Biopharmaceutics, University of Bonn, Bonn, Germany
| | - Sangeeta Bhaskar
- Product Development Cell, National Institute of Immunology, New Delhi, India
| |
Collapse
|
92
|
Synergistic effects of glycated chitosan with high-intensity focused ultrasound on suppression of metastases in a syngeneic breast tumor model. Cell Death Dis 2014; 5:e1178. [PMID: 24743733 PMCID: PMC4001313 DOI: 10.1038/cddis.2014.159] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 03/13/2014] [Accepted: 03/13/2014] [Indexed: 01/12/2023]
Abstract
Stimulation of the host immune system is crucial in cancer treatment. In particular, nonspecific immunotherapies, when combined with other traditional therapies such as radiation and chemotherapy, may induce immunity against primary and metastatic tumors. In this study, we demonstrate that a novel, non-toxic immunoadjuvant, glycated chitosan (GC), decreases the motility and invasion of mammalian breast cancer cells in vitro and in vivo. Lung metastatic ratios were reduced in 4T1 tumor-bearing mice when intratumoral GC injection was combined with local high-intensity focused ultrasound (HIFU) treatment. We postulate that this treatment modality stimulates the host immune system to combat cancer cells, as macrophage accumulation in tumor lesions was detected after GC-HIFU treatment. In addition, plasma collected from GC-HIFU-treated tumor-bearing mice exhibited tumor-specific cytotoxicity. We also investigated the effect of GC on epithelial–mesenchymal transition-related markers. Our results showed that GC decreased the expression of Twist-1 and Slug, proto-oncogenes commonly implicated in metastasis. Epithelial-cadherin, which is regulated by these genes, was also upregulated. Taken together, our current data suggest that GC alone can reduce cancer cell motility and invasion, whereas GC-HIFU treatment can induce immune responses to suppress tumor metastasis in vivo.
Collapse
|
93
|
Azmi F, Ahmad Fuaad AAH, Skwarczynski M, Toth I. Recent progress in adjuvant discovery for peptide-based subunit vaccines. Hum Vaccin Immunother 2013; 10:778-96. [PMID: 24300669 DOI: 10.4161/hv.27332] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Peptide-based subunit vaccines are of great interest in modern immunotherapy as they are safe, easy to produce and well defined. However, peptide antigens produce a relatively weak immune response, and thus require the use of immunostimulants (adjuvants) for optimal efficacy. Developing a safe and effective adjuvant remains a challenge for peptide-based vaccine design. Recent advances in immunology have allowed researchers to have a better understanding of the immunological implication of related diseases, which facilitates more rational design of adjuvant systems. Understanding the molecular structure of the adjuvants allows the establishment of their structure-activity relationships which is useful for the development of next-generation adjuvants. This review summarizes the current state of adjuvants development in the field of synthetic peptide-based vaccines. The structural, chemical and biological properties of adjuvants associated with their immunomodulatory effects are discussed.
Collapse
Affiliation(s)
- Fazren Azmi
- School of Chemistry and Molecular Biosciences; The University of Queensland; Brisbane, QLD Australia; Faculty of Pharmacy; National University Malaysia; Kuala Lumpur, Malaysia
| | | | | | | |
Collapse
|
94
|
Bolhassani A, Javanzad S, Saleh T, Hashemi M, Aghasadeghi MR, Sadat SM. Polymeric nanoparticles: potent vectors for vaccine delivery targeting cancer and infectious diseases. Hum Vaccin Immunother 2013; 10:321-32. [PMID: 24128651 PMCID: PMC4185908 DOI: 10.4161/hv.26796] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 10/06/2013] [Accepted: 10/12/2013] [Indexed: 12/13/2022] Open
Abstract
Nanocarriers with various compositions and biological properties have been extensively applied for in vitro/in vivo drug and gene delivery. The family of nanocarriers includes polymeric nanoparticles, lipid-based carriers (liposomes/micelles), dendrimers, carbon nanotubes, and gold nanoparticles (nanoshells/nanocages). Among different delivery systems, polymeric carriers have several properties such as: easy to synthesize, inexpensive, biocompatible, biodegradable, non-immunogenic, non-toxic, and water soluble. In addition, cationic polymers seem to produce more stable complexes led to a more protection during cellular trafficking than cationic lipids. Nanoparticles often show significant adjuvant effects in vaccine delivery since they may be easily taken up by antigen presenting cells (APCs). Natural polymers such as polysaccharides and synthetic polymers have demonstrated great potential to form vaccine nanoparticles. The development of new adjuvants or delivery systems for DNA and protein immunization is an expanding research field. This review describes polymeric carriers especially PLGA, chitosan, and PEI as vaccine delivery systems.
Collapse
Affiliation(s)
- Azam Bolhassani
- Department of Hepatitis and AIDs; Pasteur Institute of Iran; Tehran, Iran
| | - Shabnam Javanzad
- Department of Hepatitis and AIDs; Pasteur Institute of Iran; Tehran, Iran
- Department of genetics; Islamic Azad University; Tehran Medical Branch; Tehran, Iran
| | - Tayebeh Saleh
- Department of Nanobiotechnology; Faculty of Biological Sciences; Tarbiat Modares University; Tehran, Iran
| | - Mehrdad Hashemi
- Department of genetics; Islamic Azad University; Tehran Medical Branch; Tehran, Iran
| | | | - Seyed Mehdi Sadat
- Department of Hepatitis and AIDs; Pasteur Institute of Iran; Tehran, Iran
| |
Collapse
|