1
|
Duymaz D, Kebabci AO, Kizilel S. Harnessing the immunomodulatory potential of chitosan and its derivatives for advanced biomedical applications. Int J Biol Macromol 2025; 307:142055. [PMID: 40090654 DOI: 10.1016/j.ijbiomac.2025.142055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 02/21/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025]
Abstract
The success of biomaterial applications in medicine, particularly in tissue engineering, relies on achieving a balance between promoting tissue regeneration and controlling the immune response. Due to its natural origin, high biocompatibility, and versatility, chitosan has emerged as a promising biomaterial especially for immunomodulation purposes. Immunomodulation, refers to the deliberate alteration of the immune system's activity to achieve a desired therapeutic effect either by enhancing or suppressing the function of specific immune cells, signaling pathways, or cytokine production. This modulation opens up the unlimited possibilities for the use of biomaterials, especially about the use of natural polymers such as chitosan. Although numerous chitosan-based immunoregulatory strategies have been demonstrated over the past two decades, the lack of in-depth exploration hinders the full potential of strategies that include chitosan and its derivatives in biomedical applications. Thus, in this review, the possible immunomodulatory effects of chitosan, chitosan derivatives and their potential combined with various agents and therapies are investigated in detail. Moreover, this report includes agents for localized immune response control, chitosan-based strategies with complementary immunomodulatory properties to create synergistic effects that will influence the success of cell therapies for enhanced tissue acceptance and regeneration. Finally, the challenges and outlook of chitosan-based therapies as a powerful tool for improving immunomodulatory applications are discussed for paving the way for further studies.
Collapse
Affiliation(s)
- Doğukan Duymaz
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye
| | - Aybaran O Kebabci
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye
| | - Seda Kizilel
- Chemical and Biological Engineering, Koç University, Sariyer, İstanbul, Turkiye.
| |
Collapse
|
2
|
Kim YH, Park CH, Kim JM, Yoon YC. Chitooligosaccharides suppress airway inflammation, fibrosis, and mucus hypersecretion in a house dust mite-induced allergy model. FRONTIERS IN ALLERGY 2025; 6:1533928. [PMID: 39927112 PMCID: PMC11799285 DOI: 10.3389/falgy.2025.1533928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/09/2025] [Indexed: 02/11/2025] Open
Abstract
Background Respiratory allergy is a serious respiratory disorder characterized by inflammation, mucus hypersecretion, and airway tissue sclerosis. Disruption of the T helper 1 (Th1) and T helper 2 (Th2) immune systems by stimuli induced by house dust mites (HDM) and fine particulate matter leads to the secretion of various inflammatory cytokines, resulting in immune respiratory diseases characterized by airway inflammation. Chitooligosaccharides (COS) are known for their antioxidant and anti-inflammatory properties. Methods Human airway epithelial cells (BEAS-2B) were cultured in DMEM/F12 medium containing COS at concentrations of 25-100 µg/ml for 24 h. No intracellular toxicity was observed up to 1,000 µg/ml. Cell experiments were conducted at COS concentrations below 100 µg/ml, while animal experiments were performed at concentrations below 100 mg/kg body weight for 4 weeks. Samples of right lung tissue obtained from the experimental animals were used for gene and protein expression analysis, whereas samples of contralateral lung tissue were used for immunohistochemical analysis. Results COS regulated Th1 immunity by inhibiting major cytokines, including inflammatory tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), in BEAS-2B cells. In the HDM-induced allergic respiratory model, COS suppressed the infiltration of inflammatory cells around the airways and inhibited the mRNA expression of Th1 immune cytokines in lung tissues, while also reducing the expression of nuclear factor kappa B (NF-κB)-related proteins. Furthermore, the results confirmed the suppression of the levels of immunoglobulin E (IgE) in the blood secreted by mast cells activated by HDM, which led to a reduction in allergic mucus hypersecretion and airway sclerosis. Conclusion In summary, COS are thought to improve airway resistance by alleviating inflammatory allergic respiratory diseases caused by HDM and are regarded as substances that regulate the balance of the Th1 and Th2 immune systems in epithelial cells affected by mucus hypersecretion.
Collapse
Affiliation(s)
| | | | | | - Yeo Cho Yoon
- Healthcare & Nutrition Laboratory, Amicogen, Inc., Seongnam, Republic of Korea
| |
Collapse
|
3
|
Ghattas M, Dwivedi G, Chevrier A, Horn-Bourque D, Alameh MG, Lavertu M. Chitosan immunomodulation: insights into mechanisms of action on immune cells and signaling pathways. RSC Adv 2025; 15:896-909. [PMID: 39802469 PMCID: PMC11719903 DOI: 10.1039/d4ra08406c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 12/22/2024] [Indexed: 01/16/2025] Open
Abstract
Chitosan, a biodegradable and biocompatible natural polymer composed of β-(1-4)-linked N-acetyl glucosamine (GlcNAc) and d-glucosamine (GlcN) and derived from crustacean shells, has been widely studied for various biomedical applications, including drug delivery, cartilage repair, wound healing, and tissue engineering, because of its unique physicochemical properties. One of the most promising areas of research is the investigation of the immunomodulatory properties of chitosan, since the biopolymer has been shown to modulate the maturation, activation, cytokine production, and polarization of dendritic cells and macrophages, two key immune cells involved in the initiation and regulation of innate and adaptive immune responses, leading to enhanced immune responses. Several signaling pathways, including the cGAS-STING, STAT-1, and NLRP3 inflammasomes, are involved in chitosan-induced immunomodulation. This review provides a comprehensive overview of the current understanding of the in vitro immunomodulatory effects of chitosan. This information may facilitate the development of chitosan-based therapies and vaccine adjuvants for various immune-related diseases.
Collapse
Affiliation(s)
- Majed Ghattas
- Department of Chemical Engineering, Polytechnique Montreal Montreal QC Canada
- Institute of Biomedical Engineering, Polytechnique Montreal Montreal QC Canada
| | - Garima Dwivedi
- Perelman School of Medicine, University of Pennsylvania Philadelphia PA USA
| | - Anik Chevrier
- Department of Chemical Engineering, Polytechnique Montreal Montreal QC Canada
| | - Delano Horn-Bourque
- Department of Chemical Engineering, Polytechnique Montreal Montreal QC Canada
- Institute of Biomedical Engineering, Polytechnique Montreal Montreal QC Canada
| | - Mohamad-Gabriel Alameh
- Perelman School of Medicine, University of Pennsylvania Philadelphia PA USA
- Penn Institute for RNA Innovation, University of Pennsylvania Philadelphia PA USA
| | - Marc Lavertu
- Department of Chemical Engineering, Polytechnique Montreal Montreal QC Canada
- Institute of Biomedical Engineering, Polytechnique Montreal Montreal QC Canada
| |
Collapse
|
4
|
Elkhenany H, Soliman MW, Atta D, El-Badri N. Innovative Marine-Sourced Hydroxyapatite, Chitosan, Collagen, and Gelatin for Eco-Friendly Bone and Cartilage Regeneration. J Biomed Mater Res A 2025; 113:e37833. [PMID: 39508545 DOI: 10.1002/jbm.a.37833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/16/2024] [Accepted: 10/25/2024] [Indexed: 11/15/2024]
Abstract
In recent years, the exploration of sustainable alternatives in the field of bone tissue engineering has led researchers to focus on marine waste byproducts as a valuable resource. These marine resources, often overlooked remnants of various industries, exhibit a rich composition of hydroxyapatite, collagen, calcium carbonate, and other minerals essential to the complex framework of bone structure. Marine waste by-products can emit gases such as methane and carbon dioxide, highlighting the urgency to repurpose these materials for innovative tissue regeneration solutions, offering a sustainable approach to address environmental challenges while advancing medical science. Using these discarded materials offers a promising pathway for sustainable development in regenerative medicine. This review investigates the distinctive properties of marine waste byproducts, emphasizing their capacity to be recycled effectively to contribute to the rebuilding of bone and cartilage tissue during regeneration processes. We also highlight the compatibility of these resources with biological materials such as platelet-rich plasma (PRP), stem cells, exosomes, and natural bioproducts, as well as nanoparticles (NPs) and polymers. By using the natural potential of these resources, we simultaneously address environmental challenges and promote innovative solutions in skeletal tissue engineering, initiating a new era of environmentally green biomedical research.
Collapse
Affiliation(s)
- Hoda Elkhenany
- Department of Surgery, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| | - Mariam Waleed Soliman
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Giza, Egypt
| | - Dina Atta
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Giza, Egypt
| | - Nagwa El-Badri
- Center of Excellence for Stem Cells and Regenerative Medicine (CESC), Zewail City of Science and Technology, Giza, Egypt
| |
Collapse
|
5
|
Reay SL, Marina Ferreira A, Hilkens CMU, Novakovic K. The Paradoxical Immunomodulatory Effects of Chitosan in Biomedicine. Polymers (Basel) 2024; 17:19. [PMID: 39795422 PMCID: PMC11723117 DOI: 10.3390/polym17010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/13/2025] Open
Abstract
Chitosan is widely explored in the field of biomedicine due to its abundance and reported properties, including biocompatibility, biodegradability, non-toxicity, mucoadhesion, and anti-microbial activity. Although our understanding of the immune response to chitosan has evolved, confusion remains regarding whether chitosan is a pro- or anti-inflammatory biomaterial. Tackling this knowledge gap is essential for the translation of chitosan-based biomaterials to clinical use. Herein, we provide an overview of the immune responses to chitosan, exploring the roles of endotoxin contamination and physiochemical properties in immunomodulation. Ultimately, this literature review concludes that various physiochemical properties, including molecular weight, degree of deacetylation and polydispersity, endotoxin contamination, and cellular environment, interplay in the complex process of chitosan immunomodulation, which can lead to both pro- and anti-inflammatory effects.
Collapse
Affiliation(s)
- Sophie L. Reay
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK; (A.M.F.); (K.N.)
| | - Ana Marina Ferreira
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK; (A.M.F.); (K.N.)
| | - Catharien M. U. Hilkens
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK;
| | - Katarina Novakovic
- School of Engineering, Newcastle University, Newcastle Upon Tyne NE1 7RU, UK; (A.M.F.); (K.N.)
| |
Collapse
|
6
|
Hoover AR, Liu K, Furrer C, Lam SSK, Anderson DW, Zhou Z, Yang J, Wong CF, Medcalf AD, Sun XH, Hode T, Alleruzzo L, Delawder A, Raker J, Abousleiman G, Valerio TI, Sun Y, Papin JF, Li M, Chen WR. N-Dihydrogalactochitosan Drives Conventional and Alternative Activations of STING to Synergize Type I IFN and IL-1β Productions for Antitumor Immunity. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2410079. [PMID: 39896882 PMCID: PMC11784597 DOI: 10.1002/adfm.202410079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Indexed: 02/04/2025]
Abstract
N-dihydrogalactochitosan (GC) is an immune stimulant/adjuvant. Synthesized from chitosan and galactose, GC is a new chemical entity that significantly enhances the immune-stimulating properties of its parental material, chitosan, making it a promising therapeutic agent. When used in combination with antigenic material, GC stimulates innate and adaptive antitumor and antiviral immunities. However, its mechanism has not been fully investigated. Herein we demonstrate that GC drives type I IFN activation in antigen-presenting cells (APCs). More importantly, GC drives alternative STING pathways, leading to inflammatory cell death that enhances dendritic cell (DC) activation. GC-activated DCs trigger a variety of nucleic acid sensing pattern recognition receptors (PRRs) pathways and IL-1β production via the activation of the inflammasome. In vivo, GC induces a potent response of type I IFNs and upregulates genes associated with STING signaling within the tumor microenvironment (TME). Moreover, intratumoral delivery of GC reduces the numbers of M2-like macrophages and increases M1-like macrophages residing within the TME, while subsequently increasing the number of activated DCs. Our findings demonstrate that GC acts as a multimodal immune stimulant via STING to generate a broad type I IFN response. This uniquely broad response holds therapeutic promise in generating enhanced antitumor and antiviral immunities.
Collapse
Affiliation(s)
- Ashley R. Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Coline Furrer
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | | | | | - Zhijun Zhou
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jingxuan Yang
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | - Alexandra D. Medcalf
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Xiao-Hong Sun
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Tomas Hode
- Immunophotonics, Inc., Saint Louis, MO, USA
| | | | | | | | - Ghainaa Abousleiman
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Trisha I. Valerio
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Yuanhong Sun
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - James F. Papin
- Department Pathology and Division of Comparative Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, USA
| | - Min Li
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Wei R. Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
7
|
Lin J, Zhou Y, Li C, Li B, Hao H, Tian F, Li H, Liu Z, Wang G, Shen XC, Tang R, Wang X. Hydrogel activation of Mincle receptors for tumor cell processing: A novel approach in cancer immunotherapy. Biomaterials 2024; 311:122703. [PMID: 39002516 DOI: 10.1016/j.biomaterials.2024.122703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
An obstacle in current tumor immunotherapies lies in the challenge of achieving sustained and tumor-targeting T cell immunity, impeded by the limited antigen processing and cross-presentation of tumor antigens. Here, we propose a hydrogel-based multicellular immune factory within the body that autonomously converts tumor cells into an antitumor vaccine. Within the body, the scaffold, formed by a calcium-containing chitosan hydrogel complex (ChitoCa) entraps tumor cells and attracts immune cells to establish a durable and multicellular microenvironment. Within this context, tumor cells are completely eliminated by antigen-presenting cells (APCs) and processed for cross-antigen presentation. The regulatory mechanism relies on the Mincle receptor, a cell-phagocytosis-inducing C-type lectin receptor specifically activated on ChitoCa-recruited APCs, which serves as a recognition synapse, facilitating a tenfold increase in tumor cell engulfment and subsequent elimination. The ChitoCa-induced tumor cell processing further promotes the cross-presentation of tumor antigens to prime protective CD8+ T cell responses. Therefore, the ChitoCa treatment establishes an immune niche within the tumor microenvironment, resulting in effective tumor regression either used alone or in combination with other immunotherapies. This hydrogel-induced immune factory establishes a functional organ-like multicellular colony for tumor-specific immunotherapy, paving the way for innovative strategies in cancer treatment.
Collapse
Affiliation(s)
- Jiake Lin
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Liangzhu Laboratory, Hangzhou, Zhejiang, 311113, China
| | - Yuemin Zhou
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Chen Li
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Benke Li
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Haibin Hao
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Fengchao Tian
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Huixin Li
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Zhenyu Liu
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Guangchuan Wang
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xing-Can Shen
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Liangzhu Laboratory, Hangzhou, Zhejiang, 311113, China; Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China.
| | - Xiaoyu Wang
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Liangzhu Laboratory, Hangzhou, Zhejiang, 311113, China; Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China.
| |
Collapse
|
8
|
Alameh MG, Tavakoli Naeini A, Dwivedi G, Lesage F, Buschmann MD, Lavertu M. Chitosan siRNA Nanoparticles Produce Significant Non-Toxic Functional Gene Silencing in Kidney Cortices. Polymers (Basel) 2024; 16:2547. [PMID: 39274180 PMCID: PMC11398103 DOI: 10.3390/polym16172547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/16/2024] Open
Abstract
Chitosan shows effective nucleic acid delivery. To understand the influence of chitosan's molecular weight, dose, payload, and hyaluronic acid coating on in vivo toxicity, immune stimulation, biodistribution and efficacy, precisely characterized chitosans were formulated with unmodified or chemically modified siRNA to control for innate immune stimulation. The hemocompatibility, cytokine induction, hematological and serological responses were assessed. Body weight, clinical signs, in vivo biodistribution and functional target knockdown were monitored. Hemolysis was found to be dose- and MW-dependent with the HA coating abrogating hemolysis. Compared to cationic lipid nanoparticles, uncoated and HA-coated chitosan nanoparticles did not induce immune stimulation or hematologic toxicity. Liver and kidney biomarkers remained unchanged with chitosan formulations, while high doses of cationic lipid nanoparticles led to increased transaminase levels and a decrease in body weight. Uncoated and HA-coated nanoparticles accumulated in kidneys with functional knockdown for uncoated chitosan formulations reaching 60%, suggesting potential applications in the treatment of kidney diseases.
Collapse
Affiliation(s)
- Mohamad-Gabriel Alameh
- Institute of Biomedical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montréal, QC H3T 1J4, Canada
- Department of Chemical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montreal, QC H3T 1J4, Canada
| | - Ashkan Tavakoli Naeini
- Institute of Biomedical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montréal, QC H3T 1J4, Canada
| | - Garima Dwivedi
- Institute of Biomedical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montréal, QC H3T 1J4, Canada
| | - Frederic Lesage
- Institute of Biomedical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montréal, QC H3T 1J4, Canada
- Department of Chemical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montreal, QC H3T 1J4, Canada
| | - Michael D Buschmann
- Institute of Biomedical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montréal, QC H3T 1J4, Canada
- Department of Chemical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montreal, QC H3T 1J4, Canada
| | - Marc Lavertu
- Institute of Biomedical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montréal, QC H3T 1J4, Canada
- Department of Chemical Engineering, Polytechnique Montreal, 2500 Chem. de Polytechnique, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
9
|
Yin X, Wang L, Niu Y, Xie D, Zhang Q, Xiao J, Dong L, Wang C. Unmasking Chemokine-Inducing Specificity in Oligosaccharide Biomaterial to Promote Hair Growth. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2304655. [PMID: 37567583 DOI: 10.1002/adma.202304655] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/23/2023] [Indexed: 08/13/2023]
Abstract
Hair loss affects over 50 million people worldwide with limited therapeutic options. Despite evidence highlighting the vital role of local immune cells in regulating the life cycle of hair follicles (HFs), accurate regulation of immunocytes to directly promote hair growth remains unachieved. Here, inspired by the physiological feedback in the skin immunity to suppress microbe-triggered inflammation, an oligosaccharide biomaterial with "unmasked" specific activity is developed to recruit regulatory T (Treg ) cells around HFs, leading to accelerated hair growth in mice. By processing the glucomannan polysaccharide via controllable enzymatic cleavage, a series of oligosaccharide fractions with more specific chemokine-inducing functions is obtained. Notably, a hexasaccharide-based fraction (OG6) stimulates macrophages to selectively express Treg -chemoattractant C-C Motif Chemokine Ligand 5 (CCL5) through a mannose receptor-mediated endocytosis and NOD1/2-dependent signaling, as evidenced by molecular docking, inhibition assays, and a Foxp3-reporter mouse model. Intradermal delivery of OG6 to the depilated mouse skin promotes Treg mobilization around HFs and stimulates de novo regeneration of robust hairs. This study demonstrates that unmasking precise immunomodulatory functions in oligosaccharides from their parental polysaccharide can potentially solve the long-lasting dilemma with polysaccharide biomaterials that are widely renowned for versatile activities yet high heterogeneity, opening new avenues to designing glycan-based therapeutic tools with improved specificity and safety.
Collapse
Affiliation(s)
- Xiaoyu Yin
- State Key Laboratory in Quality Research of Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, 999078, China
- State Key Laboratory in Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Lintao Wang
- State Key Laboratory in Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
| | - Yiming Niu
- State Key Laboratory in Quality Research of Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, 999078, China
| | - Daping Xie
- State Key Laboratory in Quality Research of Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, 999078, China
| | - Qingwen Zhang
- State Key Laboratory in Quality Research of Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, 999078, China
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Lei Dong
- State Key Laboratory in Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, China
- National Resource Center For Mutant Mice, Nanjing, 210023, China
| | - Chunming Wang
- State Key Laboratory in Quality Research of Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, 999078, China
- Zhuhai UM Science & Technology Research Institute, University of Macau, Hengqin, 519000, China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau SAR, 999078, China
| |
Collapse
|
10
|
Xia Y, Liu K, Wang F, Xu Z, Wang Y, Zong R, Xu Y, Li P, Deng B, Xu M, Chen G. Self-Assembled Virus-Like Particle Vaccines via Fluorophilic Interactions Enable Infection Mimicry and Immune Protection. Adv Healthc Mater 2023; 12:e2301647. [PMID: 37703498 DOI: 10.1002/adhm.202301647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 09/10/2023] [Indexed: 09/15/2023]
Abstract
Influenza epidemics persistently threaten global health. Vaccines based on virus-like particles (VLPs), which resemble the native conformation of viruses, have emerged as vaccine candidates. However, the production of VLPs via genetic engineering remains constrained by challenges such as low yields, high costs, and being time consuming. In this study, a novel VLP platform is developed that could mimic infection and confer influenza protection through fluorination-driven self-assembly. The VLPs closely mimick the key steps in viral infection including dendritic cell (DC) attachment and pH-responsive endo-lysosomal escape, which enhances DC maturation and antigen cross-presentation. It is also observed that the VLPs migrate from the injection site to the draining lymph nodes efficiently. Immunization with VLPs triggers both Th1 and Th2 cellular responses, thereby inducing an improved CD8+ T cell response along with strong antigen-specific antibody responses. In several infected mouse models, VLP vaccines ameliorate weight loss, lung virus titers, pulmonary pathologies, and confer full protection against H1N1, H6N2, H9N2, and mixed influenza viruses. Therefore, the results support the potential of VLPs as an effective influenza vaccine with improved immune potency against infection. A methodology to generate VLPs based on fluorophilic interactions, which can be a general approach for development of pathogenic VLPs, is reported.
Collapse
Affiliation(s)
- Yinhe Xia
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Kai Liu
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Fei Wang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Zhou Xu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Yuesheng Wang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Rongling Zong
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, P. R. China
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Yemin Xu
- Department of Gastroenterology, Affiliated Hospital, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Ping Li
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| | - Bin Deng
- Department of Gastroenterology, Affiliated Hospital, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Maolei Xu
- The Key Laboratory of Traditional Chinese Medicine Prescription Effect and Clinical Evaluation of State Administration of Traditional Chinese Medicine, School of Pharmacy, Binzhou Medical University, Yantai, 264003, P. R. China
| | - Gang Chen
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
- Qingdao Hospital (Qingdao Municipal Hospital), University of Health and Rehabilitation Sciences, Qingdao, 266024, P. R. China
| |
Collapse
|
11
|
Wu Y, Liang X, Mao C, Jiang Y. The Distinct Properties of Polysaccharide Nanoparticles Tune Immune Responses against mRNA Antigen via Stimulator of Interferon Genes-Mediated Autophagy and Inflammasome. ACS NANO 2023; 17:21782-21798. [PMID: 37922196 DOI: 10.1021/acsnano.3c07632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2023]
Abstract
mRNA antigens require powerful nanocarriers for efficient delivery, as well as immunomodulators for controlling their excessive immunogenicity. While lipid nanoparticles (LNPs) used in mRNA vaccines exhibited systemic toxicity, there is an urgent need for developing potential nanoparticles with strong immunoenhancing effects for mRNA antigens. Although natural polysaccharides as adjuvants assisted various types of antigens in triggering potent immune responses, they have been rarely investigated in mRNA vaccines. Here, we constructed four polysaccharide nanoparticles with different molecular weights (MWs) to deliver and protect mRNA antigens, and boosted antigen cross-presentation, DC maturation, CD4+/CD8+T cell responses and humoral immune responses. Importantly, the immunoenhancing capacities of polysaccharide nanoparticles were highly dependent on their MW properties. CS NPs with high MW initiated stimulator of interferon genes (STING)-mediated autophagy and NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome signaling, consequently possessing superior mRNA antigen-specific immune responses in vitro and in vivo. In contrast, CS NPs with low MWs induced NLRP3 signaling without STING or autophagy activation, which failed to induce robust immune responses. Therefore, it uncovered the MW-dependent immunoenhancing effects and mechanism of polysaccharide nanoparticles, providing a platform for designing potential nanosized polysaccharide immunomodulators for mRNA vaccines.
Collapse
Affiliation(s)
- Yue Wu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xiaoyu Liang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Canquan Mao
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yuhong Jiang
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
12
|
Zaiki Y, Iskandar A, Wong TW. Functionalized chitosan for cancer nano drug delivery. Biotechnol Adv 2023; 67:108200. [PMID: 37331671 DOI: 10.1016/j.biotechadv.2023.108200] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/18/2023] [Accepted: 06/11/2023] [Indexed: 06/20/2023]
Abstract
Chitosan is a biotechnological derivative of chitin receiving a widespread pharmaceutical and biomedical applications. It can be used to encapsulate and deliver cancer therapeutics with inherent pH-dependent solubility to confer drug targeting at tumour microenvironment and anti-cancer activity synergizing cancer cytotoxic drug actions. To further reduce the off-target and by-stander adverse effects of drugs, a high targeted drug delivery efficiency at the lowest possible drug doses is clinically required. The chitosan has been functionalized with covalent conjugates or complexes and processed into nanoparticles to encapsulate and control drug release, to avoid premature drug clearance, to deliver drugs passively and actively to cancer site at tissue, cell or subcellular levels, and to promote cancer cell uptake of nanoparticles through membrane permeabilization at higher specificity and scale. Nanomedicine developed using functionalized chitosan translates to significant preclinical improvements. Future challenges related to nanotoxicity, manufacturability, selection precision of conjugates and complexes as a function of cancer omics and their biological responses from administration site to cancer target need critical assessments.
Collapse
Affiliation(s)
- Yazid Zaiki
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia; Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia
| | - Athirah Iskandar
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia; Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia
| | - Tin Wui Wong
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia; Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia; Sino-Malaysia Molecular Oncology and Traditional Chinese Medicine Delivery Joint Research Centre, Medical College, Yangzhou University, 136, Jiangyang Middle Road, Yangzhou, Jiangsu Province, China; Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
13
|
Uchida S, Lau CYJ, Oba M, Miyata K. Polyplex designs for improving the stability and safety of RNA therapeutics. Adv Drug Deliv Rev 2023; 199:114972. [PMID: 37364611 DOI: 10.1016/j.addr.2023.114972] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023]
Abstract
Nanoparticle-based delivery systems have contributed to the recent clinical success of RNA therapeutics, including siRNA and mRNA. RNA delivery using polymers has several distinct properties, such as enabling RNA delivery into extra-hepatic organs, modulation of immune responses to RNA, and regulation of intracellular RNA release. However, delivery systems should overcome safety and stability issues to achieve widespread therapeutic applications. Safety concerns include direct damage to cellular components, innate and adaptive immune responses, complement activation, and interaction with surrounding molecules and cells in the blood circulation. The stability of the delivery systems should balance extracellular RNA protection and controlled intracellular RNA release, which requires optimization for each RNA species. Further, polymer designs for improving safety and stability often conflict with each other. This review covers advances in polymer-based approaches to address these issues over several years, focusing on biological understanding and design concepts for delivery systems rather than material chemistry.
Collapse
Affiliation(s)
- Satoshi Uchida
- Department of Advanced Nanomedical Engineering, Medical Research Institute, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan; Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto, 606-0823, Japan; Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan.
| | - Chun Yin Jerry Lau
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Makoto Oba
- Medical Chemistry, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 1-5 Shimogamohangi-cho, Sakyo-ku, Kyoto, 606-0823, Japan
| | - Kanjiro Miyata
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan; Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| |
Collapse
|
14
|
Eswar K, Mukherjee S, Ganesan P, Kumar Rengan A. Immunomodulatory Natural Polysaccharides: An Overview of the Mechanisms Involved. Eur Polym J 2023. [DOI: 10.1016/j.eurpolymj.2023.111935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
|
15
|
MTX-PEG-modified CG/DMMA polymeric micelles for targeted delivery of doxorubicin to induce synergistic autophagic death against triple-negative breast cancer. Breast Cancer Res 2023; 25:3. [PMID: 36635685 PMCID: PMC9837947 DOI: 10.1186/s13058-022-01599-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023] Open
Abstract
The chemotherapy of triple-negative breast cancer based on doxorubicin (DOX) regimens suffers from great challenges on toxicity and autophagy raised off-target. In this study, a conjugate methotrexate-polyethylene glycol (shorten as MTX-PEG)-modified CG/DMMA polymeric micelles were prepared to endue DOX tumor selectivity and synergistic autophagic flux interference to reduce systematic toxicity and to improve anti-tumor capacity. The micelles could effectively promote the accumulation of autophagosomes in tumor cells and interfere with the degradation process of autophagic flux, collectively inducing autophagic death of tumor cells. In vivo and in vitro experiments showed that the micelles could exert improved anti-tumor effect and specificity, as well as reduced accumulation and damage of chemotherapeutic drugs in normal organs. The potential mechanism of synergistic autophagic death exerted by the synthesized micelles in MDA-MB-231 cells has been performed by autophagic flux-related pathway.
Collapse
|
16
|
Weiss AM, Hossainy S, Rowan SJ, Hubbell JA, Esser-Kahn AP. Immunostimulatory Polymers as Adjuvants, Immunotherapies, and Delivery Systems. Macromolecules 2022; 55:6913-6937. [PMID: 36034324 PMCID: PMC9404695 DOI: 10.1021/acs.macromol.2c00854] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/16/2022] [Indexed: 12/14/2022]
Abstract
![]()
Activating innate immunity in a controlled manner is
necessary
for the development of next-generation therapeutics. Adjuvants, or
molecules that modulate the immune response, are critical components
of vaccines and immunotherapies. While small molecules and biologics
dominate the adjuvant market, emerging evidence supports the use of
immunostimulatory polymers in therapeutics. Such polymers can stabilize
and deliver cargo while stimulating the immune system by functioning
as pattern recognition receptor (PRR) agonists. At the same time,
in designing polymers that engage the immune system, it is important
to consider any unintended initiation of an immune response that results
in adverse immune-related events. Here, we highlight biologically
derived and synthetic polymer scaffolds, as well as polymer–adjuvant
systems and stimuli-responsive polymers loaded with adjuvants, that
can invoke an immune response. We present synthetic considerations
for the design of such immunostimulatory polymers, outline methods
to target their delivery, and discuss their application in therapeutics.
Finally, we conclude with our opinions on the design of next-generation
immunostimulatory polymers, new applications of immunostimulatory
polymers, and the development of improved preclinical immunocompatibility
tests for new polymers.
Collapse
Affiliation(s)
- Adam M. Weiss
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
- Department of Chemistry, University of Chicago 5735 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Samir Hossainy
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
| | - Stuart J. Rowan
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
- Department of Chemistry, University of Chicago 5735 S Ellis Ave., Chicago, Illinois 60637, United States
| | - Jeffrey A. Hubbell
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
| | - Aaron P. Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago 5640 S. Ellis Ave., Chicago, Illinois 60637, United States
| |
Collapse
|
17
|
V. Lima B, Oliveira MJ, Barbosa MA, Gonçalves RM, Castro F. Harnessing chitosan and poly-(γ-glutamic acid)-based biomaterials towards cancer immunotherapy. MATERIALS TODAY ADVANCES 2022; 15:100252. [DOI: 10.1016/j.mtadv.2022.100252] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
18
|
Kumar S, Gogoi AS, Shukla S, Trivedi M, Gulati S. Conclusion and Future Prospects of Chitosan-Based Nanocomposites. CHITOSAN-BASED NANOCOMPOSITE MATERIALS 2022:305-341. [DOI: 10.1007/978-981-19-5338-5_14] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
19
|
Chitosan coatings with distinct innate immune bioactivities differentially stimulate angiogenesis, osteogenesis and chondrogenesis in poly-caprolactone scaffolds with controlled interconnecting pore size. Bioact Mater 2021; 10:430-442. [PMID: 34901558 PMCID: PMC8636821 DOI: 10.1016/j.bioactmat.2021.09.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 12/16/2022] Open
Abstract
This study tested whether osseous integration into poly (ε-caprolactone) (PCL) bioplastic scaffolds with fully-interconnecting 155 ± 8 μm pores is enhanced by an adhesive, non-inflammatory 99% degree of deacetylation (DDA) chitosan coating (99-PCL), or further incorporation of pro-inflammatory 83% DDA chitosan microparticles (83-99-PCL) to accelerate angiogenesis. New Zealand White rabbit osteochondral knee defects were press-fit with PCL, 99-PCL, 83-99-PCL, or allowed to bleed (drill-only). Between day 1 and 6 weeks of repair, drill-only defects repaired by endochondral ossification, with an 8-fold higher bone volume fraction (BVF) versus initial defects, compared to a 2-fold (99-PCL), 1.1-fold (PCL), or 0.4-fold (83-99-PCL) change in BVF. Hematoma innate immune cells swarmed to 83-99-PCL, elicited angiogenesis throughout the pores and induced slight bone resorption. PCL and 99-PCL pores were variably filled with cartilage or avascular mesenchyme near the bone plate, or angiogenic mesenchyme into which repairing trabecular bone infiltrated up to 1 mm deep. More repair cartilage covered the 99-PCL scaffold (65%) than PCL (18%) or 83-99-PCL (0%) (p < 0.005). We report the novel finding that non-inflammatory chitosan coatings promoted cartilage infiltration into and over a bioplastic scaffold, and were compatible with trabecular bone integration. This study also revealed that in vitro osteogenesis assays have limited ability to predict osseous integration into porous scaffolds, because (1) in vivo, woven bone integrates from the leading edge of regenerating trabecular bone and not from mesenchymal cells adhering to scaffold surfaces, and (2) bioactive coatings that attract inflammatory cells induce bone resorption. Porous polycaprolactone scaffolds elicited drawn-out osteochondral wound repair. Regenerating trabecular bone only infiltrated angiogenic mesenchyme free of inflammatory cells. 83% DDA chitosan stimulated sterile inflammatory angiogenesis and trabecular bone resorption. 99% DDA chitosan coatings promoted chondrogenesis inside and over the PCL articular surface.
Collapse
|
20
|
Sun Q, Hu X, Zheng H, An Y, Qu J, Zhang Z, Khan S. Permanganate release from silica-based hollow mesoporous coagulant combined with UV for spatiotemporal enrichment and degradation of diclofenac sodium. CHEMOSPHERE 2021; 284:131306. [PMID: 34225128 DOI: 10.1016/j.chemosphere.2021.131306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 06/13/2023]
Abstract
In this work, the novel hollow mesoporous coagulant was prepared by chitosan-polydopamine coating and permanganate loading into silica nanoparticles for investigating the simultaneous enrichment and degradation of diclofenac sodium (DCFS) combined with ultraviolet irradiation. The enrichment kinetic of DCFS was explained well with pseudo-second-order model, indicating the exist of hydrogen bonding. Based on the correlation coefficients, the enriched isotherms were fitted by models which accorded with the BET > Freundlich > Langmuir sequence. The result showed that, in addition to the coagulant and DCFS, there were aromatic stackings among DCFS molecules. Due to both effects of which, the DCFS enrichment could be realized significantly in the range of pH 4.0-9.0. It was degraded at the copresence of ultraviolet and permanganate released from coagulant in acidic aqueous medium. The release mechanism was simulated through Korsmayer-Peppas model, implying case-II transport and Fickian diffusion. Additionally, Mn (V) and •OH radicals were vital in the DCFS degradation process. The coagulant could be reloaded at least ten times and that from each cycle was used directly for DCFS removal for six times without rinse process, which provided a potential application in environmental remediation.
Collapse
Affiliation(s)
- Qiang Sun
- Key Laboratory of the Three Gorges Reservoir Region's Eco-Environment, State Ministry of Education, Chongqing University, Chongqing, 400045, China
| | - Xuebin Hu
- State Key Laboratory of Coal Mine Disaster Dynamics and Control, Chongqing University, Chongqing, 400044, China.
| | - Huaili Zheng
- Key Laboratory of the Three Gorges Reservoir Region's Eco-Environment, State Ministry of Education, Chongqing University, Chongqing, 400045, China.
| | - Yanyan An
- Key Laboratory of the Three Gorges Reservoir Region's Eco-Environment, State Ministry of Education, Chongqing University, Chongqing, 400045, China
| | - Jinyao Qu
- State Key Laboratory of Coal Mine Disaster Dynamics and Control, Chongqing University, Chongqing, 400044, China
| | - Zhanmei Zhang
- School of River and Ocean Engineering, Chongqing Jiaotong University, Chongqing, 400074, PR China
| | - Sarfaraz Khan
- Key Laboratory of the Three Gorges Reservoir Region's Eco-Environment, State Ministry of Education, Chongqing University, Chongqing, 400045, China
| |
Collapse
|
21
|
Moine L, Canali MM, Porporatto C, Correa SG. Reviewing the biological activity of chitosan in the mucosa: Focus on intestinal immunity. Int J Biol Macromol 2021; 189:324-334. [PMID: 34419549 DOI: 10.1016/j.ijbiomac.2021.08.098] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/08/2021] [Accepted: 08/12/2021] [Indexed: 12/13/2022]
Abstract
Chitosan is a polymer derived from the partial deacetylation of chitin with particular characteristics, such as mucoadhesiveness, tolerability, biocompatibility and biodegradability. Biomedical uses of chitosan cover a wide spectrum of applications as dietary fiber, immunoadjuvant and regulator of the intestinal microbiota or delivery agent. Chemical modification of chitosan is feasible because its reactive amino and hydroxyl groups can be modified by a diverse array of ligands, functional groups and molecules. This gives rise to numerous derivatives that allow different formulation types influencing their activity. Considering the multiple events resulting from the interaction with mucosal tissues, chitosan is a singular candidate for strategies targeting immune stimulation (i.e., tolerance induction, vaccination). Its role as a prebiotic and probiotic carrier represents an effective option to manage intestinal dysbiosis. In the intestinal scenario where the exposure of the immune system to a wide variety of antigens is permanent, chitosan increases IgA levels and favors a tolerogenic environment, thus becoming a key ally for host homeostasis.
Collapse
Affiliation(s)
- L Moine
- Departamento de Bioquímica Clínica-Facultad de Ciencias Químicas-Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, CP: 5016 Córdoba Capital, Córdoba, Argentina
| | - M M Canali
- Université Côte d'Azur, INSERM, CNRS, IPMC, France
| | - C Porporatto
- Instituto Multidisciplinario de Investigación y Transferencia Agroalimentaria y Biotecnológica (IMITAB-CONICET), Universidad Nacional de Villa María (UNVM), Arturo Jauretche 1555, CP: 5900 Villa María, Córdoba, Argentina
| | - S G Correa
- Departamento de Bioquímica Clínica-Facultad de Ciencias Químicas-Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Universidad Nacional de Córdoba, Haya de la Torre y Medina Allende, CP: 5016 Córdoba Capital, Córdoba, Argentina.
| |
Collapse
|
22
|
Trincado V, Gala RP, Morales JO. Buccal and Sublingual Vaccines: A Review on Oral Mucosal Immunization and Delivery Systems. Vaccines (Basel) 2021; 9:vaccines9101177. [PMID: 34696284 PMCID: PMC8539688 DOI: 10.3390/vaccines9101177] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 12/13/2022] Open
Abstract
Currently, most vaccines available on the market are for parental use; however, this may not be the best option on several occasions. Mucosal routes of administration such as intranasal, sublingual, and buccal generate great interest due to the benefits they offer. These range from increasing patient compliance to inducing a more effective immune response than that achieved through conventional routes. Due to the activation of the common mucosal immune system, it is possible to generate an effective systemic and local immune response, which is not achieved through parenteral administration. Protection against pathogens that use mucosal entry routes is provided by an effective induction of mucosal immunity. Mucosal delivery systems are being developed, such as films and microneedles, which have proven to be effective, safe, and easy to administer. These systems have multiple advantages over commonly used injections, which are simple to manufacture, stable at room temperature, painless for the patient since they do not require puncture. Therefore, these delivery systems do not require to be administered by medical personnel; in fact, they could be self-administered.
Collapse
Affiliation(s)
- Valeria Trincado
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago 8380494, Chile;
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380494, Chile
- Center of New Drugs for Hypertension (CENDHY), Santiago 8380494, Chile
| | - Rikhav P. Gala
- Biotechnology Division, Center Mid-Atlantic, Fraunhofer USA, Newark, DE 19702, USA;
| | - Javier O. Morales
- Drug Delivery Laboratory, Departamento de Ciencias y Tecnología Farmacéuticas, Universidad de Chile, Santiago 8380494, Chile;
- Advanced Center for Chronic Diseases (ACCDiS), Santiago 8380494, Chile
- Center of New Drugs for Hypertension (CENDHY), Santiago 8380494, Chile
- Correspondence:
| |
Collapse
|
23
|
Aibani N, Rai R, Patel P, Cuddihy G, Wasan EK. Chitosan Nanoparticles at the Biological Interface: Implications for Drug Delivery. Pharmaceutics 2021; 13:1686. [PMID: 34683979 PMCID: PMC8540112 DOI: 10.3390/pharmaceutics13101686] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/24/2021] [Accepted: 10/07/2021] [Indexed: 12/13/2022] Open
Abstract
The unique properties of chitosan make it a useful choice for various nanoparticulate drug delivery applications. Although chitosan is biocompatible and enables cellular uptake, its interactions at cellular and systemic levels need to be studied in more depth. This review focuses on the various physical and chemical properties of chitosan that affect its performance in biological systems. We aim to analyze recent research studying interactions of chitosan nanoparticles (NPs) upon their cellular uptake and their journey through the various compartments of the cell. The positive charge of chitosan enables it to efficiently attach to cells, increasing the probability of cellular uptake. Chitosan NPs are taken up by cells via different pathways and escape endosomal degradation due to the proton sponge effect. Furthermore, we have reviewed the interaction of chitosan NPs upon in vivo administration. Chitosan NPs are immediately surrounded by a serum protein corona in systemic circulation upon intravenous administration, and their biodistribution is mainly to the liver and spleen indicating RES uptake. However, the evasion of RES system as well as the targeting ability and bioavailability of chitosan NPs can be improved by utilizing specific routes of administration and covalent modifications of surface properties. Ongoing clinical trials of chitosan formulations for therapeutic applications are paving the way for the introduction of chitosan into the pharmaceutical market and for their toxicological evaluation. Chitosan provides specific biophysical properties for effective and tunable cellular uptake and systemic delivery for a wide range of applications.
Collapse
Affiliation(s)
| | | | | | | | - Ellen K. Wasan
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, SK S7N 5E5, Canada; (N.A.); (R.R.); (P.P.); (G.C.)
| |
Collapse
|
24
|
Hou J, Yang R, Vuong I, Li F, Kong J, Mao HQ. Biomaterials strategies to balance inflammation and tenogenesis for tendon repair. Acta Biomater 2021; 130:1-16. [PMID: 34082095 DOI: 10.1016/j.actbio.2021.05.043] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 05/15/2021] [Accepted: 05/24/2021] [Indexed: 12/17/2022]
Abstract
Adult tendon tissue demonstrates a limited regenerative capacity, and the natural repair process leaves fibrotic scar tissue with inferior mechanical properties. Surgical treatment is insufficient to provide the mechanical, structural, and biochemical environment necessary to restore functional tissue. While numerous strategies including biodegradable scaffolds, bioactive factor delivery, and cell-based therapies have been investigated, most studies have focused exclusively on either suppressing inflammation or promoting tenogenesis, which includes tenocyte proliferation, ECM production, and tissue formation. New biomaterials-based approaches represent an opportunity to more effectively balance the two processes and improve regenerative outcomes from tendon injuries. Biomaterials applications that have been explored for tendon regeneration include formation of biodegradable scaffolds presenting topographical, mechanical, and/or immunomodulatory cues conducive to tendon repair; delivery of immunomodulatory or tenogenic biomolecules; and delivery of therapeutic cells such as tenocytes and stem cells. In this review, we provide the biological context for the challenges in tendon repair, discuss biomaterials approaches to modulate the immune and regenerative environment during the healing process, and consider the future development of comprehensive biomaterials-based strategies that can better restore the function of injured tendon. STATEMENT OF SIGNIFICANCE: Current strategies for tendon repair focus on suppressing inflammation or enhancing tenogenesis. Evidence indicates that regulated inflammation is beneficial to tendon healing and that excessive tissue remodeling can cause fibrosis. Thus, it is necessary to adopt an approach that balances the benefits of regulated inflammation and tenogenesis. By reviewing potential treatments involving biodegradable scaffolds, biological cues, and therapeutic cells, we contrast how each strategy promotes or suppresses specific repair steps to improve the healing outcome, and highlight the advantages of a comprehensive approach that facilitates the clearance of necrotic tissue and recruitment of cells during the inflammatory stage, followed by ECM synthesis and organization in the proliferative and remodeling stages with the goal of restoring function to the tendon.
Collapse
|
25
|
Heydari P, Kharaziha M, Varshosaz J, Javanmard SH. Current knowledge of immunomodulation strategies for chronic skin wound repair. J Biomed Mater Res B Appl Biomater 2021; 110:265-288. [PMID: 34318595 DOI: 10.1002/jbm.b.34921] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/16/2021] [Accepted: 07/18/2021] [Indexed: 12/11/2022]
Abstract
In orchestrating the wound healing process, the immune system plays a critical role. Hence, controlling the immune system to repair skin defects is an attractive approach. The highly complex immune system includes the coordinated actions of several immune cells, which can produce various inflammatory and antiinflammatory cytokines and affect the healing of skin wounds. This process can be optimized using biomaterials, bioactive molecules, and cell delivery. The present review discusses various immunomodulation strategies for supporting the healing of chronic wounds. In this regard, following the evolution of the immune system and its role in the wound healing mechanism, the interaction between the extracellular mechanism and immune cells for acceleration wound healing will be firstly investigated. Consequently, the immune-based chronic wounds will be briefly examined and the mechanism of progression, and conventional methods of their treatment are evaluated. In the following, various biomaterials-based immunomodulation strategies are introduced to stimulate and control the immune system to treat and regenerate skin defects. Other effective methods of controlling the immune system in wound healing which is the release of bioactive agents (such as antiinflammatory, antigens, and immunomodulators) and stem cell therapy at the site of injury are reviewed.
Collapse
Affiliation(s)
- Parisa Heydari
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Mahshid Kharaziha
- Department of Materials Engineering, Isfahan University of Technology, Isfahan, Iran
| | - Jaleh Varshosaz
- School of Pharmacy and Pharmaceutical Science, Isfahan University of Medical Science, Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
26
|
Turley JL, Moran HBT, McEntee CP, O'Grady K, Muñoz-Wolf N, Jin L, Follmann F, Andersen P, Andersson M, Lavelle EC. Chitin-derived polymer deacetylation regulates mitochondrial reactive oxygen species dependent cGAS-STING and NLRP3 inflammasome activation. Biomaterials 2021; 275:120961. [PMID: 34171753 DOI: 10.1016/j.biomaterials.2021.120961] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 06/02/2021] [Indexed: 01/03/2023]
Abstract
Chitosan is a cationic polysaccharide that has been evaluated as an adjuvant due to its biocompatible and biodegradable nature. The polysaccharide can enhance antibody responses and cell-mediated immunity following vaccination by injection or mucosal routes. However, the optimal polymer characteristics for activation of dendritic cells (DCs) and induction of antigen-specific cellular immune responses have not been resolved. Here, we demonstrate that only chitin-derived polymers with a high degree of deacetylation (DDA) enhance generation of mitochondrial reactive oxygen species (mtROS), leading to cGAS-STING mediated induction of type I IFN. Additionally, the capacity of the polymers to activate the NLRP3 inflammasome was strictly dependent on the degree and pattern of deacetylation and mtROS generation. Polymers with a DDA below 80% are poor adjuvants while a fully deacetylated polyglucosamine polymer is most effective as a vaccine adjuvant. Furthermore, this polyglucosamine polymer enhanced antigen-specific Th1 responses in a NLRP3 and STING-type I IFN-dependent manner. Overall these results indicate that the degree of chitin deacetylation, the acetylation pattern and its regulation of mitochondrial ROS are the key determinants of its immune enhancing effects.
Collapse
Affiliation(s)
- Joanna L Turley
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, D02R590, Ireland
| | - Hannah B T Moran
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, D02R590, Ireland
| | - Craig P McEntee
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, D02R590, Ireland
| | - Katie O'Grady
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, D02R590, Ireland
| | - Natalia Muñoz-Wolf
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, D02R590, Ireland
| | - Lei Jin
- Department of Medicine, Division of Pulmonary, Critical Care and Sleep Medicine, University of Florida, Gainesville, FL, 32610, USA
| | - Frank Follmann
- Center for Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Peter Andersen
- Center for Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institute, Copenhagen, Denmark
| | - Mats Andersson
- Division Bioscience and Materials, RISE (Research Institutes of Sweden), Forskargatan 18, 151 36, Södertälje, Sweden
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, D02R590, Ireland; Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN) & Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2, D02 PN40, Ireland.
| |
Collapse
|
27
|
Murugesan S, Scheibel T. Chitosan‐based
nanocomposites for medical applications. JOURNAL OF POLYMER SCIENCE 2021. [DOI: 10.1002/pol.20210251] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Selvakumar Murugesan
- Lehrstuhl Biomaterialien Universität Bayreuth Bayreuth Germany
- Department of Metallurgical and Materials Engineering National Institute of Technology Karnataka Mangalore India
| | - Thomas Scheibel
- Lehrstuhl Biomaterialien Universität Bayreuth Bayreuth Germany
- Bayreuther Zentrum für Kolloide und Grenzflächen (BZKG), Bayreuther Zentrum für Molekulare Biowissenschaften (BZMB), Bayreuther Materialzentrum (BayMAT), Bayerisches Polymerinstitut (BPI) University Bayreuth Bayreuth Germany
| |
Collapse
|
28
|
Li Z, Bratlie KM. The Influence of Polysaccharides-Based Material on Macrophage Phenotypes. Macromol Biosci 2021; 21:e2100031. [PMID: 33969643 DOI: 10.1002/mabi.202100031] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Indexed: 02/03/2023]
Abstract
Macrophage polarization is a key factor in determining the success of implanted tissue engineering scaffolds. Polysaccharides (derived from plants, animals, and microorganisms) are known to modulate macrophage phenotypes by recognizing cell membrane receptors. Numerous studies have developed polysaccharide-based materials into functional biomaterial substrates for tissue regeneration and pharmaceutical application due to their immunostimulatory activities and anti-inflammatory response. They are used as hydrogel substrates, surface coatings, and drug delivery carriers. In addition to their innate immunological functions, the newly endowed physical and chemical properties, including substrate modulus, pore size/porosity, surface binding chemistry, and the mole ratio of polysaccharides in hybrid materials may regulate macrophage phenotypes more precisely. Growing evidence indicates that the sulfation pattern of glycosaminoglycans and proteoglycans expressed on polarized macrophages leads to the changes in protein binding, which may alter macrophage phenotype and influence the immune response. A comprehensive understanding of how different types of polysaccharide-based materials alter macrophage phenotypic changes can be beneficial to predict transplantation/implantation outcomes. This review focuses on recent advances in promoting wound healing and balancing macrophage phenotypes using polysaccharide-based substrates/coatings and new directions to address the limitations in the current understanding of macrophage responses to polysaccharides.
Collapse
Affiliation(s)
- Zhuqing Li
- Department of Materials Science & Engineering, Iowa State University, Ames, IA, 50011, USA
| | - Kaitlin M Bratlie
- Department of Materials Science & Engineering, Iowa State University, Ames, IA, 50011, USA.,Department of Chemical & Biological Engineering, Iowa State University, Ames, IA, 50011, USA
| |
Collapse
|
29
|
Nadine S, Correia CR, Mano JF. An Immunomodulatory Miniaturized 3D Screening Platform Using Liquefied Capsules. Adv Healthc Mater 2021; 10:e2001993. [PMID: 33506631 DOI: 10.1002/adhm.202001993] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/12/2021] [Indexed: 12/11/2022]
Abstract
A critical determinant of successful clinical outcomes is the host's response to the biomaterial. Therefore, the prediction of the immunomodulatory bioperformance of biomedical devices following implantation is of utmost importance. Herein, liquefied capsules are proposed as immunomodulatory miniaturized 3D platforms for the high-content combinatorial screening of different polymers that could be used generically in scaffolds. Additionally, the confined and liquefied core of capsules affords a cell-mediated 3D assembly with bioinstructive microplatforms, allowing to study the potential synergistic effect that cells in tissue engineering therapies have on the immunological environment before implantation. As a proof-of-concept, three different polyelectrolytes, ranging in charge density and source, are used. Poly(L-lysine)-, alginate-, and chitosan-ending capsules with or without encapsulated mesenchymal stem/stromal cells (MSCs) are placed on top of a 2D culture of macrophages. Results show that chitosan-ending capsules, as well as the presence of MSCs, favor the balance of macrophage polarization toward a more regenerative profile, through the up-regulation of anti-inflammatory markers, and the release of pro-regenerative cytokines. Overall, the developed system enables the study of the immunomodulatory bioperformance of several polymers in a cost-effective and scalable fashion, while the paracrine signaling between encapsulated cells and the immunological environment can be simultaneously evaluated.
Collapse
Affiliation(s)
- Sara Nadine
- CICECO – Aveiro Institute of Materials Department of Chemistry University of Aveiro Campus Universitário de Santiago Aveiro 3810‐193 Portugal
| | - Clara R. Correia
- CICECO – Aveiro Institute of Materials Department of Chemistry University of Aveiro Campus Universitário de Santiago Aveiro 3810‐193 Portugal
| | - João F. Mano
- CICECO – Aveiro Institute of Materials Department of Chemistry University of Aveiro Campus Universitário de Santiago Aveiro 3810‐193 Portugal
| |
Collapse
|
30
|
Shi GN, Hu M, Chen C, Fu J, Shao S, Zhou Y, Wu L, Zhang T. Methotrexate enhances antigen presentation and maturation of tumour antigen-loaded dendritic cells through NLRP3 inflammasome activation: a strategy for dendritic cell-based cancer vaccine. Ther Adv Med Oncol 2021; 13:1758835920987056. [PMID: 33613696 PMCID: PMC7841859 DOI: 10.1177/1758835920987056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Dendritic cells (DCs) are antigen-presenting cells that play a pivotal role in adaptive cell-mediated immunity by priming and activating T cells against specific tumour and pathogenic antigens. Methotrexate (MTX), a folate derivative, functions as an immunoregulatory agent. However, the possible effect of MTX on tumour antigen-loaded DCs has not yet been investigated. Methods: We analysed the effect of MTX on the maturation and function of DCs along with tumour cell lysates (TCLs). Using bone marrow-derived DCs, we investigated the effect of MTX combined TCL-loaded DCs on T cells priming and proliferation. We also tested the anti-tumour immune effect on DCs when treated with MTX and/or TCL in vivo. Results: MTX combined with TCL not only enhanced DC maturation and stimulated cytokine release but also promoted CD8+ T cell activation and proliferation. The latter was associated with increased tumour antigen uptake and cross-presentation to T cells. Mechanistically, DC maturation and antigen presentation were partly modulated by NLRP3 inflammasome activation. Furthermore, immunisation of mice with MTX and TCL-pulsed DCs before a tumour challenge significantly delayed tumour onset and retarded its growth. This protective effect was due to priming of IFN-γ releasing CD8+ T cells and enhanced killing of tumour cells by cytotoxic T lymphocytes isolated from these immunised mice. Conclusion: MTX can function as a potent adjuvant in DC vaccines by increasing antigen presentation and T cell priming. Our findings provide a new strategy for the application of DC-based anti-tumour immunotherapy.
Collapse
Affiliation(s)
- Gao-Na Shi
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Min Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chengjuan Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Junmin Fu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuai Shao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yu Zhou
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lei Wu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tiantai Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Room 216, Beijing, 100050, China
| |
Collapse
|
31
|
Natural and synthetic carbohydrate-based vaccine adjuvants and their mechanisms of action. Nat Rev Chem 2021; 5:197-216. [PMID: 37117529 PMCID: PMC7829660 DOI: 10.1038/s41570-020-00244-3] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/03/2020] [Indexed: 01/31/2023]
Abstract
Modern subunit vaccines based on homogeneous antigens offer more precise targeting and improved safety compared with traditional whole-pathogen vaccines. However, they are also less immunogenic and require an adjuvant to increase the immunogenicity of the antigen and potentiate the immune response. Unfortunately, few adjuvants have sufficient potency and low enough toxicity for clinical use, highlighting the urgent need for new, potent and safe adjuvants. Notably, a number of natural and synthetic carbohydrate structures have been used as adjuvants in clinical trials, and two have recently been approved in human vaccines. However, naturally derived carbohydrate adjuvants are heterogeneous, difficult to obtain and, in some cases, unstable. In addition, their molecular mechanisms of action are generally not fully understood, partly owing to the lack of tools to elucidate their immune-potentiating effects, thus hampering the rational development of optimized adjuvants. To address these challenges, modification of the natural product structure using synthetic chemistry emerges as an attractive approach to develop well-defined, improved carbohydrate-containing adjuvants and chemical probes for mechanistic investigation. This Review describes selected examples of natural and synthetic carbohydrate-based adjuvants and their application in synthetic self-adjuvanting vaccines, while also discussing current understanding of their molecular mechanisms of action.
Collapse
|
32
|
Molecularly Distinct NLRP3 Inducers Mediate Diverse Ratios of Interleukin-1 β and Interleukin-18 from Human Monocytes. Mediators Inflamm 2020; 2020:4651090. [PMID: 33144845 PMCID: PMC7599400 DOI: 10.1155/2020/4651090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/10/2020] [Accepted: 09/29/2020] [Indexed: 11/17/2022] Open
Abstract
Inflammasomes cleave and activate interleukin- (IL-) 1β and IL-18 which have both shared and unique biological functions. IL-1β is an important mediator of the acute phase response to infections and tissue damage, whereas IL-18 takes part in activation and tailoring of the adaptive immune response. While IL-1β has served as the prototypic indicator of inflammasome activation, few studies have compared the potential differences in IL-1β and IL-18 production during inflammasome activation. Since these cytokines partake in different immune pathways, the involvement of inflammasome activity in different conditions needs to be described beyond IL-1β production alone. To address a potential heterogeneity in inflammasome functionality, ATP, chitosan, or silica oxide (SiO2) were used to induce NLRP3 inflammasome activation in THP-1 cells and the subsequent outcomes were quantified. Despite using doses of the inflammasome inducers yielding similar release of IL-1β, SiO2-stimulated cells showed a lower concentration of released IL-18 compared to ATP and chitosan. Hence, the cells stimulated with SiO2 responded with a distinctly different IL-18 : IL-1β ratio. The difference in the IL-18 : IL-1β ratio for SiO2 was constant over different doses. While all downstream responses were strictly dependent on a functional NLRP3 inflammasome, the differences did not depend on the level of gene expression, caspase-1 activity, or pyroptosis. We suggest that the NLRP3 inflammasome response should be considered a dynamic process, which can be described by taking the ratio between IL-1β and IL-18 into account and moving away from an on/off perspective of inflammasome activation.
Collapse
|
33
|
Yoncheva K, Tzankov B, Yordanov Y, Spassova I, Kovacheva D, Frosini M, Valoti M, Tzankova V. Encapsulation of doxorubicin in chitosan-alginate nanoparticles improves its stability and cytotoxicity in resistant lymphoma L5178 MDR cells. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101870] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
34
|
Rahmati M, Silva EA, Reseland JE, A Heyward C, Haugen HJ. Biological responses to physicochemical properties of biomaterial surface. Chem Soc Rev 2020; 49:5178-5224. [PMID: 32642749 DOI: 10.1039/d0cs00103a] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Biomedical scientists use chemistry-driven processes found in nature as an inspiration to design biomaterials as promising diagnostic tools, therapeutic solutions, or tissue substitutes. While substantial consideration is devoted to the design and validation of biomaterials, the nature of their interactions with the surrounding biological microenvironment is commonly neglected. This gap of knowledge could be owing to our poor understanding of biochemical signaling pathways, lack of reliable techniques for designing biomaterials with optimal physicochemical properties, and/or poor stability of biomaterial properties after implantation. The success of host responses to biomaterials, known as biocompatibility, depends on chemical principles as the root of both cell signaling pathways in the body and how the biomaterial surface is designed. Most of the current review papers have discussed chemical engineering and biological principles of designing biomaterials as separate topics, which has resulted in neglecting the main role of chemistry in this field. In this review, we discuss biocompatibility in the context of chemistry, what it is and how to assess it, while describing contributions from both biochemical cues and biomaterials as well as the means of harmonizing them. We address both biochemical signal-transduction pathways and engineering principles of designing a biomaterial with an emphasis on its surface physicochemistry. As we aim to show the role of chemistry in the crosstalk between the surface physicochemical properties and body responses, we concisely highlight the main biochemical signal-transduction pathways involved in the biocompatibility complex. Finally, we discuss the progress and challenges associated with the current strategies used for improving the chemical and physical interactions between cells and biomaterial surface.
Collapse
Affiliation(s)
- Maryam Rahmati
- Department of Biomaterials, Institute of Clinical Dentistry, University of Oslo, 0317 Oslo, Norway. h.j.haugen.odont.uio.no
| | | | | | | | | |
Collapse
|
35
|
Choi YM, Kim H, Lee SA, Lee SY, Kim BJ. A Telomerase-Derived Peptide Exerts an Anti-Hepatitis B Virus Effect via Mitochondrial DNA Stress-Dependent Type I Interferon Production. Front Immunol 2020; 11:652. [PMID: 32508804 PMCID: PMC7253625 DOI: 10.3389/fimmu.2020.00652] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 03/23/2020] [Indexed: 01/14/2023] Open
Abstract
Previously, a telomerase-derived 16-mer peptide, GV1001, developed as an anticancer vaccine, was reported to exert antiviral effects on human immunodeficiency virus or hepatitis C virus in a heat shock protein-dependent manner. Here we investigated whether GV1001 exerts antiviral effects on hepatitis B virus (HBV) and elucidated its underlying mechanisms. GV1001 inhibited HBV replication and hepatitis B surface antigen (HBsAg) secretion in a dose-dependent manner, showing synergistic antiviral effects with nucleos(t)ide analogs (NAs) including entecavir and lamivudine. This peptide also inhibited viral cccDNA and pgRNA. The intravenous GV1001 treatment of transgenic mice had anti-HBV effects. Our mechanistic studies revealed that GV1001 suppresses HBV replication by inhibiting capsid formation via type I interferon-mediated induction of heme oxygenase-1 (HO-1). GV1001 promoted the mitochondrial DNA stress-mediated release of oxidized DNA into the cytosol, resulting in IFN-I-dependent anti-HBV effects via the STING-IRF3 axis. We found that the anti-HBV effect of GV1001 was due to its ability to penetrate into the cytosol via extracellular heat shock protein, leading to phagosomal escape-mediated mtDNA stress. We demonstrated that the cell-penetrating and cytosolic localization capacity of GV1001 results in antiviral effects on HBV infections via mtDNA stress-mediated IFN-I production. Thus, GV1001, a peptide proven to be safe for human use, may be an anti-HBV drug that can be synergistically used with nucleot(s)ide analog.
Collapse
Affiliation(s)
| | | | | | | | - Bum-Joon Kim
- Department of Microbiology and Immunology, Biomedical Sciences, Liver Research Institute and Cancer Research Institute, College of Medicine, Seoul National University, Seoul, South Korea
| |
Collapse
|
36
|
Activity of Chitosan and Its Derivatives against Leishmania major and Leishmania mexicana In Vitro. Antimicrob Agents Chemother 2020; 64:AAC.01772-19. [PMID: 31871082 PMCID: PMC7038302 DOI: 10.1128/aac.01772-19] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/08/2019] [Indexed: 12/18/2022] Open
Abstract
There is an urgent need for safe, efficacious, affordable, and field-adapted drugs for the treatment of cutaneous leishmaniasis, which newly affects around 1.5 million people worldwide annually. Chitosan, a biodegradable cationic polysaccharide, has previously been reported to have antimicrobial, antileishmanial, and immunostimulatory activities. There is an urgent need for safe, efficacious, affordable, and field-adapted drugs for the treatment of cutaneous leishmaniasis, which newly affects around 1.5 million people worldwide annually. Chitosan, a biodegradable cationic polysaccharide, has previously been reported to have antimicrobial, antileishmanial, and immunostimulatory activities. We investigated the in vitro activity of chitosan and several of its derivatives and showed that the pH of the culture medium plays a critical role in antileishmanial activity of chitosan against both extracellular promastigotes and intracellular amastigotes of Leishmania major and Leishmania mexicana. Chitosan and its derivatives were approximately 7 to 20 times more active at pH 6.5 than at pH 7.5, with high-molecular-weight chitosan being the most potent. High-molecular-weight chitosan stimulated the production of nitric oxide and reactive oxygen species by uninfected and Leishmania-infected macrophages in a time- and dose-dependent manner at pH 6.5. Despite the in vitro activation of bone marrow macrophages by chitosan to produce nitric oxide and reactive oxygen species, we showed that the antileishmanial activity of chitosan was not mediated by these metabolites. Finally, we showed that rhodamine-labeled chitosan is taken up by pinocytosis and accumulates in the parasitophorous vacuole of Leishmania-infected macrophages.
Collapse
|
37
|
Sultankulov B, Berillo D, Sultankulova K, Tokay T, Saparov A. Progress in the Development of Chitosan-Based Biomaterials for Tissue Engineering and Regenerative Medicine. Biomolecules 2019; 9:E470. [PMID: 31509976 PMCID: PMC6770583 DOI: 10.3390/biom9090470] [Citation(s) in RCA: 169] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 12/16/2022] Open
Abstract
Over the last few decades, chitosan has become a good candidate for tissue engineering applications. Derived from chitin, chitosan is a unique natural polysaccharide with outstanding properties in line with excellent biodegradability, biocompatibility, and antimicrobial activity. Due to the presence of free amine groups in its backbone chain, chitosan could be further chemically modified to possess additional functional properties useful for the development of different biomaterials in regenerative medicine. In the current review, we will highlight the progress made in the development of chitosan-containing bioscaffolds, such as gels, sponges, films, and fibers, and their possible applications in tissue repair and regeneration, as well as the use of chitosan as a component for drug delivery applications.
Collapse
Affiliation(s)
- Bolat Sultankulov
- Department of Chemical Engineering, School of Engineering, Nazarbayev University, Nur-Sultan 010000, Kazakhstan
| | - Dmitriy Berillo
- Water Technology Center (WATEC) Department of Bioscience - Microbiology, Aarhus University, Aarhus 8000, Denmark
- Department of Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan
| | | | - Tursonjan Tokay
- School of Science and Technology, Nazarbayev University, Nur-Sultan 010000, Kazakhstan
| | - Arman Saparov
- School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan.
| |
Collapse
|
38
|
Vasconcelos DP, de Torre-Minguela C, Gomez AI, Águas AP, Barbosa MA, Pelegrín P, Barbosa JN. 3D chitosan scaffolds impair NLRP3 inflammasome response in macrophages. Acta Biomater 2019; 91:123-134. [PMID: 31003033 DOI: 10.1016/j.actbio.2019.04.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/11/2019] [Accepted: 04/12/2019] [Indexed: 12/21/2022]
Abstract
Chitosan (Ch) is used in different biomedical applications to promote tissue repair. However, tissue injury caused by biomaterial implantation lead to the release of danger signals that engage different inflammatory pathways on the host. Different implanted materials activate the inflammasome leading to the modulation of the immune response. Here we have studied how macroscopic biomaterials, Ch scaffolds with different chemical composition: 4% or 15% degree of acetylation (DA) modulate the activation of the NLRP3 inflammasome in vitro. For that, we assessed the NLRP3 inflammasome in bone marrow derived mouse macrophages (BMDM) and human macrophages cultured within 3D Ch scaffolds. We found that both Ch scaffolds did not trigger the NLRP3 inflammasome activation in macrophages. Furthermore, BMDMs and human macrophages cultured in both Ch scaffolds presented a reduction in the number of apoptosis-associated speck-like protein containing a caspase activating recruitment domain (ASC) specks and in IL-1β release upon classical NLRP3 inflammasome stimulation. We also found a decrease in proIL-1β in BMDMs after priming with LPS when cultured in Ch scaffolds with DA 4% DA after priming with LPS when compared to Ch scaffolds with 15% DA or to macrophages cultured in cell-culture plates. Our results shows that 3D Ch scaffolds with different DA impair NLRP3 inflammasome priming and activation. STATEMENT OF SIGNIFICANCE: In this research work we have assessed the role of the NLRP3 inflammasome in the macrophage response to 3D chitosan scaffolds with different degrees of acetylation (DA). To our knowledge this is the first work that demonstrates the modulatory capacity of 3D porous chitosan scaffolds in the NLRP3 inflammasome activation, because our results show that Ch scaffolds impair NLRP3 inflammasome assembly in macrophages. Interestingly, our results are in contrast with studies reported in the literature that indicate that chitosan is a powerful activator of the NLRP3 inflammasome in nanoscale chitosan products. Our studies that were performed in large scale chitosan scaffolds, stress out that the process of phagocytosis is pivotal in inflammasome assembly and activation, are rather important since they clearly illustrate the different role of the inflammasome in the biological response to large scale and nanoscale biomaterials.
Collapse
Affiliation(s)
- Daniela P Vasconcelos
- i3S - Instituto de Inovação e Investigação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-125 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Rua Alfredo Allen, 208, 4200-125 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Carlos de Torre-Minguela
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Hospital Clínico Universitario Virgen de la Arrixaca, Carretera Buenavista, 30120 Murcia, Spain
| | - Ana I Gomez
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Hospital Clínico Universitario Virgen de la Arrixaca, Carretera Buenavista, 30120 Murcia, Spain
| | - Artur P Águas
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; UMIB - Unit for Multidisciplinary Biomedical Research of ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Mário A Barbosa
- i3S - Instituto de Inovação e Investigação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-125 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Rua Alfredo Allen, 208, 4200-125 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Pablo Pelegrín
- Molecular Inflammation Group, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Hospital Clínico Universitario Virgen de la Arrixaca, Carretera Buenavista, 30120 Murcia, Spain.
| | - Judite N Barbosa
- i3S - Instituto de Inovação e Investigação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-125 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Rua Alfredo Allen, 208, 4200-125 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
39
|
Fermaintt CS, Sano K, Liu Z, Ishii N, Seino J, Dobbs N, Suzuki T, Fu YX, Lehrman MA, Matsuo I, Yan N. A bioactive mammalian disaccharide associated with autoimmunity activates STING-TBK1-dependent immune response. Nat Commun 2019; 10:2377. [PMID: 31147550 PMCID: PMC6542856 DOI: 10.1038/s41467-019-10319-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 04/25/2019] [Indexed: 01/07/2023] Open
Abstract
Glycans from microbial pathogens are well known pathogen-associated molecular patterns that are recognized by the host immunity; however, little is known about whether and how mammalian self-glycans activate the host immune response, especially in the context of autoimmune disease. Using biochemical fractionation and two-dimensional HPLC, we identify an abundant and bioactive free glycan, the Manβ1-4GlcNAc disaccharide in TREX1-associated autoimmune diseases. We report that both monosaccharide residues and the β1-4 linkage are critical for bioactivity of this disaccharide. We also show that Manβ1-4GlcNAc is produced by oligosaccharyltransferase hydrolysis of lipid-linked oligosaccharides in the ER lumen, followed by ENGase and mannosidase processing in the cytosol and lysosomes. Furthermore, synthetic Manβ1-4GlcNAc disaccharide stimulates a broad immune response in vitro, which is in part dependent on the STING-TBK1 pathway, and enhances antibody response in vivo. Together, our data identify Manβ1-4GlcNAc as a novel innate immune modulator associated with chronic autoimmune diseases.
Collapse
Affiliation(s)
- Charles S Fermaintt
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kanae Sano
- Division of Molecular Science, Gunma University, Maebashi, 371-8510, Japan
| | - Zhida Liu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Nozomi Ishii
- Division of Molecular Science, Gunma University, Maebashi, 371-8510, Japan
| | - Junichi Seino
- Glycometabolic Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Wako, 351-0198, Japan
| | - Nicole Dobbs
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tadashi Suzuki
- Glycometabolic Biochemistry Laboratory, RIKEN Cluster for Pioneering Research, Wako, 351-0198, Japan
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Mark A Lehrman
- Department of Pharmacology, Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Ichiro Matsuo
- Division of Molecular Science, Gunma University, Maebashi, 371-8510, Japan
| | - Nan Yan
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA. .,Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
40
|
Torosyan Y, Spece H, Goodacre N, Azarbaijani Y, Marinac-Dabic D, Kurtz SM. In silico approaches for enhancing retrieval analysis as a source for discovery of implant reactivity-related mechanisms and biomarkers. J Biomed Mater Res B Appl Biomater 2019; 108:263-271. [PMID: 31012261 DOI: 10.1002/jbm.b.34386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/26/2018] [Accepted: 03/24/2019] [Indexed: 11/06/2022]
Abstract
The ability to characterize implant debris in conjunction with corresponding immune and tissue-destructive responses renders retrieval analysis as an important tool for evaluating orthopedic devices. We applied advanced analytics and in silico approaches to illustrate the retrieval-based potential to elucidate host responses and enable discovery of corresponding biomarkers indicative of in vivo implant performance. Hip retrieval analysis was performed using variables based on immunostaining, polarized microscopy, and fretting-corrosion and oxidation analyses. Statistical analyses were performed in R. Hierarchical/k-means clustering and principal component analysis were used for data analysis and visualization. Correlation Engine (CE) and Ingenuity Pathway Analysis (IPA) were employed for in silico corroboration of putative biomarkers. Higher giant cell and histiocyte scores and positivity for CD68 and CD3 indicating infiltration with macrophages and T-cells, respectively, were detected mainly among older generation hips with higher ultra-high-molecular-weight-polyethylene loads. Our in silico analysis using pre-existing data on wear particle-induced loosening substantiated the role of CD68 in implant-induced innate responses and identified the CD68-related molecular signature that can be indicative of development of aseptic loosening and can be further corroborated for diagnostic/prognostic testing in clinical setting. Thus, this study confirmed the great potential of advanced analytics and in silico approaches for enhancing retrieval analysis applications to discovery of new biomarkers for optimizing implant-related preclinical testing and clinical management. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 108B:263-271, 2020.
Collapse
Affiliation(s)
- Yelizaveta Torosyan
- Center for Devices and Radiological Health, Office of Clinical Evidence and Analysis, Food and Drug Administration, Silver Spring, Maryland
| | - Hannah Spece
- Center for Devices and Radiological Health, Office of Clinical Evidence and Analysis, Food and Drug Administration, Silver Spring, Maryland.,Drexel University, Philadelphia, Pennsylvania
| | - Norman Goodacre
- Center for Devices and Radiological Health, Office of Clinical Evidence and Analysis, Food and Drug Administration, Silver Spring, Maryland
| | - Yasameen Azarbaijani
- Center for Devices and Radiological Health, Office of Clinical Evidence and Analysis, Food and Drug Administration, Silver Spring, Maryland
| | - Danica Marinac-Dabic
- Center for Devices and Radiological Health, Office of Clinical Evidence and Analysis, Food and Drug Administration, Silver Spring, Maryland
| | - Steven M Kurtz
- Drexel University, Philadelphia, Pennsylvania.,Exponent, Inc., Philadelphia, Pennsylvania
| |
Collapse
|
41
|
Baljon JJ, Dandy A, Wang-Bishop L, Wehbe M, Jacobson ME, Wilson JT. The efficiency of cytosolic drug delivery using pH-responsive endosomolytic polymers does not correlate with activation of the NLRP3 inflammasome. Biomater Sci 2019; 7:1888-1897. [PMID: 30843539 PMCID: PMC6478565 DOI: 10.1039/c8bm01643g] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Inefficient cytosolic delivery has limited the development of many promising biomacromolecular drugs, a long-standing challenge that has prompted extensive development of drug carriers that facilitate endosomal escape. Although many such carriers have shown considerable promise for cytosolic delivery of a diversity of therapeutics, the rupture or destabilization of endo/lysosomal membranes has also been associated with activation of the inflammasome with attendant risk of inflammation and toxicity. In this study, we investigated relationships between pH-dependent membrane destabilization, cytosolic drug delivery, and inflammasome activation using a series of well-defined poly[(ethylene glycol)-block-[(2-(dimethylamino)ethyl methacrylate)-co-(butyl methacrylate)] copolymers of variable second block composition and pH-responsive properties. We found that polymers that demonstrated the most potent membrane-destabilizing activity at early endosomal pH values in an erythrocyte hemolysis assay were most efficient at delivery of siRNA, yet tended to be associated with the least amount of NOD-like related protein 3 (NLRP3) inflammasome activation. By contrast, polymers that displayed minimal hemolysis activity and poor siRNA knockdown, and instead mediated lysosomal rupture likely due to a proton sponge mechanism, strongly induced NLPR3 inflammasome activation in a caspase- and cathepsin-dependent manner. Collectively, these findings reinforce the importance of early endosomal escape in minimizing inflammasome activation and also demonstrate the ability to tune the degree inflammasome activation via control of polymer structure with potential implications for design of vaccine adjuvants and immunotherapeutics.
Collapse
|
42
|
Zhou X, Guo L, Shi D, Duan S, Li J. Biocompatible Chitosan Nanobubbles for Ultrasound-Mediated Targeted Delivery of Doxorubicin. NANOSCALE RESEARCH LETTERS 2019; 14:24. [PMID: 30649655 PMCID: PMC6335234 DOI: 10.1186/s11671-019-2853-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 01/03/2019] [Indexed: 05/18/2023]
Abstract
Ultrasound-targeted delivery of nanobubbles (NBs) has become a promising strategy for noninvasive drug delivery. The biosafety and drug-transporting ability of NBs have been a research hotspot, especially regarding chitosan NBs due to their biocompatibility and high biosafety. Since the drug-carrying capacity of chitosan NBs and the performance of ultrasound-assisted drug delivery remain unclear, the aim of this study was to synthesize doxorubicin hydrochloride (DOX)-loaded biocompatible chitosan NBs and assess their drug delivery capacity. In this study, the size distribution of chitosan NBs was measured by dynamic light scattering, while their drug-loading capacity and ultrasound-mediated DOX release were determined by a UV spectrophotometer. In addition, a clinical ultrasound imaging system was used to evaluate the ability of chitosan NBs to achieve imaging enhancement, while the biosafety profile of free chitosan NBs was evaluated by a cytotoxicity assay in MCF-7 cells. Furthermore, NB-mediated DOX uptake and the apoptosis of Michigan Cancer Foundation-7 (MCF-7) cells were measured by flow cytometry. The results showed that the DOX-loaded NBs (DOX-NBs) exhibited excellent drug-loading ability as well as the ability to achieve ultrasound enhancement. Ultrasound (US) irradiation promoted the release of DOX from DOX-NBs in vitro. Furthermore, DOX-NBs effectively delivered DOX into mammalian cancer cells. In conclusion, biocompatible chitosan NBs are suitable for ultrasound-targeted DOX delivery and are thus a promising strategy for noninvasive and targeted drug delivery worthy of further investigation.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Department of Ultrasound, Qilu Hospital of Shandong University, West Wenhua Road, Jinan, Shandong China
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital of Shandong University, West Wenhua Road, Jinan, Shandong China
| | - Dandan Shi
- Department of Ultrasound, Qilu Hospital of Shandong University, West Wenhua Road, Jinan, Shandong China
| | - Sujuan Duan
- Department of Ultrasound, Qilu Hospital of Shandong University, West Wenhua Road, Jinan, Shandong China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital of Shandong University, West Wenhua Road, Jinan, Shandong China
| |
Collapse
|
43
|
The inflammasome in host response to biomaterials: Bridging inflammation and tissue regeneration. Acta Biomater 2019; 83:1-12. [PMID: 30273748 DOI: 10.1016/j.actbio.2018.09.056] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 09/05/2018] [Accepted: 09/27/2018] [Indexed: 12/16/2022]
Abstract
The development of new biomaterials to be used in tissue engineering applications is creating new solutions for a range of healthcare problems. The trend in biomaterials research has shifted from biocompatible "immune-evasive" biomaterials to "immune-interactive" materials that modulate the inflammatory response supporting implant integration as well as improving healing and tissue regeneration. Inflammasomes are large intracellular multiprotein complexes that are key players in host defence during innate immune responses and assemble after recognition of pathogens or danger signals. The process of biomaterial implantation causes injury to tissues that will consequently release danger signals that could be sensed by the inflammasome. There are increasing evidences that the inflammasome has a role in several inflammatory processes, from pathogen clearance to chronic inflammation or tissue repair. Thus, modulation of the inflammasome activity appears as an important target in the development of effective approaches in regenerative medicine. In this review, we discuss the main points of the current understanding on the host response to implanted biomaterials and how the paradigm of "immune-evasive" biomaterials has shifted over the last years; the significance of the inflammasome in the inflammatory response to biomaterials; and the growing idea that the immune system is of key importance in an effective tissue repair and regeneration. STATEMENT OF SIGNIFICANCE: We herein discuss the main points of the current understanding on the host response to implanted biomaterials and how the paradigm of "immune-evasive" biomaterials has shifted to "immune-interactive" over the last years; the significance of the inflammasome in the inflammatory response to biomaterials; and the growing idea that the immune system is of key importance in an effective tissue repair and regeneration, supporting the emerging concept of Regenerative Immunology. The inflammasome is a recent and central concept in immunology research. Since the beginning of this century the inflammasome is viewed as key platform of the innate immune response. We believe that, successful modulation of the inflammasome activity will become a milestone in the fields of tissue engineering and regenerative medicine.
Collapse
|
44
|
Shardlow E, Mold M, Exley C. Unraveling the enigma: elucidating the relationship between the physicochemical properties of aluminium-based adjuvants and their immunological mechanisms of action. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2018; 14:80. [PMID: 30455719 PMCID: PMC6223008 DOI: 10.1186/s13223-018-0305-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 10/26/2018] [Indexed: 01/02/2023]
Abstract
Aluminium salts are by far the most commonly used adjuvants in vaccines. There are only two aluminium salts which are used in clinically-approved vaccines, Alhydrogel® and AdjuPhos®, while the novel aluminium adjuvant used in Gardasil® is a sulphated version of the latter. We have investigated the physicochemical properties of these two aluminium adjuvants and specifically in milieus approximating to both vaccine vehicles and the composition of injection sites. Additionally we have used a monocytic cell line to establish the relationship between their physicochemical properties and their internalisation and cytotoxicity. We emphasise that aluminium adjuvants used in clinically approved vaccines are chemically and biologically dissimilar with concomitantly potentially distinct roles in vaccine-related adverse events.
Collapse
Affiliation(s)
- Emma Shardlow
- The Birchall Centre, Lennard Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG UK
| | - Matthew Mold
- The Birchall Centre, Lennard Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG UK
| | - Christopher Exley
- The Birchall Centre, Lennard Jones Laboratories, Keele University, Keele, Staffordshire ST5 5BG UK
| |
Collapse
|
45
|
Immunomodulatory properties of chitosan polymers. Biomaterials 2018; 184:1-9. [DOI: 10.1016/j.biomaterials.2018.08.054] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [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]
|
46
|
Gubaeva E, Gubaev A, Melcher RLJ, Cord-Landwehr S, Singh R, El Gueddari NE, Moerschbacher BM. 'Slipped Sandwich' Model for Chitin and Chitosan Perception in Arabidopsis. MOLECULAR PLANT-MICROBE INTERACTIONS : MPMI 2018; 31:1145-1153. [PMID: 29787346 DOI: 10.1094/mpmi-04-18-0098-r] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Chitin, a linear polymer of N-acetyl-d-glucosamine, and chitosans, fully or partially deacetylated derivatives of chitin, are known to elicit defense reactions in higher plants. We compared the ability of chitin and chitosan oligomers and polymers (chitin oligomers with degree of polymerization [DP] 3 to 8; chitosan oligomers with degree of acetylation [DA] 0 to 35% and DP 3 to 15; chitosan polymers with DA 1 to 60% and DP approximately 1,300) to elicit an oxidative burst indicative of induced defense reactions in Arabidopsis thaliana seedlings. Fully deacetylated chitosans were not able to trigger a response; elicitor activity increased with increasing DA of chitosan polymers. Partially acetylated chitosan oligomers required a minimum DP of 6 and at least four N-acetyl groups to trigger a response. Invariably, elicitation of an oxidative burst required the presence of the chitin receptor AtCERK1. Our results as well as previously published studies on chitin and chitosan perception in plants are best explained by a new general model of LysM-containing receptor complexes in which two partners form a long but off-set chitin-binding groove and are, thus, dimerized by one chitin or chitosan molecule, sharing a central GlcNAc unit with which both LysM domains interact. To verify this model and to distinguish it from earlier models, we assayed elicitor and inhibitor activities of selected partially acetylated chitosan oligomers with fully defined structures. In contrast to the initial 'continuous groove', the original 'sandwich', or the current 'sliding mode' models for the chitin/chitosan receptor, the here-proposed 'slipped sandwich' model-which builds on these earlier models and represents a consensus combination of these-is in agreement with all experimental observations.
Collapse
Affiliation(s)
- Ekaterina Gubaeva
- 1 Institute for Biology and Biotechnology of Plants, University of Münster, Schlossplatz 8, 48143 Münster, Germany; and
| | - Airat Gubaev
- 2 Institute for Physical Chemistry, University of Münster, Corrensstraße 30, 48149 Münster, Germany
| | - Rebecca L J Melcher
- 1 Institute for Biology and Biotechnology of Plants, University of Münster, Schlossplatz 8, 48143 Münster, Germany; and
| | - Stefan Cord-Landwehr
- 1 Institute for Biology and Biotechnology of Plants, University of Münster, Schlossplatz 8, 48143 Münster, Germany; and
| | - Ratna Singh
- 1 Institute for Biology and Biotechnology of Plants, University of Münster, Schlossplatz 8, 48143 Münster, Germany; and
| | - Nour Eddine El Gueddari
- 1 Institute for Biology and Biotechnology of Plants, University of Münster, Schlossplatz 8, 48143 Münster, Germany; and
| | - Bruno M Moerschbacher
- 1 Institute for Biology and Biotechnology of Plants, University of Münster, Schlossplatz 8, 48143 Münster, Germany; and
| |
Collapse
|
47
|
Lebre F, Pedroso de Lima MC, Lavelle EC, Borges O. Mechanistic study of the adjuvant effect of chitosan-aluminum nanoparticles. Int J Pharm 2018; 552:7-15. [PMID: 30244149 DOI: 10.1016/j.ijpharm.2018.09.044] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/14/2018] [Accepted: 09/18/2018] [Indexed: 12/31/2022]
Abstract
The use of tailored particle-based adjuvants constitutes a promising way to enhance antigen-specific humoral and cellular immune responses. However, a thorough understanding of the mechanisms underlying their adjuvanticity is crucial to generate more effective vaccines. We studied the ability of chitosan-aluminum nanoparticles (CH-Al NPs), which combine the immunostimulatory effects of chitosan and aluminum salts, to promote dendritic cell activation, assess their impact on innate and adaptive immune responses, and compare the results to those reported for conventional chitosan particles (CH-Na NPs). All tested CH-NP formulations were capable of modulating cytokine secretion by dendritic cells. CH-Al NPs promoted NLRP3 inflammasome activation, enhancing the release of IL-1β without significantly inhibiting Th1 and Th17 cell-polarizing cytokines, IL-12p70 or IL-23, and induced DC maturation, but did not promote pro-inflammatory cytokine production on their own. In vivo results showed that mice injected with CH-Al NPs generated a local inflammatory response comparable to that elicited by the vaccine adjuvant alum. Importantly, after subcutaneous immunization with CH-Al NPs combined with the hepatitis B surface antigen (HBsAg), mice developed antigen-specific IgG titers in serum, nasal and vaginal washes. Overall, our results established CH-Al NPs as a potential adjuvant to enhance both innate and adaptive immune responses.
Collapse
Affiliation(s)
- F Lebre
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 PN40, Ireland; Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2 D02 PN40, Ireland
| | - M C Pedroso de Lima
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2 D02 PN40, Ireland; Advanced Materials Bio-Engineering Research Centre (AMBER), Trinity College Dublin, Dublin 2 D02 PN40, Ireland
| | - O Borges
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
48
|
Bezrodnykh EA, Blagodatskikh IV, Kulikov SN, Zelenikhin PV, Yamskov IA, Tikhonov VE. Consequences of chitosan decomposition by nitrous acid: Approach to non-branched oligochitosan oxime. Carbohydr Polym 2018; 195:551-557. [DOI: 10.1016/j.carbpol.2018.05.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/20/2018] [Accepted: 05/03/2018] [Indexed: 01/28/2023]
|
49
|
Manna S, Howitz WJ, Oldenhuis NJ, Eldredge AC, Shen J, Nihesh FN, Lodoen MB, Guan Z, Esser-Kahn AP. Immunomodulation of the NLRP3 Inflammasome through Structure-Based Activator Design and Functional Regulation via Lysosomal Rupture. ACS CENTRAL SCIENCE 2018; 4:982-995. [PMID: 30159395 PMCID: PMC6107863 DOI: 10.1021/acscentsci.8b00218] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Indexed: 05/06/2023]
Abstract
The NLRP3 inflammasome plays a role in the inflammatory response to vaccines, in antimicrobial host defense, and in autoimmune diseases. However, its mechanism of action remains incompletely understood. NLRP3 has been shown to be activated by diverse stimuli including microbial toxins, ATP, particulate matter, etc. that activate multiple cellular processes. There have been two major challenges in translating inflammasome activators into controlled adjuvants. Both stem from their chemical and structural diversity. First, it is difficult to identify a minimum requirement for inflammasome activation. Second, no current activator can be tuned to generate a desired degree of activation. Thus, in order to design such immunomodulatory biomaterials, we developed a new tunable lysosomal rupture probe that leads to significant differences in inflammasome activation owing to structural changes as small as a single amino acid. Using these probes, we conduct experiments that suggest that rupturing lysosomes is a critical, initial step necessary to activate an inflammasome and that it precedes other pathways of activation. We demonstrate that each molecule differentially activates the inflammasome based solely on their degree of lysosomal rupture. We have employed this understanding of chemical control in structure-based design of immunomodulatory NLRP3 agonists on a semipredictive basis. This information may guide therapeutic interventions to prevent or mitigate lysosomal rupture and will also provide a predictive framework for dosable activation of the NLRP3 inflammasome for potential applications in vaccines and immunotherapies.
Collapse
Affiliation(s)
- Saikat Manna
- Department
of Chemistry, University of California,
Irvine, Irvine, California 92697, United States
- Institute for Molecular Engineering, University of Chicago, Chicago, Illinois 60637, United States
| | - William J. Howitz
- Department
of Chemistry, University of California,
Irvine, Irvine, California 92697, United States
| | - Nathan J. Oldenhuis
- Department
of Chemistry, University of California,
Irvine, Irvine, California 92697, United States
| | - Alexander C. Eldredge
- Department
of Chemistry, University of California,
Irvine, Irvine, California 92697, United States
| | - Jingjing Shen
- Institute for Molecular Engineering, University of Chicago, Chicago, Illinois 60637, United States
| | - Fnu Naorem Nihesh
- Department
of Chemistry, University of California,
Irvine, Irvine, California 92697, United States
- Institute for Molecular Engineering, University of Chicago, Chicago, Illinois 60637, United States
| | - Melissa B. Lodoen
- Department of Molecular Biology and Biochemistry and The Institute for Immunology, University
of California, Irvine, Irvine, California 92697, United States
| | - Zhibin Guan
- Department
of Chemistry, University of California,
Irvine, Irvine, California 92697, United States
| | - Aaron P. Esser-Kahn
- Department
of Chemistry, University of California,
Irvine, Irvine, California 92697, United States
- Institute for Molecular Engineering, University of Chicago, Chicago, Illinois 60637, United States
- E-mail:
| |
Collapse
|
50
|
Chokradjaroen C, Theeramunkong S, Yui H, Saito N, Rujiravanit R. Cytotoxicity against cancer cells of chitosan oligosaccharides prepared from chitosan powder degraded by electrical discharge plasma. Carbohydr Polym 2018; 201:20-30. [PMID: 30241811 DOI: 10.1016/j.carbpol.2018.08.037] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 08/09/2018] [Accepted: 08/09/2018] [Indexed: 02/02/2023]
Abstract
Chitosan oligosaccharides, which obtain from degradation of chitosan, possess some interesting molecular weight-dependent biological properties, especially anticancer activity. Therefore, the conversion of chitosan to chitosan oligosaccharides with specific molecular weight has been continuously investigated in order to find effective strategies that can achieve both economic feasibility and environmental concerns. In this study, a novel process was developed to heterogeneously degrade chitosan powder by highly active species generated by electrical discharge plasma in a dilute salt solution (0.02 M) without the addition of other chemicals. The degradation rate obtained from the proposed process was comparable to that obtained from some other methods with the addition of acids and oxidizing agents. Separation of the water-soluble degraded products containing chitosan oligosaccharides from the reaction solution was simply done by filtration. The obtained chitosan oligosaccharides were further evaluated for an influence of their molecular weights on cytotoxicity against cancer cells and the selectivity toward cancer and normal cells.
Collapse
Affiliation(s)
| | | | - Hiroharu Yui
- Department of Chemistry, Tokyo University of Science, Tokyo 162-8601, Japan; Water Frontier Science & Technology Research Center, Tokyo University of Science, Tokyo 162-8601, Japan
| | - Nagahiro Saito
- Department of Chemical Systems Engineering, Graduate School of Engineering, Nagoya University, Nagoya 464-8603, Japan
| | - Ratana Rujiravanit
- The Petroleum and Petrochemical College, Chulalongkorn University, Bangkok 10330, Thailand; Center of Excellence on Petrochemical and Materials Technology, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|