1
|
Zhao C, Wen S, Xu R, Wang K, Zhong Y, Huang D, Zhao B, Chen W. Oral delivery of ultra-small zwitterionic nanoparticles to overcome mucus and epithelial barriers for macrophage modulation and colitis therapy. Acta Biomater 2025; 196:399-409. [PMID: 39983856 DOI: 10.1016/j.actbio.2025.02.039] [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: 11/09/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease of the colon that poses significant therapeutic challenges due to the intestinal mucus and epithelial barriers. In this study, ultra-small zwitterionic nanoparticles (HC-CB NPs) is developed based on glutathione (GSH)-responsive hyperbranched polycarbonate to enhance the oral delivery of drugs and overcome these physiological barriers. HC-CB NPs demonstrate high colloidal stability across a wide range of pH environments and physiological fluids, preventing premature drug release within the gastrointestinal tract. The ultra-small sized HC-CB NPs demonstrate minimal mucin adsorption and effectively penetrate through the mucus layer, and the zwitterion surface further facilitate epithelial barrier crossing via the proton-assisted amino acid transporter 1 (PAT1) pathway. HC-CB NPs mediate enhanced macrophage uptake via monocarboxylate transporters (MCTs) pathway and ultimately improved therapy efficacy on colitis. The in vivo results reveal that FK506-loaded HC-CB NPs (HC-CB NPs@FK506) significantly reduce inflammatory markers (TNF-α, IL-6) and myeloperoxidase (MPO) levels, while promoting epithelial integrity by increasing E-cadherin expression. This study offers a promising approach to overcoming intestinal barriers in oral UC treatment, offering biocompatibility and potential for clinical translation. STATEMENT OF SIGNIFICANCE: Ulcerative colitis (UC) is a chronic inflammatory disease of the colon that poses significant therapeutic challenges due to the intestinal mucus and epithelial barriers. This study explores an oral UC therapy using ultra-small zwitterionic nanoparticles (HC-CB NPs) constructed from GSH-responsive hyperbranched polycarbonate. Compared to existing strategies, HC-CB NPs demonstrate minimal mucin adsorption and effectively penetrate through the mucus layer, and the zwitterion surface further facilitate epithelial barrier crossing via the proton-assisted amino acid transporter 1 (PAT1) pathway. Additionally, HC-CB NPs mediate enhanced macrophage uptake via monocarboxylate transporters (MCTs) pathway, resulting in improved therapeutic efficacy. These findings underscore the potential of HC-CB NPs as a transformative platform for overcoming intestinal barriers in UC treatment.
Collapse
Affiliation(s)
- Changshun Zhao
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Suchen Wen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Rui Xu
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Ke Wang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Yinan Zhong
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Dechun Huang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China; Engineering Research Center for Smart Pharmaceutical Manufacturing Technologies, Ministry of Education, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Bingbing Zhao
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Wei Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China; Engineering Research Center for Smart Pharmaceutical Manufacturing Technologies, Ministry of Education, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
2
|
Gao C, Yang Z, Song R, Sheng H, Zhu L. Nanotechnology-based drug delivery system for targeted therapy of ulcerative colitis from traditional Chinese medicine: A review. Int J Pharm 2025; 673:125375. [PMID: 39965734 DOI: 10.1016/j.ijpharm.2025.125375] [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: 12/01/2024] [Revised: 01/27/2025] [Accepted: 02/15/2025] [Indexed: 02/20/2025]
Abstract
Ulcerative colitis (UC) is a chronic autoimmune disease and seriously affects the normal life of patients. Conventional therapeutic drugs are difficult to meet clinical needs. Traditional Chinese medicine (TCM) ingredients could effectively alleviate the symptoms of UC by anti-inflammatory, anti-oxidative, regulating the gut microbiota, and repairing the colonic epithelial barrier, but their low solubility and bioavailability severely limit their clinical application. Nano-drug delivery systems (NDDS) combined with TCM ingredients is a promising option for treating UC, and they could significantly enhance the stability, solubility, and bioavailability of TCM ingredients. The review describes the anti-UC mechanisms of TCM ingredients, systematically summarizes various kinds of NDDS for TCM ingredients according to different routes of administration, and highlights the advantages of NDDS for TCM ingredients in the treatmentof UC. In addition, we discuss the limitations of existing NDDS for TCM ingredients and the development direction in the future. This review will provide a basis for the future development of anti-UC NDDS for TCM ingredients.
Collapse
Affiliation(s)
- Chengcheng Gao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zerun Yang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ruirui Song
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
3
|
Xu Y, Michalowski CB, Koehler J, Darwish T, Guccio N, Alcaino C, Domingues I, Zhang W, Marotti V, Van Hul M, Paone P, Koutsoviti M, Boyd BJ, Drucker DJ, Cani PD, Reimann F, Gribble FM, Beloqui A. Smart control lipid-based nanocarriers for fine-tuning gut hormone secretion. SCIENCE ADVANCES 2024; 10:eadq9909. [PMID: 39671480 PMCID: PMC11641013 DOI: 10.1126/sciadv.adq9909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 11/08/2024] [Indexed: 12/15/2024]
Abstract
Modulating the endogenous stores of gastrointestinal hormones is considered a promising strategy to mimic gut endocrine function, improving metabolic dysfunction. Here, we exploit mouse and human knock-in and knockout intestinal organoids and show that agents used as commercial lipid excipients can activate nutrient-sensitive receptors on enteroendocrine cells (EECs) and, when formulated as lipid nanocarriers, can bestow biological effects through the release of GLP-1, GIP, and PYY from K and L cells. Studies in wild-type, dysglycemic, and gut Gcg knockout mice demonstrated that the effect exerted by lipid nanocarriers could be modulated by varying the excipients (e.g., nature and quantities), the formulation methodology, and their physiochemical properties (e.g., size and composition). This study demonstrates the therapeutic potential of using nanotechnology to modulate release of multiple endogenous hormones from the enteroendocrine system through a patient-friendly, inexpensive, and noninvasive manner.
Collapse
Affiliation(s)
- Yining Xu
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université catholique de Louvain, 1200 Brussels, Belgium
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
- Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Cécilia Bohns Michalowski
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université catholique de Louvain, 1200 Brussels, Belgium
| | - Jackie Koehler
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 2J7, Canada
| | - Tamana Darwish
- Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Nunzio Guccio
- Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Constanza Alcaino
- Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Inês Domingues
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université catholique de Louvain, 1200 Brussels, Belgium
| | - Wunan Zhang
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université catholique de Louvain, 1200 Brussels, Belgium
| | - Valentina Marotti
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université catholique de Louvain, 1200 Brussels, Belgium
| | - Matthias Van Hul
- Louvain Drug Research Institute, Metabolism and Nutrition Group, Université catholique de Louvain, 1200 Brussels, Belgium
| | - Paola Paone
- Louvain Drug Research Institute, Metabolism and Nutrition Group, Université catholique de Louvain, 1200 Brussels, Belgium
| | - Melitini Koutsoviti
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
- Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Ben J. Boyd
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen Ø, Denmark
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Daniel J. Drucker
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 2J7, Canada
| | - Patrice D. Cani
- Louvain Drug Research Institute, Metabolism and Nutrition Group, Université catholique de Louvain, 1200 Brussels, Belgium
- WEL Research Institute, Avenue Pasteur, 6, 1300 Wavre, Belgium
- Institute of Experimental and Clinical Research (IREC), Université catholique de Louvain, 1200 Brussels, Belgium
| | - Frank Reimann
- Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Fiona M. Gribble
- Institute of Metabolic Science, Addenbrooke’s Hospital, University of Cambridge, Hills Road, Cambridge, CB2 0QQ, UK
| | - Ana Beloqui
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université catholique de Louvain, 1200 Brussels, Belgium
- WEL Research Institute, Avenue Pasteur, 6, 1300 Wavre, Belgium
| |
Collapse
|
4
|
Li S, Wu T, Wu J, Chen W, Zhang D. Recognizing the biological barriers and pathophysiological characteristics of the gastrointestinal tract for the design and application of nanotherapeutics. Drug Deliv 2024; 31:2415580. [PMID: 39404464 PMCID: PMC11485891 DOI: 10.1080/10717544.2024.2415580] [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: 05/07/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
The gastrointestinal tract (GIT) is an important and complex system by which humans to digest food and absorb nutrients. The GIT is vulnerable to diseases, which may led to discomfort or even death in humans. Therapeutics for GIT disease treatment face multiple biological barriers, which significantly decrease the efficacy of therapeutics. Recognizing the biological barriers and pathophysiological characteristics of GIT may be helpful to design innovative therapeutics. Nanotherapeutics, which have special targeting and controlled therapeutic release profiles, have been widely used for the treatment of GIT diseases. Herein, we provide a comprehensive review of the biological barrier and pathophysiological characteristics of GIT, which may aid in the design of promising nanotherapeutics for GIT disease treatment. Furthermore, several typical diseases of the upper and lower digestive tracts, such as Helicobacter pylori infection and inflammatory bowel disease, were selected to investigate the application of nanotherapeutics for GIT disease treatment.
Collapse
Affiliation(s)
- Shan Li
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Gastroenterology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Army Medical University (Third Military Medical University), Shigatse, Tibet Autonomous Region, China
| | - Tianyu Wu
- Department of Gastroenterology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jingfeng Wu
- Department of Gastroenterology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wensheng Chen
- Department of Gastroenterology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Dinglin Zhang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
5
|
Liang W, Zhang W, Tian J, Zhang X, Lv X, Qu A, Chen J, Wu Z. Advances in carbohydrate-based nanoparticles for targeted therapy of inflammatory bowel diseases: A review. Int J Biol Macromol 2024; 281:136392. [PMID: 39423983 DOI: 10.1016/j.ijbiomac.2024.136392] [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: 05/18/2024] [Revised: 09/13/2024] [Accepted: 10/05/2024] [Indexed: 10/21/2024]
Abstract
The incidence of inflammatory bowel disease (IBD), a chronic gastrointestinal disorder, is rapidly increasing worldwide. Unfortunately, the current therapies for IBD are often hindered by premature drug release and undesirable side effects. With the advancement of nanotechnology, the innovative targeted nanotherapeutics are explored to ensure the accurate delivery of drugs to specific sites in the colon, thereby reducing side effects and improving the efficacy of oral administration. The emphasis of this review is to summarize the potential pathogenesis of IBD and highlight recent breakthroughs in carbohydrate-based nanoparticles for IBD treatment, including their construction, release mechanism, potential targeting ability, and their therapeutic efficacy. Specifically, we summarize the latest knowledge regarding environmental-responsive nano-systems and active targeted nanoparticles. The environmental-responsive drug delivery systems crafted with carbohydrates or other biological macromolecules like chitosan and sodium alginate, exhibit a remarkable capacity to enhance the accumulation of therapeutic drugs in the inflamed regions of the digestive tract. Active targeting strategies improve the specificity and accuracy of oral drug delivery to the colon by modifying carbohydrates such as hyaluronic acid and mannose onto nanocarriers. Finally, we discuss the challenges and provide insight into the future perspectives of colon-targeted delivery systems for IBD treatment.
Collapse
Affiliation(s)
- Wenjing Liang
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Wen Zhang
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China; Key Laboratory of Low Carbon Cold Chain for Agricultural Products, Ministry of Agriculture and Rural Affairs, China.
| | - Jiayi Tian
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Xinping Zhang
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Xinyi Lv
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Ao Qu
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China
| | - Jinyu Chen
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China; Key Laboratory of Low Carbon Cold Chain for Agricultural Products, Ministry of Agriculture and Rural Affairs, China
| | - Zijian Wu
- Tianjin Key Laboratory of Food Science and Biotechnology, School of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin 300134, China; Key Laboratory of Low Carbon Cold Chain for Agricultural Products, Ministry of Agriculture and Rural Affairs, China.
| |
Collapse
|
6
|
Wu H, Shi C, Li Q, Wang L, Wang R, Chen F, Li R, Guo X, Chen Y, She J. Oral Administration of Bioactive Nanoparticulates for Inflammatory Bowel Disease Therapy by Mitigating Oxidative Stress and Restoring Intestinal Microbiota Homeostasis. Mol Pharm 2024. [PMID: 39462848 DOI: 10.1021/acs.molpharmaceut.4c00499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The management of inflammatory bowel disease (IBD) continues to pose significant challenges due to the absence of curative therapies and a high rate of recurrence. Therefore, it is imperative to explore novel approaches to enhance the efficacy of IBD therapy. Herein, a bioactive nanoparticulate s is tailored designed to achieve a "Pull-Push" approach for efficient and safe IBD treatment by integrating reactive oxygen species (ROS) scavenging (Pull) with anti-inflammatory agent delivery (Push) in the inflammatory microenvironment. The multifunctional nanomedicine, designated MON-PAMAM@SASP, is developed through the encapsulation of sulfasalazine (SASP), a widely utilized clinical drug for the treatment of IBD, within cationic diselenide-bridged mesoporous organosilica nanoparticles (MONs) that possess significant antioxidant properties. Herein, poly(amidoamine) (PAMAM) endows the original MONs with positive charge characteristics. The MON-PAMAM@SASP not only displays the remarkable capability of neutralizing ROS to ameliorates intestinal damage, but also achieves controllable release of SASP to mitigate intestinal inflammation. Consequently, this nanomedicine effectively mitigates IBD by colitis in mouse models, and our current research has not identified any significant drug toxicity. Beyond regulating inflammatory microenvironment in intestine, treatment with MON-PAMAM@SASP results in increased richness and restores intestinal microbiota homeostasis, thereby mitigating IBD to a certain extent. Together, our work provides a highly versatile "Pull-Push" approach for IBD management and encourages the development of similar nanomedicine to treating multiple inflammatory diseases of gastrointestinal tract.
Collapse
Affiliation(s)
- Hong Wu
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- The Third Department of General Surgery, Xi'an Daxing Hospital Affiliated to Yan'an University, Xi'an 710016, China
| | - Chengxin Shi
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Qixin Li
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Lizhao Wang
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Ruochen Wang
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Fangman Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Ruizhe Li
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Xiaolong Guo
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Yinnan Chen
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| | - Junjun She
- Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi 710049, China
| |
Collapse
|
7
|
Liu X, Dong Y, Wang C, Guo Z. Application of chitosan as nano carrier in the treatment of inflammatory bowel disease. Int J Biol Macromol 2024; 278:134899. [PMID: 39187100 DOI: 10.1016/j.ijbiomac.2024.134899] [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: 12/11/2023] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 08/28/2024]
Abstract
Inflammatory bowel disease (IBD), encompassing ulcerative colitis (UC) and Crohn's disease (CD), is characterized by persistent and recurrent gastrointestinal inflammation. Conventional IBD therapies often involve the use of antibiotics, NSAIDs, biological agents, and immunomodulators. While these medications can mitigate acute inflammatory symptoms, their long-term efficacy is frequently compromised due to cumulative toxic effects. In recent years, significant attention has shifted toward nanoparticle (NP)-based therapies as potential alternatives for IBD management. Various drug delivery strategies, including those targeting microbiota interactions, ligand-receptor binding, pH sensitivity, biodegradability, pressure response, and specific charge and size parameters, have been explored and optimized in animal studies. This review provides a comprehensive overview of the current landscape of chitosan NP-mediated drug delivery systems for IBD treatment. Additionally, it will discuss the prevailing challenges and propose future research directions to advance chitosan NP-based therapeutic strategies for IBD.
Collapse
Affiliation(s)
- Xiaoming Liu
- Department of Gastroenterology, Huaihe Hospital of Henan University, 115 Ximen Street, Kaifeng 475000, Henan, China
| | - Yunrui Dong
- Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, China
| | - Chenyu Wang
- Department of General Surgery, Huaihe Hospital of Henan University, 115 Ximen Street, Kaifeng 475000, Henan, China
| | - Zhiguo Guo
- Department of Gastroenterology, Suzhou Hospital of Anhui Medical University (Suzhou Municipal Hospital of Anhui Province), No.616 Bianyangsan Road, Suzhou 234000, Anhui, China.
| |
Collapse
|
8
|
Gardey E, Cseresnyes Z, Sobotta FH, Eberhardt J, Haziri D, Grunert PC, Kuchenbrod MT, Gruschwitz FV, Hoeppener S, Schumann M, Gaßler N, Figge MT, Stallmach A, Brendel JC. Selective Uptake Into Inflamed Human Intestinal Tissue and Immune Cell Targeting by Wormlike Polymer Micelles. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306482. [PMID: 38109123 DOI: 10.1002/smll.202306482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 11/10/2023] [Indexed: 12/19/2023]
Abstract
Inflammatory bowel disease (IBD) has become a globally prevalent chronic disease with no causal therapeutic options. Targeted drug delivery systems with selectivity for inflamed areas in the gastrointestinal tract promise to reduce severe drug-related side effects. By creating three distinct nanostructures (vesicles, spherical, and wormlike micelles) from the same amphiphilic block copolymer poly(butyl acrylate)-block-poly(ethylene oxide) (PBA-b-PEO), the effect of nanoparticle shape on human mucosal penetration is systematically identified. An Ussing chamber technique is established to perform the ex vivo experiments on human colonic biopsies, demonstrating that the shape of polymeric nanostructures represents a rarely addressed key to tissue selectivity required for efficient IBD treatment. Wormlike micelles specifically enter inflamed mucosa from patients with IBD, but no significant uptake is observed in healthy tissue. Spheres (≈25 nm) and vesicles (≈120 nm) enter either both normal and inflamed tissue types or do not penetrate any tissue. According to quantitative image analysis, the wormlike nanoparticles localize mainly within immune cells, facilitating specific targeting, which is crucial for further increasing the efficacy of IBD treatment. These findings therefore demonstrate the untapped potential of wormlike nanoparticles not only to selectively target the inflamed human mucosa, but also to target key pro-inflammatory cells.
Collapse
Affiliation(s)
- Elena Gardey
- Department of Internal Medicine IV (Gastroenterology, Hepatology, Infectious Diseases and Central Endoscopy), Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Zoltan Cseresnyes
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology Hans Knöll Institute (HKI), Beutenbergstraße 11a, 07745, Jena, Germany
| | - Fabian H Sobotta
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743, Jena, Germany
- Department of Chemical Engineering and Chemistry & Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5612 AZ, the Netherlands
| | - Juliane Eberhardt
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743, Jena, Germany
| | - Drilon Haziri
- Department of Internal Medicine IV (Gastroenterology, Hepatology, Infectious Diseases and Central Endoscopy), Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Philip C Grunert
- Department of Internal Medicine IV (Gastroenterology, Hepatology, Infectious Diseases and Central Endoscopy), Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
| | - Maren T Kuchenbrod
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743, Jena, Germany
| | - Franka V Gruschwitz
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743, Jena, Germany
| | - Stephanie Hoeppener
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Michael Schumann
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité-University Medicine, Hindenburgdamm 30, 12200, Berlin, Germany
| | - Nikolaus Gaßler
- Jena University Hospital, Section of Pathology, Institute of Forensic Medicine, Friedrich Schiller University Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Marc T Figge
- Applied Systems Biology, Leibniz Institute for Natural Product Research and Infection Biology Hans Knöll Institute (HKI), Beutenbergstraße 11a, 07745, Jena, Germany
- Institute of Microbiology, Faculty of Biological Sciences, Friedrich Schiller University Jena, Neugasse 25, 07743, Jena, Germany
| | - Andreas Stallmach
- Department of Internal Medicine IV (Gastroenterology, Hepatology, Infectious Diseases and Central Endoscopy), Jena University Hospital, Am Klinikum 1, 07747, Jena, Germany
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
| | - Johannes C Brendel
- Jena Center for Soft Matter (JCSM), Friedrich Schiller University Jena, Philosophenweg 7, 07743, Jena, Germany
- Laboratory of Organic and Macromolecular Chemistry (IOMC), Friedrich Schiller University Jena, Humboldtstraße 10, 07743, Jena, Germany
| |
Collapse
|
9
|
Hu D, Tian T, Ren Q, Han S, Li Z, Deng Y, Lu Z, Zhang L. Novel biomimetic peptide-loaded chitosan nanoparticles improve dentin bonding via promoting dentin remineralization and inhibiting endogenous matrix metalloproteinases. Dent Mater 2024; 40:160-172. [PMID: 37951748 DOI: 10.1016/j.dental.2023.11.003] [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: 06/22/2023] [Revised: 10/25/2023] [Accepted: 11/04/2023] [Indexed: 11/14/2023]
Abstract
OBJECTIVE This study aims to synthesize novel chitosan nanoparticles loaded with an amelogenin-derived peptide QP5 (TMC-QP5/NPs), investigate their remineralization capability and inhibitory effects on endogenous matrix metalloproteinases (MMPs), and evaluate the dentin bonding properties of remineralized dentin regulated by TMC-QP5/NPs. METHODS TMC-QP5/NPs were prepared by ionic crosslinking method and characterized by dynamic light scattering method, scanning electron microscopy, transmission electron microscope, atomic force microscope, Fourier transform infrared spectroscopy, and differential scanning calorimetry. The encapsulation and loading efficiency of TMC-QP5/NPs and the release of QP5 were examined. To evaluate the remineralization capability of TMC-QP5/NPs, the mechanical properties, and the changes in structure and composition of differently conditioned dentin were characterized. The MMPs inhibitory effects of TMC-QP5/NPs were explored by MMP Activity Assay and in-situ zymography. The dentin bonding performance was detected by interfacial microleakage and microshear bond strength (μSBS). RESULTS TMC-QP5/NPs were successfully synthesized, with uniform size, good stability and biosafety. The encapsulation and loading efficiency of TMC-QP5/NPs was respectively 69.63 ± 2.22% and 13.21 ± 0.73%, with a sustained release of QP5. TMC-QP5/NPs could induce mineral deposits on demineralized collagen fibers and partial occlusion of dentin tubules, and recover the surface microhardness of dentin, showing better remineralization effects than QP5. Besides, TMC-QP5/NPs significantly inhibited the endogenous MMPs activity. The remineralized dentin induced by TMC-QP5/NPs exhibited less interfacial microleakage and higher μSBS, greatly improved dentin bonding. SIGNIFICANCE This novel peptide-loaded chitosan nanoparticles improved resin-dentin bonding by promoting dentin remineralization and inactivating MMPs, suggesting a promising strategy for optimizing dentin adhesive restorations.
Collapse
Affiliation(s)
- Die Hu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Tian Tian
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qian Ren
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Sili Han
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhongcheng Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yudi Deng
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ziqian Lu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Linglin Zhang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
10
|
Li Q, Lin L, Zhang C, Zhang H, Ma Y, Qian H, Chen XL, Wang X. The progression of inorganic nanoparticles and natural products for inflammatory bowel disease. J Nanobiotechnology 2024; 22:17. [PMID: 38172992 PMCID: PMC10763270 DOI: 10.1186/s12951-023-02246-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024] Open
Abstract
There is a growing body of evidence indicating a close association between inflammatory bowel disease (IBD) and disrupted intestinal homeostasis. Excessive production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), along with an increase in M1 proinflammatory macrophage infiltration during the activation of intestinal inflammation, plays a pivotal role in disrupting intestinal homeostasis in IBD. The overabundance of ROS/RNS can cause intestinal tissue damage and the disruption of crucial gut proteins, which ultimately compromises the integrity of the intestinal barrier. The proliferation of M1 macrophages contributes to an exaggerated immune response, further compromising the intestinal immune barrier. Currently, intestinal nanomaterials have gained widespread attention in the context of IBD due to their notable characteristics, including the ability to specifically target regions of interest, clear excess ROS/RNS, and mimic biological enzymes. In this review, we initially elucidated the gut microenvironment in IBD. Subsequently, we delineate therapeutic strategies involving two distinct types of nanomedicine, namely inorganic nanoparticles and natural product nanomaterials. Finally, we present a comprehensive overview of the promising prospects associated with the application of nanomedicine in future clinical settings for the treatment of IBD (graphic abstract). Different classes of nanomedicine are used to treat IBD. This review primarily elucidates the current etiology of inflammatory bowel disease and explores two prominent nanomaterial-based therapeutic approaches. First, it aims to eliminate excessive reactive oxygen species and reactive nitrogen species. Second, they focus on modulating the polarization of inflammatory macrophages and reducing the proportion of pro-inflammatory macrophages. Additionally, this article delves into the treatment of inflammatory bowel disease using inorganic metal nanomaterials and natural product nanomaterials.
Collapse
Affiliation(s)
- Qingrong Li
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Liting Lin
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Cong Zhang
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China
| | - Hengguo Zhang
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Yan Ma
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China
| | - Haisheng Qian
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China.
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| | - Xianwen Wang
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China.
| |
Collapse
|
11
|
Hu S, Zhao R, Xu Y, Gu Z, Zhu B, Hu J. Orally-administered nanomedicine systems targeting colon inflammation for the treatment of inflammatory bowel disease: latest advances. J Mater Chem B 2023; 12:13-38. [PMID: 38018424 DOI: 10.1039/d3tb02302h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic and idiopathic condition that results in inflammation of the gastrointestinal tract, leading to conditions such as ulcerative colitis and Crohn's disease. Commonly used treatments for IBD include anti-inflammatory drugs, immunosuppressants, and antibiotics. Fecal microbiota transplantation is also being explored as a potential treatment method; however, these drugs may lead to systemic side effects. Oral administration is preferred for IBD treatment, but accurately locating the inflamed area in the colon is challenging due to multiple physiological barriers. Nanoparticle drug delivery systems possess unique physicochemical properties that enable precise delivery to the target site for IBD treatment, exploiting the increased permeability and retention effect of inflamed intestines. The first part of this review comprehensively introduces the pathophysiological environment of IBD, covering the gastrointestinal pH, various enzymes in the pathway, transport time, intestinal mucus, intestinal epithelium, intestinal immune cells, and intestinal microbiota. The second part focuses on the latest advances in the mechanism and strategies of targeted delivery using oral nanoparticle drug delivery systems for colitis-related fields. Finally, we present challenges and potential directions for future IBD treatment with the assistance of nanotechnology.
Collapse
Affiliation(s)
- Shumeng Hu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, P. R. China.
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
| | - Runan Zhao
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- College of Biosystems Engineering and Food Science, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, P. R. China
| | - Yu Xu
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, P. R. China
| | - Zelin Gu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, P. R. China.
| | - Beiwei Zhu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, 130118, P. R. China.
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, P. R. China
| | - Jiangning Hu
- State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, 116034, P. R. China.
- School of Food Science and Technology, Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, 116034, P. R. China
| |
Collapse
|
12
|
Zhang Y, Liu L, Wang T, Mao C, Shan P, Lau CS, Li Z, Guo W, Wang W. Reactive Oxygen Species-Responsive Polymeric Prodrug Nanoparticles for Selective and Effective Treatment of Inflammatory Diseases. Adv Healthc Mater 2023; 12:e2301394. [PMID: 37540810 PMCID: PMC11468797 DOI: 10.1002/adhm.202301394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/11/2023] [Indexed: 08/06/2023]
Abstract
It is challenging to manage inflammatory diseases using traditional anti-inflammatory drugs due to their limited efficacy and systemic side effects, which are a result of their lack of selectivity, poor stability, and low solubility. Herein, it reports the development of a novel nanoparticle system, called ROS-CA-NPs, which is formed using polymer-cinnamaldehyde (CA) conjugates and is responsive to reactive oxygen species (ROS). ROS-CA-NPs exhibit excellent drug stability, tissue selectivity, and controlled drug release upon oxidative stress activation. Using mouse models of chronic rheumatoid arthritis and acute ulcerative colitis, this study demonstrates that the systemic administration of ROS-CA-NPs results in their accumulation at inflamed lesions and leads to greater therapeutic efficacy compared to traditional drugs. Furthermore, ROS-CA-NPs present excellent biocompatibility. The findings suggest that ROS-CA-NPs have the potential to be developed as safe and effective nanotherapeutic agents for a broad range of inflammatory diseases.
Collapse
Affiliation(s)
- Yaming Zhang
- State Key Laboratory of Pharmaceutical Biotechnology & Dr. Li Dak-Sum Research Centre & Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Lu Liu
- State Key Laboratory of Pharmaceutical Biotechnology & Dr. Li Dak-Sum Research Centre & Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Tianyi Wang
- State Key Laboratory of Pharmaceutical Biotechnology & Dr. Li Dak-Sum Research Centre & Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Cong Mao
- Department of Minimally Invasive Interventional Radiology, State Key Laboratory of Respiratory Disease, School of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Pengfei Shan
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, 325027, China
| | - Chak Sing Lau
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Zhongyu Li
- College of Chemistry and Materials Engineering, Wenzhou University, Wenzhou, 325027, China
| | - Weisheng Guo
- Department of Minimally Invasive Interventional Radiology, State Key Laboratory of Respiratory Disease, School of Biomedical Engineering & The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, China
| | - Weiping Wang
- State Key Laboratory of Pharmaceutical Biotechnology & Dr. Li Dak-Sum Research Centre & Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
13
|
Wang Y, Li Z, Bao Y, Cui H, Li J, Song B, Wang M, Li H, Cui X, Chen Y, Chen W, Yang S, Yang Y, Jin Z, Si X, Li B. Colon-targeted delivery of polyphenols: construction principles, targeting mechanisms and evaluation methods. Crit Rev Food Sci Nutr 2023; 65:64-86. [PMID: 37823723 DOI: 10.1080/10408398.2023.2266842] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Polyphenols have received considerable attention for their promotive effects on colonic health. However, polyphenols are mostly sensitive to harsh gastrointestinal environments, thus, must be protected. It is necessary to design and develop a colon-targeted delivery system to improve the stability, colon-targeting and bioavailability of polyphenols. This paper mainly introduces research on colon-targeted controlled release of polyphenols. The physiological features affecting the dissolution, release and absorption of polyphenol-loaded delivery systems in the colon are first discussed. Simultaneously, the types of colon-targeted carriers with different release mechanisms are described, and colon-targeting assessment models that have been studied so far and their advantages and limitations are summarized. Based on the current research on polyphenols colon-targeting, outlook and reflections are proposed, with the goal of inspiring strategic development of new colon-targeted therapeutics to ensure that the polyphenols reach the colon with complete bioactivity.
Collapse
Affiliation(s)
- Yidi Wang
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Zhiying Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Yiwen Bao
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Huijun Cui
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Jiaxin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Baoge Song
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Mengzhu Wang
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Haikun Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Xingyue Cui
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Yi Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, China
| | - Wei Chen
- Faculty of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - Shufang Yang
- Zhejiang Lanmei Technology Co., Ltd, Zhu-ji City, Zhejiang Province, China
| | - Yiyun Yang
- Zhejiang Lanmei Technology Co., Ltd, Zhu-ji City, Zhejiang Province, China
| | - Zhufeng Jin
- Zhejiang Lanmei Technology Co., Ltd, Zhu-ji City, Zhejiang Province, China
| | - Xu Si
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| | - Bin Li
- College of Food Science, Shenyang Agricultural University, Shenyang, Liaoning, China
| |
Collapse
|
14
|
Zhang C, Zeng F, Fan Z, He Z, Tai L, Peng Q, Zhang Y, Chao Z, Jiang W, Jia L, Han L. An oral polyphenol host-guest nanoparticle for targeted therapy of inflammatory bowel disease. Acta Biomater 2023; 169:422-433. [PMID: 37597682 DOI: 10.1016/j.actbio.2023.08.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 08/04/2023] [Accepted: 08/11/2023] [Indexed: 08/21/2023]
Abstract
Inflammatory bowel disease (IBD) is a global public health challenge that affects millions of people. Current medical treatments for IBD are not fully effective and may cause undesirable side effects on patients. Thus, there is an urgent need for safe, simple, and efficacious strategies to treat IBD in clinical settings. Here, we develop an oral polyphenol nanoparticle (PDT) by assembling dexamethasone sodium phosphate (DSP)-loaded poly-β-cyclodextrin with tannic acid via host-guest interactions for treating IBD. This one-step assembly process is rapid (within 10 s), reproducible, and free of harmful chemical agents, which can facilitate its clinical translation. PDT is negatively charged due to the three components, which enable it to specifically target the positively charged inflamed colonic mucosa through electrostatic attraction, thus localizing the drug at the inflamed site to reduce systemic exposure and side effects. Furthermore, PDT exhibits a strong reactive oxygen species (ROS)-scavenging ability derived from the tannic acid component, which can alleviate ROS-mediated inflammatory responses and ameliorate IBD symptoms. Compared with free DSP, PDT demonstrates sustained DSP release behavior in vitro and in vivo, as well as enhanced therapeutic efficacy in a colitis mouse model. These results suggest that PDT might be a potential therapeutic agent for the treatment of IBD. Moreover, this facile polyphenol host-guest assembly strategy may provide a promising drug-delivery platform for treating various diseases STATEMENT OF SIGNIFICANCE: To develop safe and effective treatments for inflammatory bowel disease (IBD), we have designed an oral polyphenol nanoparticle (PDT) using the host-guest assembly of dexamethasone sodium phosphate (DSP)-loaded poly-β-cyclodextrin with tannic acid. Through in vitro and in vivo experiments, PDT has demonstrated remarkable inflammation-targeting, ROS-scavenging, and anti-inflammatory properties, along with sustained release of DSP. Moreover, in an IBD mouse model, PDT has shown significantly improved therapeutic efficacy compared to free DSP. The host-guest assembly strategy employed for PDT is noteworthy for its rapidity, reproducibility, and safety due to the absence of harmful chemicals, holding great promise for designing a diverse range of nanomedicines customized for treating various diseases.
Collapse
Affiliation(s)
- Chong Zhang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, PR China
| | - Fen Zeng
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, PR China
| | - Zhengyang Fan
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, PR China
| | - Zhen He
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, PR China
| | - Liang Tai
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, PR China
| | - Qiang Peng
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, PR China
| | - Yixin Zhang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, PR China
| | - Zhenhua Chao
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, PR China
| | - Wenning Jiang
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, PR China
| | - Lingyun Jia
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, PR China.
| | - Lulu Han
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian 116023, PR China.
| |
Collapse
|
15
|
Di Vincenzo F, Yadid Y, Petito V, Emoli V, Masi L, Gerovska D, Araúzo-Bravo MJ, Gasbarrini A, Regenberg B, Scaldaferri F. Circular and Circulating DNA in Inflammatory Bowel Disease: From Pathogenesis to Potential Molecular Therapies. Cells 2023; 12:1953. [PMID: 37566032 PMCID: PMC10417561 DOI: 10.3390/cells12151953] [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: 06/19/2023] [Revised: 07/15/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023] Open
Abstract
Inflammatory bowel diseases (IBD), including Crohn's Disease (CD) and Ulcerative Colitis (UC) are chronic multifactorial disorders which affect the gastrointestinal tract with variable extent. Despite extensive research, their etiology and exact pathogenesis are still unknown. Cell-free DNAs (cfDNAs) are defined as any DNA fragments which are free from the origin cell and able to circulate into the bloodstream with or without microvescicles. CfDNAs are now being increasingly studied in different human diseases, like cancer or inflammatory diseases. However, to date it is unclear how IBD etiology is linked to cfDNAs in plasma. Extrachromosomal circular DNA (eccDNA) are non-plasmidic, nuclear, circular and closed DNA molecules found in all eukaryotes tested. CfDNAs appear to play an important role in autoimmune diseases, inflammatory processes, and cancer; recently, interest has also grown in IBD, and their role in the pathogenesis of IBD has been suggested. We now suggest that eccDNAs also play a role in IBD. In this review, we have comprehensively collected available knowledge in literature regarding cfDNA, eccDNA, and structures involving them such as neutrophil extracellular traps and exosomes, and their role in IBD. Finally, we focused on old and novel potential molecular therapies and drug delivery systems, such as nanoparticles, for IBD treatment.
Collapse
Affiliation(s)
- Federica Di Vincenzo
- IBD Unit, Centro di Malattie dell’Apparato Digerente (CeMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.D.V.); (L.M.); (A.G.); (F.S.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (Y.Y.); (V.E.)
| | - Ylenia Yadid
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (Y.Y.); (V.E.)
| | - Valentina Petito
- IBD Unit, Centro di Malattie dell’Apparato Digerente (CeMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.D.V.); (L.M.); (A.G.); (F.S.)
| | - Valeria Emoli
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (Y.Y.); (V.E.)
| | - Letizia Masi
- IBD Unit, Centro di Malattie dell’Apparato Digerente (CeMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.D.V.); (L.M.); (A.G.); (F.S.)
| | - Daniela Gerovska
- Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, Calle Doctor Begiristain s/n, 20014 San Sebastian, Spain; (D.G.); (M.J.A.-B.)
| | - Marcos Jesus Araúzo-Bravo
- Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, Calle Doctor Begiristain s/n, 20014 San Sebastian, Spain; (D.G.); (M.J.A.-B.)
- IKERBASQUE, Basque Foundation for Science, Calle María Díaz Harokoa 3, 48013 Bilbao, Spain
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Antonio Gasbarrini
- IBD Unit, Centro di Malattie dell’Apparato Digerente (CeMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.D.V.); (L.M.); (A.G.); (F.S.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (Y.Y.); (V.E.)
| | - Birgitte Regenberg
- Section for Ecology and Evolution, Department of Biology, University of Copenhagen, Universitetsparken 13, Room 426, DK-2100 Copenhagen, Denmark;
| | - Franco Scaldaferri
- IBD Unit, Centro di Malattie dell’Apparato Digerente (CeMAD), Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy; (F.D.V.); (L.M.); (A.G.); (F.S.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (Y.Y.); (V.E.)
| |
Collapse
|
16
|
Liu D, Wei M, Yan W, Xie H, Sun Y, Yuan B, Jin Y. Potential applications of drug delivery technologies against radiation enteritis. Expert Opin Drug Deliv 2023; 20:435-455. [PMID: 36809906 DOI: 10.1080/17425247.2023.2183948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023]
Abstract
INTRODUCTION The incidence of abdominal tumors, such as colorectal and prostate cancers, continually increases. Radiation therapy is widely applied in the clinical treatment of patients with abdominal/pelvic cancers, but it often unfortunately causes radiation enteritis (RE) involving the intestine, colon, and rectum. However, there is a lack of suitable treatment options for effective prevention and treatment of RE. AREAS COVERED Conventional clinical drugs for preventing and treating RE are usually applied by enemas and oral administration. Innovative gut-targeted drug delivery systems including hydrogels, microspheres, and nanoparticles are proposed to improve the prevention and curation of RE. EXPERT OPINION The prevention and treatment of RE have not attracted sufficient attention in the clinical practice, especially compared to the treatment of tumors, although RE takes patients great pains. Drug delivery to the pathological sites of RE is a huge challenge. The short retention and weak targeting of conventional drug delivery systems affect the therapeutic efficiency of anti-RE drugs. Novel drug delivery systems including hydrogels, microspheres, and nanoparticles can allow drugs long-term retention in the gut and targeting the inflammation sites to alleviate radiation-induced injury.
Collapse
Affiliation(s)
- Dongdong Liu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Meng Wei
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Wenrui Yan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Hua Xie
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yingbao Sun
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Bochuan Yuan
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| | - Yiguang Jin
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, China
| |
Collapse
|
17
|
Elahi E, Ali ME, Zimmermann J, Getts DR, Müller M, Lamprecht A. Immune Modifying Effect of Drug Free Biodegradable Nanoparticles on Disease Course of Experimental Autoimmune Neuritis. Pharmaceutics 2022; 14:2410. [PMID: 36365228 PMCID: PMC9695102 DOI: 10.3390/pharmaceutics14112410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/21/2023] Open
Abstract
Guillain-Barre syndrome (GBS) is an autoimmune disease of demyelination and inflammation of peripheral nerves. Current treatments are limited to plasma exchange and intravenous immunoglobulins. Cargo-free nanoparticles (NPs) have been evaluated here for their therapeutic benefit on the disease course of experimental autoimmune neuritis (EAN), mimicking the human GBS. NPs prepared from poly-lactic co-glycolic acid (PLGA) with variable size and surface charge (i.e., 500 nm vs. 130 nm, polyvinyl alcohol (PVA) vs. sodium cholate), were intravenously administered in before- or early-onset treatment schedules in a rat EAN model. NP treatment mitigated distinctly the clinical severity of EAN as compared to the P2-peptide control group (P2) in all treatments and reduced the trafficking of inflammatory monocytes at inflammatory loci and diverted them towards the spleen. Therapeutic treatment with NPs reduced the expression of proinflammatory markers (CD68 (P2: 34.8 ± 6.6 vs. NP: 11.9 ± 2.3), IL-1β (P2: 18.3 ± 0.8 vs. NP: 5.8 ± 2.2), TNF-α (P2: 23.5 ± 3.7 vs. NP: 8.3 ± 1.7) and elevated the expression levels of anti-inflammatory markers CD163 (P2: 19.7 ± 3.0 vs. NP: 41.1 ± 6.5; all for NP-PVA of 130 nm; relative to healthy control). These results highlight the therapeutic potential of such cargo-free NPs in treating EAN, which would be easily translatable into clinical use due to their well-known low-toxicity profile.
Collapse
Affiliation(s)
- Ehsan Elahi
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, 53127 Bonn, Germany
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Gerhard-Domagk Str. 3, 53121 Bonn, Germany
| | - Mohamed Ehab Ali
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Gerhard-Domagk Str. 3, 53121 Bonn, Germany
| | - Julian Zimmermann
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, 53127 Bonn, Germany
| | - Daniel R. Getts
- Myeloid Therapeutics, 300 Technology Sq., Suite 203, Cambridge, MA 02139, USA
| | - Marcus Müller
- Department of Neurology, University Clinic Bonn, Campus Venusberg 1, 53127 Bonn, Germany
| | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Gerhard-Domagk Str. 3, 53121 Bonn, Germany
| |
Collapse
|
18
|
Das R, Bhasarkar J, Rastogi A, Saxena R, Bal DK. Artificial neural network-based pore size prediction of alginate gel scaffold for targeted drug delivery. Neural Comput Appl 2022. [DOI: 10.1007/s00521-022-07958-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
19
|
Fei Y, Ma Y, Zhang H, Li H, Feng G, Fang J. Nanotechnology for research and treatment of the intestine. J Nanobiotechnology 2022; 20:430. [PMID: 36175955 PMCID: PMC9523975 DOI: 10.1186/s12951-022-01517-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
The establishment of intestinal in vitro models is crucial for elucidating intestinal cell-microbe intrinsic connections and interaction mechanisms to advance normalized intestinal diagnosis and precision therapy. This review discusses the application of nanomaterials in mucosal therapy and mechanism research in combination with the study of nanoscaffold in vitro models of the gut. By reviewing the original properties of nanomaterials synthesized by different physicochemical principles and modifying the original properties, the contribution of nanomaterials to solving the problems of short survival period, low cell differentiation rate, and poor reduction ability in traditional intestinal models is explored. According to nanomaterials’ different diagnostic mediators and therapeutic targets, the current diagnostic principles in inflammatory bowel disease, intestinal cancer, and other diseases are summarized inductively. In addition, the mechanism of action of nanomedicines in repairing mucosa, inhibiting inflammation, and alleviating the disease process is also discussed. Through such systematic elaboration, it offers a basis for nanomaterials to help advance in vitro research on the intestine and provide precision treatments in the clinic.
Collapse
Affiliation(s)
- Yanquan Fei
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, Hunan, China
| | - Yong Ma
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, Hunan, China
| | - Huaizu Zhang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, Hunan, China
| | - Hao Li
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Guangfu Feng
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, Hunan, China.
| | - Jun Fang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, Hunan, China.
| |
Collapse
|
20
|
Li DF, Yang MF, Xu HM, Zhu MZ, Zhang Y, Tian CM, Nie YQ, Wang JY, Liang YJ, Yao J, Wang LS. Nanoparticles for oral delivery: targeted therapy for inflammatory bowel disease. J Mater Chem B 2022; 10:5853-5872. [PMID: 35876136 DOI: 10.1039/d2tb01190e] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
As a group of chronic and idiopathic gastrointestinal (GI) disorders, inflammatory bowel disease (IBD) is characterized by recurrent intestinal mucosal inflammation. Oral administration is critical for the treatment of IBD. Unfortunately, it is difficult to target the bowel located in the GI tract due to multiple physical barriers. The unique physicochemical properties of nanoparticle-based drug delivery systems (DDSs) and their enhanced permeability and retention effects in the inflamed bowel, render nanomedicines to be used to implement precise drug delivery at diseased sites in IBD therapy. In this review, we described the pathophysiological features of IBD, and designed strategies to exploit these features for intestinal targeting. In addition, we introduced the types of currently developed nano-targeted carriers, including synthetic nanoparticle-based and emerging naturally derived nanoparticles (e.g., extracellular vesicles and plant-derived nanoparticles). Moreover, recent developments in targeted oral nanoparticles for IBD therapy were also highlighted. Finally, we presented challenges associated with nanotechnology and potential directions for future IBD treatment.
Collapse
Affiliation(s)
- De-Feng Li
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen 518020, Guangdong, China.
| | - Mei-Feng Yang
- Department of Hematology, Yantian District People's Hospital, Shenzhen 518020, Guangdong, China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510030, China
| | - Min-Zheng Zhu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510030, China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou 516000, Guangdong, China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Yu-Qiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510030, China
| | - Jian-Yao Wang
- Department of General Surgery, Shenzhen Children's Hospital, No. 7019, Yitian Road, Futian District, Shenzhen 518026, Guangdong, China.
| | - Yu-Jie Liang
- Shenzhen Kangning Hospital, No. 1080, Cuizu Road, Luohu District, Shenzhen 518020, Guangdong, China.
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen 518020, Guangdong, China.
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (the Second Clinical Medical College, Jinan University, the First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen 518020, Guangdong, China.
| |
Collapse
|
21
|
Pathological features-based targeted delivery strategies in IBD therapy: A mini review. Biomed Pharmacother 2022; 151:113079. [PMID: 35605297 DOI: 10.1016/j.biopha.2022.113079] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 11/22/2022] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is characterized by a complex and dysfunctional immune response. Currently, IBD is incurable, and patients with IBD often need to take drugs for life. However, as the traditional systemic treatment strategies for IBD do not target the site of inflammation, only limited efficacy can be obtained from them. Moreover, the possibility of serious side effects stemming from the systemic administration or redistribution of drugs in the body is high when conventional drug formulations are used. Therefore, a targeted drug-delivery system for IBD should be considered. Based on the pathological features related to IBD, the new targeted drug-delivery strategy can directly transfer the drug to the inflammatory site, thus enhancing the accumulation of the drugs and reducing side effects. This article reviews the pathological features of IBD and the application of the IBD-targeted delivery system based on different pathological features, and discusses the challenges and new prospects in this field.
Collapse
|
22
|
Yasmin F, Najeeb H, Shaikh S, Hasanain M, Naeem U, Moeed A, Koritala T, Hasan S, Surani S. Novel drug delivery systems for inflammatory bowel disease. World J Gastroenterol 2022; 28:1922-1933. [PMID: 35664964 PMCID: PMC9150062 DOI: 10.3748/wjg.v28.i18.1922] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/22/2022] [Accepted: 03/26/2022] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic illness characterized by relapsing inflammation of the intestines. The disorder is stratified according to the severity and is marked by its two main phenotypical representations: Ulcerative colitis and Crohn's disease. Pathogenesis of the disease is ambiguous and is expected to have interactivity between genetic disposition, environmental factors such as bacterial agents, and dysregulated immune response. Treatment for IBD aims to reduce symptom extent and severity and halt disease progression. The mainstay drugs have been 5-aminosalicylates (5-ASAs), corticosteroids, and immunosuppressive agents. Parenteral, oral and rectal routes are the conventional methods of drug delivery, and among all, oral administration is most widely adopted. However, problems of systematic drug reactions and low specificity in delivering drugs to the inflamed sites have emerged with these regular routes of delivery. Novel drug delivery systems have been introduced to overcome several therapeutic obstacles and for localized drug delivery to target tissues. Enteric-coated microneedle pills, various nano-drug delivery techniques, prodrug systems, lipid-based vesicular systems, hybrid drug delivery systems, and biologic drug delivery systems constitute some of these novel methods. Microneedles are painless, they dislodge their content at the affected site, and their release can be prolonged. Recombinant bacteria such as genetically engineered Lactococcus Lactis and eukaryotic cells, including GM immune cells and red blood cells as nanoparticle carriers, can be plausible delivery methods when evaluating biologic systems. Nano-particle drug delivery systems consisting of various techniques are also employed as nanoparticles can penetrate through inflamed regions and adhere to the thick mucus of the diseased site. Prodrug systems such as 5-ASAs formulations or their derivatives are effective in reducing colonic damage. Liposomes can be modified with both hydrophilic and lipophilic particles and act as lipid-based vesicular systems, while hybrid drug delivery systems containing an internal nanoparticle section for loading drugs are potential routes too. Leukosomes are also considered as possible carrier systems, and results from mouse models have revealed that they control anti- and pro-inflammatory molecules.
Collapse
Affiliation(s)
- Farah Yasmin
- Department of Medicine, Dow University of Health Science, Karachi 74200, Pakistan
| | - Hala Najeeb
- Department of Medicine, Dow University of Health Science, Karachi 74200, Pakistan
| | - Shehryar Shaikh
- Department of Medicine, Dow OJha University Hospital, Karachi 74200, Pakistan
| | - Muhammad Hasanain
- Department of Medicine, Dow University of Health Science, Karachi 74200, Pakistan
| | - Unaiza Naeem
- Department of Medicine, Dow University of Health Science, Karachi 74200, Pakistan
| | - Abdul Moeed
- Department of Medicine, Dow University of Health Science, Karachi 74200, Pakistan
| | - Thoyaja Koritala
- Department of Medicine, Mayo Clinic Health System, Mankato, MN 56001, United States
| | - Syedadeel Hasan
- Department of Medicine, University of Louisville, Louisville, KY 40292, United States
| | - Salim Surani
- Department of Medicine, Texas A&M University, College Station, TX 77843, United States
- Department of Anesthesiology, Mayo Clinic, Rochester, MN 55901, United States
| |
Collapse
|
23
|
Kędzierska M, Drabczyk A, Jamroży M, Kudłacik-Kramarczyk S, Głąb M, Tyliszczak B, Bańkosz W, Potemski P. The Synthesis Methodology and Characterization of Nanogold-Coated Fe 3O 4 Magnetic Nanoparticles. MATERIALS 2022; 15:ma15093383. [PMID: 35591718 PMCID: PMC9105358 DOI: 10.3390/ma15093383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 04/29/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023]
Abstract
Core-shell nanostructures are widely used in many fields, including medicine and the related areas. An example of such structures are nanogold-shelled Fe3O4 magnetic nanoparticles. Systems consisting of a magnetic core and a shell made from nanogold show unique optical and magnetic properties. Thus, it is essential to develop the methodology of their preparation. Here, we report the synthesis methodology of Fe3O4@Au developed so as to limit their agglomeration and increase their stability. For this purpose, the impact of the reaction environment was verified. The properties of the particles were characterized via UV-Vis spectrophotometry, dynamic light scattering (DLS), X-ray diffraction (XRD), and Scanning Electron Microscopy-Energy Dispersive X-ray analysis (SEM-EDS technique). Moreover, biological investigations, including determining the cytotoxicity of the particles towards murine fibroblasts and the pro-inflammatory activity were also performed. It was demonstrated that the application of an oil and water reaction environment leads to the preparation of the particles with lower polydispersity, whose agglomerates’ disintegration is 24 times faster than the disintegration of nanoparticle agglomerates formed as a result of the reaction performed in a water environment. Importantly, developed Fe3O4@Au nanoparticles showed no pro-inflammatory activity regardless of their concentration and the reaction environment applied during their synthesis and the viability of cell lines incubated for 24 h with the particle suspensions was at least 92.88%. Thus, the developed synthesis methodology of the particles as well as performed investigations confirmed a great application potential of developed materials for biomedical purposes.
Collapse
Affiliation(s)
- Magdalena Kędzierska
- Department of Chemotherapy, Medical University of Lodz, WWCOiT Copernicus Hospital, 90-001 Lodz, Poland; (M.K.); (P.P.)
| | - Anna Drabczyk
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland; (M.G.); (B.T.)
- Correspondence: (A.D.); (M.J.); (S.K.-K.)
| | - Mateusz Jamroży
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland; (M.G.); (B.T.)
- Correspondence: (A.D.); (M.J.); (S.K.-K.)
| | - Sonia Kudłacik-Kramarczyk
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland; (M.G.); (B.T.)
- Correspondence: (A.D.); (M.J.); (S.K.-K.)
| | - Magdalena Głąb
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland; (M.G.); (B.T.)
| | - Bożena Tyliszczak
- Department of Materials Science, Faculty of Materials Engineering and Physics, Cracow University of Technology, 37 Jana Pawła II Av., 31-864 Krakow, Poland; (M.G.); (B.T.)
| | - Wojciech Bańkosz
- Department of Automation and Robotics, Faculty of Electrical and Computer Engineering, Cracow University of Technology, 24 Warszawska St., 31-155 Krakow, Poland;
| | - Piotr Potemski
- Department of Chemotherapy, Medical University of Lodz, WWCOiT Copernicus Hospital, 90-001 Lodz, Poland; (M.K.); (P.P.)
| |
Collapse
|
24
|
Tie S, Xiang S, Chen Y, Qiao F, Cui W, Su W, Tan M. Facile synthesis of food-grade and size-controlled nanocarriers based on self-assembly of procyanidins and phycocyanin. Food Funct 2022; 13:4023-4031. [PMID: 35315469 DOI: 10.1039/d1fo04222j] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanocarriers provide the possibility to overcome the low solubility, poor stability, and low bioavailability of functional factors. However, most nanocarriers do not directly participate in the corresponding effects of functional factors, such as treating inflammatory bowel disease but lack the means to control their size accurately. Herein, nanocarriers were prepared by a one-pot method, using food-grade antioxidant procyanidins, vanillin, and phycocyanin as raw materials. The strategy involved the Mannich reaction among the phenolic hydroxyl groups of procyanidins, the aldehyde groups of vanillin, and the amino groups of phycocyanin. The obtained nanocarriers displayed controllable sizes ranging from 130 to 750 nm, showing good antioxidant capacity in scavenging free radicals and were biocompatible to Caco-2 cells and RAW 264.7 macrophages. Nanocarriers also exhibited an inhibitory effect on cell damage induced by acrylamide and H2O2. Moreover, the designed nanocarriers could be used for delivering active ingredients such as lutein, which showed a uniform spherical distribution, high encapsulation efficiency, and good biocompatibility. This work provides a facile synthesis method to prepare food-grade nanocarriers with functional properties, which can be potentially used in the delivery of functional factors.
Collapse
Affiliation(s)
- Shanshan Tie
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China. .,National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Siyuan Xiang
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China. .,National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Yannan Chen
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China. .,National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Fengzhi Qiao
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China. .,National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Weina Cui
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China. .,National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Wentao Su
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China. .,National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Mingqian Tan
- Academy of Food Interdisciplinary Science, School of Food Science and Technology, Dalian Polytechnic University, Qinggongyuan1, Ganjingzi District, Dalian 116034, Liaoning, China. .,National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, Liaoning, China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| |
Collapse
|
25
|
Tu Z, Zhong Y, Hu H, Shao D, Haag R, Schirner M, Lee J, Sullenger B, Leong KW. Design of therapeutic biomaterials to control inflammation. NATURE REVIEWS. MATERIALS 2022; 7:557-574. [PMID: 35251702 PMCID: PMC8884103 DOI: 10.1038/s41578-022-00426-z] [Citation(s) in RCA: 245] [Impact Index Per Article: 81.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 01/12/2022] [Indexed: 05/03/2023]
Abstract
Inflammation plays an important role in the response to danger signals arising from damage to our body and in restoring homeostasis. Dysregulated inflammatory responses occur in many diseases, including cancer, sepsis and autoimmunity. The efficacy of anti-inflammatory drugs, developed for the treatment of dysregulated inflammation, can be potentiated using biomaterials, by improving the bioavailability of drugs and by reducing side effects. In this Review, we first outline key elements and stages of the inflammatory environment and then discuss the design of biomaterials for different anti-inflammatory therapeutic strategies. Biomaterials can be engineered to scavenge danger signals, such as reactive oxygen and nitrogen species and cell-free DNA, in the early stages of inflammation. Materials can also be designed to prevent adhesive interactions of leukocytes and endothelial cells that initiate inflammatory responses. Furthermore, nanoscale platforms can deliver anti-inflammatory agents to inflammation sites. We conclude by discussing the challenges and opportunities for biomaterial innovations in addressing inflammation.
Collapse
Affiliation(s)
- Zhaoxu Tu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- Department of Biomedical Engineering, Columbia University, New York, NY USA
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Yiling Zhong
- Department of Biomedical Engineering, Columbia University, New York, NY USA
- School of Chemistry, University of New South Wales, Sydney, New South Wales Australia
| | - Hanze Hu
- Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Dan Shao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, China
- Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, China
| | - Rainer Haag
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin, Germany
| | - Michael Schirner
- Institut für Chemie und Biochemie, Freie Universität Berlin, Berlin, Germany
| | - Jaewoo Lee
- School of Medicine, Duke University, Durham, NC USA
| | | | - Kam W. Leong
- Department of Biomedical Engineering, Columbia University, New York, NY USA
- Department of Systems Biology, Columbia University, New York, NY USA
| |
Collapse
|
26
|
Huang F, Jiang X, Sallam MA, Zhang X, He W. A Nanocrystal Platform Based on Metal-Phenolic Network Wrapping for Drug Solubilization. AAPS PharmSciTech 2022; 23:76. [PMID: 35178657 DOI: 10.1208/s12249-022-02220-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/17/2022] [Indexed: 11/30/2022] Open
Abstract
The preparation of drugs into nanocrystals represents a practical pharmaceutical technology to solubilize poorly water-soluble drugs and enhance bioavailability. However, commonly used stabilizers in nanocrystals like polymers and surfactants are frequently inefficient and cannot stabilize nanocrystals for an expected time. This study reports an exquisite platform for nanocrystal production based on a metal-phenolic network (MPN). MPN-wrapped nanocrystal particles (MPN-NPs) were fabricated through an anti-solvent precipitation method using tannic acid and FeIII or AlIII as coupling agents and characterized by dynamic light scattering, transmission electron microscope, ultraviolet and visible spectrophotometry, fourier-transform infrared spectroscopy, and X-ray powder diffraction. In vitro release, cytotoxicity, and stability were mainly studied with MPN-NPs loading paclitaxel. The suitability of MPN as a nanocrystal stabilizer was also investigated for other classical hydrophobic drugs, including simvastatin, andrographolide, atorvastatin calcium, ferulic acid, and famotidine. The results showed that MPN could effectively wrap and stabilize various drug nanocrystals apart from famotidine. The maximum solubilization of MPN towards atorvastatin calcium was up to 1587 folds, and it also exhibited an excellent solubilizing effect on other hydrophobic drugs. We disclosed that the drug was entrapped in MPN in the nanocrystal form, and there were distinct physiochemical interactions between MPN and the payload. Our findings suggested that MPN may be a promising platform for nanocrystal production to address the challenge of low solubility associated with hydrophobic drugs. Graphical abstract.
Collapse
|
27
|
Brannon ER, Guevara MV, Pacifici NJ, Lee JK, Lewis JS, Eniola-Adefeso O. Polymeric particle-based therapies for acute inflammatory diseases. NATURE REVIEWS. MATERIALS 2022; 7:796-813. [PMID: 35874960 PMCID: PMC9295115 DOI: 10.1038/s41578-022-00458-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/09/2022] [Indexed: 05/02/2023]
Abstract
Acute inflammation is essential for initiating and coordinating the body's response to injuries and infections. However, in acute inflammatory diseases, inflammation is not resolved but propagates further, which can ultimately lead to tissue damage such as in sepsis, acute respiratory distress syndrome and deep vein thrombosis. Currently, clinical protocols are limited to systemic steroidal treatments, fluids and antibiotics that focus on eradicating inflammation rather than modulating it. Strategies based on stem cell therapeutics and selective blocking of inflammatory molecules, despite showing great promise, still lack the scalability and specificity required to treat acute inflammation. By contrast, polymeric particle systems benefit from uniform manufacturing at large scales while preserving biocompatibility and versatility, thus providing an ideal platform for immune modulation. Here, we outline design aspects of polymeric particles including material, size, shape, deformability and surface modifications, providing a strategy for optimizing the targeting of acute inflammation.
Collapse
Affiliation(s)
- Emma R. Brannon
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI USA
| | | | - Noah J. Pacifici
- Department of Biomedical Engineering, University of California, Davis, CA USA
| | - Jonathan K. Lee
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI USA
| | - Jamal S. Lewis
- Department of Biomedical Engineering, University of California, Davis, CA USA
| | | |
Collapse
|
28
|
Development of nanostructured systems using natural polymers to optimize the treatment of inflammatory bowel diseases: A prospective study. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
29
|
Hartwig O, Shetab Boushehri MA, Shalaby KS, Loretz B, Lamprecht A, Lehr CM. Drug delivery to the inflamed intestinal mucosa - targeting technologies and human cell culture models for better therapies of IBD. Adv Drug Deliv Rev 2021; 175:113828. [PMID: 34157320 DOI: 10.1016/j.addr.2021.113828] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/19/2022]
Abstract
Current treatment strategies for inflammatory bowel disease (IBD) seek to alleviate the undesirable symptoms of the disorder. Despite the higher specificity of newer generation therapeutics, e.g. monoclonal antibodies, adverse effects still arise from their interference with non-specific systemic immune cascades. To circumvent such undesirable effects, both conventional and newer therapeutic options can benefit from various targeting strategies. Of course, both the development and the assessment of the efficiency of such targeted delivery systems necessitate the use of suitable in vivo and in vitro models representing relevant pathophysiological manifestations of the disorder. Accordingly, the current review seeks to provide a comprehensive discussion of the available preclinical models with emphasis on human in vitro models of IBD, along with their potentials and limitations. This is followed by an elaboration on the advancements in the field of biology- and nanotechnology-based targeted drug delivery systems and the potential rooms for improvement to facilitate their clinical translation.
Collapse
Affiliation(s)
- Olga Hartwig
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, D-66123 Saarbrücken, Germany
| | | | - Karim S Shalaby
- Department of Pharmaceutics, University of Bonn, D-53121 Bonn, Germany; Department of Pharmaceutics and Industrial Pharmacy, Ain Shams University, Cairo, Egypt
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany
| | - Alf Lamprecht
- Department of Pharmaceutics, University of Bonn, D-53121 Bonn, Germany.
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, D-66123 Saarbrücken, Germany.
| |
Collapse
|
30
|
Chen F, Liu Q, Xiong Y, Xu L. Current Strategies and Potential Prospects of Nanomedicine-Mediated Therapy in Inflammatory Bowel Disease. Int J Nanomedicine 2021; 16:4225-4237. [PMID: 34188471 PMCID: PMC8236271 DOI: 10.2147/ijn.s310952] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/07/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel diseases (IBD) such as Crohn's disease and ulcerative colitis are highly debilitating. IBDs are associated with the imbalance of inflammatory mediators within the inflamed bowel. Conventional drugs for IBD treatment include anti-inflammatory medications and immune suppressants. However, they suffer from a lack of bioavailability and high dose-induced systemic side effects. Nanoparticle (NP)-derived therapy improves therapeutic efficacy and increases targeting specificity. Recent studies have shown that nanomedicines, based on bowel disease's pathophysiology, are a fast-growing field. NPs can prolong the circulation period and reduce side effects by improving drug encapsulation and targeted delivery. Here, this review summarizes various IBD therapies with a focus on NP-derived applications, whereas their challenges and future perspectives have also been discussed.
Collapse
Affiliation(s)
- Fengqian Chen
- Translational Research Program, Department of Anesthesiology and Center for Shock Trauma Anesthesiology Research, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Yang Xiong
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, People’s Republic of China
| | - Li Xu
- Department of Anorectal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, 310006, People’s Republic of China
| |
Collapse
|
31
|
Wang L, Yu M, Yang H. Recent Progress in the Diagnosis and Precise Nanocarrier-Mediated Therapy of Inflammatory Bowel Disease. J Inflamm Res 2021; 14:1701-1716. [PMID: 33953597 PMCID: PMC8092629 DOI: 10.2147/jir.s304101] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/30/2021] [Indexed: 12/23/2022] Open
Abstract
The effective colon drug delivery remains to be an international frontier research in inflammatory bowel disease (IBD) therapy. The exploration and research of nanocarrier-based nanomedicine with great potential brings new opportunities for IBD therapy and diagnoses. Functional nanocarriers with varying morphology and characteristics can not only effectively avoid the destruction of the complex gastrointestinal (GI) tract microenvironment but also endow drugs with target therapy and improved bioavailability, thus elevating therapeutic efficacy. In this review, we illustrated several challenges in IBD therapy, then emphasis on some latest research progress of nanoparticles based therapy of oral administration, rectal administration and parenteral administration, as well as IBD diagnoses. Finally, we described the future perspective of nanocarriers in the treatment and diagnoses of IBD.
Collapse
Affiliation(s)
- Liucan Wang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Min Yu
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
32
|
Oligonucleotide Delivery across the Caco-2 Monolayer: The Design and Evaluation of Self-Emulsifying Drug Delivery Systems (SEDDS). Pharmaceutics 2021; 13:pharmaceutics13040459. [PMID: 33800701 PMCID: PMC8066367 DOI: 10.3390/pharmaceutics13040459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 01/21/2023] Open
Abstract
Oligonucleotides (OND) represent a promising therapeutic approach. However, their instability and low intestinal permeability hamper oral bioavailability. Well-established for oral delivery, self-emulsifying drug delivery systems (SEDDS) can overcome the weakness of other delivery systems such as long-term instability of nanoparticles or complicated formulation processes. Therefore, the present study aims to prepare SEDDS for delivery of a nonspecific fluorescently labeled OND across the intestinal Caco-2 monolayer. The hydrophobic ion pairing of an OND and a cationic lipid served as an effective hydrophobization method using either dimethyldioctadecylammonium bromide (DDAB) or 1,2-dioleoyl-3-trimethylammonium propane (DOTAP). This strategy allowed a successful loading of OND-cationic lipid complexes into both negatively charged and neutral SEDDS. Subjecting both complex-loaded SEDDS to a nuclease, the negatively charged SEDDS protected about 16% of the complexed OND in contrast to 58% protected by its neutral counterpart. Furthermore, both SEDDS containing permeation-enhancing excipients facilitated delivery of OND across the intestinal Caco-2 cell monolayer. The negatively charged SEDDS showed a more stable permeability profile over 120 min, with a permeability of about 2 × 10-7 cm/s, unlike neutral SEDDS, which displayed an increasing permeability reaching up to 7 × 10-7 cm/s. In conclusion, these novel SEDDS-based formulations provide a promising tool for OND protection and delivery across the Caco-2 cell monolayer.
Collapse
|
33
|
Targeting strategies of oral nano-delivery systems for treating inflammatory bowel disease. Int J Pharm 2021; 600:120461. [PMID: 33711470 DOI: 10.1016/j.ijpharm.2021.120461] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 02/22/2021] [Accepted: 03/04/2021] [Indexed: 01/01/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic relapsing inflammatory disorder of gastrointestinal tract with rising incidence. Established treatments of IBD are characterized by significantly adverse effects, insufficient therapeutic efficacy. Employing the oral nano-drug delivery systems for targeted therapy is capable of effectively avoiding systematic absorption and increasing local drug concentration, consequently leading to decreased adverse effects and improved therapeutic outcomes. This review gives a brief profile of pathophysiological considerations in terms of developing disease-directed drug delivery systems, then focuses on mechanisms and strategies of current oral nano-drug delivery systems, including size-, enzyme-, redox-, pH-, ligand-receptor-, mucus-dependent systems, and proposes the future directions of managements for IBD.
Collapse
|
34
|
Zhang L, Li M, Zhang G, Gao C, Wang S, Zhang T, Ma C, Wang L, Zhu Q. Micro- and Nanoencapsulated Hybrid Delivery System (MNEHDS): A Novel Approach for Colon-Targeted Oral Delivery of Berberine. Mol Pharm 2021; 18:1573-1581. [PMID: 33629860 DOI: 10.1021/acs.molpharmaceut.0c00970] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Berberine (BBR) is currently explored in the oral treatment of many disorders, especially in those involving inflammatory processes. Nanotechnology-based drug delivery systems are emerging as an effective approach for improving the poor oral absorption/bioavailability of BBR. To optimize the BBR immunoregulatory effects on a specific part of the gastrointestinal tract, here we describe a micro- and nanoencapsulated hybrid delivery system (MNEHDS) for colon-targeted oral delivery of BBR and test its therapeutic efficacy in a murine colitis model. The MNEHDS is formed by encapsulation of BBR-loaded poly(lactic-co-glycolic acid) nanoparticles into a pH-sensitive, BBR-pre-entrapped Eudragit FS30D matrix to form a hybrid microparticle composed of the BBR and BBR nanoparticles. Once in the colonic environment, the microencapsulated BBR is almost completely released for immediate action, while BBR nanoparticles can provide sustained release of BBR subsequent to their intestinal absorption. One dose of oral MNEHDS/BBR treatment results in significant attenuation of acute colitis induced by dextran sulfate sodium. The MNEHDS/BBR also proves to be effective during chronically induced colitis with two doses given 1 week apart. The improved efficacy is accompanied by decreased production of colon inflammation. Comparatively, oral treatment with one or two 7-day courses of free BBR has less effect on ameliorating either acute or chronic colitis. Thus, MNEHDS represents a novel delivery system for BBR, and potentially other therapeutic agents, to treat inflammatory bowel disease.
Collapse
Affiliation(s)
- Lingzhi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Materia Medica, Beijing 100050, China
| | - Mingyan Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Materia Medica, Beijing 100050, China
| | - Guiqiu Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Materia Medica, Beijing 100050, China
| | - Changxing Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Materia Medica, Beijing 100050, China
| | - Shengfang Wang
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China.,College of Chemistry, Chemical Engineering and Resource Utilization, Northeast Forestry University, Harbin 150040, P. R. China
| | - Tingting Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Materia Medica, Beijing 100050, China
| | - Chen Ma
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Materia Medica, Beijing 100050, China
| | - Lianyan Wang
- Key Laboratory of Green Process and Engineering, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, P.R. China
| | - Qing Zhu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, and Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Chinese Academy of Medical Sciences and Peking Union Medical College Institute of Materia Medica, Beijing 100050, China
| |
Collapse
|
35
|
Gao C, Yu S, Zhang X, Dang Y, Han DD, Liu X, Han J, Hui M. Dual Functional Eudragit ® S100/L30D-55 and PLGA Colon-Targeted Nanoparticles of Iridoid Glycoside for Improved Treatment of Induced Ulcerative Colitis. Int J Nanomedicine 2021; 16:1405-1422. [PMID: 33658780 PMCID: PMC7917316 DOI: 10.2147/ijn.s291090] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 01/09/2021] [Indexed: 12/15/2022] Open
Abstract
AIM Iridoid glycosides (IG) as the major active fraction of Syringa oblata Lindl. has a proven anti-inflammatory effect for ulcerative colitis (UC). However, its current commercial formulations are hampered by low bioavailability and unable to reach inflamed colon. To overcome the limitation, dual functional IG-loaded nanoparticles (DFNPs) were prepared to increase the residence time of IG in colon. The protective mechanism of DFNPs on DSS-induced colonic injury was evaluated in rats. MATERIALS AND METHODS We prepared DFNPs using the oil-in-water emulsion method. PLGA was selected as sustained-release polymer, and ES100 and EL30D-55 as pH-responsive polymers. The morphology and size distribution of NPs were measured by SEM and DLS technique. To evaluate colon targeting of DFNPs, DiR, was encapsulated as a fluorescent probe into NPs. Fluorescent distribution of NPs were investigated. The therapeutic potential and in vivo transportation of NPs in gastrointestinal tract were evaluated in a colitis model. RESULTS SEM images and zeta data indicated the successful preparation of DFNPs. This formulation exhibited high loading capacity. Drug release results suggested DFNPs released less than 20% at the first 6 h in simulated gastric fluid (pH1.2) and simulated small intestine fluid (pH6.8). A high amount of 84.7% sustained release from NPs in simulated colonic fluid (pH7.4) was beyond 24 h. DiR-loaded NPs demonstrated a much higher colon accumulation, suggesting effective targeting due to functionalization with pH and time-dependent polymers. DFNPs could significantly ameliorate the colonic damage by reducing DAI, macroscopic score, histological damage and cell apoptosis. Our results also proved that the potent anti-inflammatory effect of DFNPs is contributed by decrease of NADPH, gene expression of COX-2 and MMP-9 and the production of TNF-α, IL-17, IL-23 and PGE2. CONCLUSION We confirm that DFNPs exert protective effects through inhibiting the inflammatory response, which could be developed as a potential colon-targeted system.
Collapse
Affiliation(s)
- Chenzhe Gao
- Food Science College, Northeast Agricultural University, Harbin, People’s Republic of China
- Department of Pharmaceutical Engineering, School of Materials Science and Chemical Engineering, Key Laboratory of Green Chemical Engineering in Heilongjiang Province, Harbin University of Science and Technology, Harbin, People’s Republic of China
| | - Shen Yu
- Department of Pharmaceutical Engineering, School of Materials Science and Chemical Engineering, Key Laboratory of Green Chemical Engineering in Heilongjiang Province, Harbin University of Science and Technology, Harbin, People’s Republic of China
| | - Xiaonan Zhang
- College of Biological and Food Engineering, Guangdong University of Petrochemical Technology, Maoming, People’s Republic of China
| | - Yanxin Dang
- Department of Pharmaceutical Engineering, School of Materials Science and Chemical Engineering, Key Laboratory of Green Chemical Engineering in Heilongjiang Province, Harbin University of Science and Technology, Harbin, People’s Republic of China
- Pharmacy Department, Fourth Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, People’s Republic of China
| | - Dan-dan Han
- Department of Pharmaceutical Engineering, School of Materials Science and Chemical Engineering, Key Laboratory of Green Chemical Engineering in Heilongjiang Province, Harbin University of Science and Technology, Harbin, People’s Republic of China
| | - Xin Liu
- Department of Pharmaceutical Engineering, School of Materials Science and Chemical Engineering, Key Laboratory of Green Chemical Engineering in Heilongjiang Province, Harbin University of Science and Technology, Harbin, People’s Republic of China
- Department of Pharmacology, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - Janchun Han
- Food Science College, Northeast Agricultural University, Harbin, People’s Republic of China
| | - Mizhou Hui
- Food Science College, Northeast Agricultural University, Harbin, People’s Republic of China
| |
Collapse
|
36
|
Ribeiro SB, de Araújo AA, Oliveira MMB, dos Santos Silva AM, da Silva-Júnior AA, Guerra GCB, Brito GADC, Leitão RFDC, de Araújo Júnior RF, Garcia VB, Vasconcelos RC, de Medeiros CACX. Effect of Dexamethasone-Loaded PLGA Nanoparticles on Oral Mucositis Induced by 5-Fluorouracil. Pharmaceutics 2021; 13:53. [PMID: 33406583 PMCID: PMC7823510 DOI: 10.3390/pharmaceutics13010053] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/23/2020] [Accepted: 12/01/2020] [Indexed: 01/06/2023] Open
Abstract
Oral mucositis (OM) is characterized by the presence of severe ulcers in the oral region that affects patients treated with chemotherapy. It occurs in almost all patients who receive radiotherapy of the head and neck, as well as patients who undergo hematopoietic cell transplantation. The pathophysiology of OM is complex, and there is no effective therapy. The aim of this study was to evaluate the effect of dexamethasone-loaded poly(d,l-Lactic-co-glycolic) nanoparticles (PLGA-DEX NPs) on an OM model induced in hamsters. The NPs were synthesized using the emulsification-solvent evaporation method and were characterized by the size, zeta potential, encapsulation efficiency, atomic force microscopy, physicochemical stability, and the in vitro release. The OM was induced by the administration of 5-FU on the first and second days and mechanical trauma on the 4th day of the experiment. PLGA-DEX NPs were administered to treat OM. The animals were euthanized on the 10th day. Macroscopic and histopathological analyses were performed, measurement of malonaldehyde (MDA) and ELISA was used to determine the levels of IL-1β and TNF-α. Immunoexpressions of NF-κB, COX-2, and TGF-β were determined by immunohistochemistry, and qRT-PCR was used to quantify the gene expression of the GILZ, MKP1, and NF-κB p65. The PLGA-DEX NPs (0.1 mg/kg) significantly reduced macroscopic and histopathological scores, decreased MDA, TNF-α and IL-1β levels, immunostaining for NF-κB, COX-2, TGF-β, and suppressed NF-κB p65 mRNA expression, but increased GILZ and MKP1 expression.
Collapse
Affiliation(s)
- Susana Barbosa Ribeiro
- Post Graduate Program Biotechnology-RENORBIO, Federal University of Rio Grande do Norte, 3000 Senador Salgado Filho Ave, Lagoa Nova, Natal RN 59078-970, Brazil; (S.B.R.); (M.M.B.O.)
| | - Aurigena Antunes de Araújo
- Post Graduate Program Dental Sciences, Post Graduate Program Pharmaceutical Science, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, 3000 Senador Salgado Filho Ave, Lagoa Nova, Natal RN 59078-970, Brazil;
| | - Maisie Mitchele Barbosa Oliveira
- Post Graduate Program Biotechnology-RENORBIO, Federal University of Rio Grande do Norte, 3000 Senador Salgado Filho Ave, Lagoa Nova, Natal RN 59078-970, Brazil; (S.B.R.); (M.M.B.O.)
| | - Alaine Maria dos Santos Silva
- Laboratory of Pharmaceutical Technology & Biotechnology (TecBioFar), Post Graduate Program Pharmaceutical Sciences, Pharmacy Department, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Faria St, Petrópolis, Natal RN 59012-570, Brazil; (A.M.d.S.S.); (A.A.d.S.-J.)
| | - Arnóbio Antônio da Silva-Júnior
- Laboratory of Pharmaceutical Technology & Biotechnology (TecBioFar), Post Graduate Program Pharmaceutical Sciences, Pharmacy Department, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Faria St, Petrópolis, Natal RN 59012-570, Brazil; (A.M.d.S.S.); (A.A.d.S.-J.)
| | - Gerlane Coelho Bernardo Guerra
- Post Graduate Program Biochemistry and Molecular Biology, Post Graduate Program Pharmaceutical Science, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, 3000 Senador Salgado Filho Ave, Lagoa Nova, Natal RN 59078-970, Brazil;
| | - Gerly Anne de Castro Brito
- Post Graduate Program Morphofunctional Sciences, Department of Morphology, Faculty of Medicine, Federal University of Ceará, Delmiro de Farias St, Rodolfo Teófilo, Fortaleza CE 60416-030, Brazil; (G.A.d.C.B.); (R.F.d.C.L.)
| | - Renata Ferreira de Carvalho Leitão
- Post Graduate Program Morphofunctional Sciences, Department of Morphology, Faculty of Medicine, Federal University of Ceará, Delmiro de Farias St, Rodolfo Teófilo, Fortaleza CE 60416-030, Brazil; (G.A.d.C.B.); (R.F.d.C.L.)
| | - Raimundo Fernandes de Araújo Júnior
- Post Graduate Program Functional and Structural Biology, Post Graduate Program Health Science, Department of Morphology, Federal University of Rio Grande do Norte, 3000 Senador Salgado Filho Ave, Lagoa Nova, Natal RN 59078-970, Brazil;
| | - Vinícius Barreto Garcia
- Post Graduate Program Health Science, Federal University of Rio Grande do Norte, General Gustavo Cordeiro de Faria St, Petrópolis, Natal RN 59012-570, Brazil;
| | | | - Caroline Addison Carvalho Xavier de Medeiros
- Post Graduate Program Biotechnology-RENORBIO, Federal University of Rio Grande do Norte, 3000 Senador Salgado Filho Ave, Lagoa Nova, Natal RN 59078-970, Brazil; (S.B.R.); (M.M.B.O.)
- Post Graduate Program Biochemistry and Molecular Biology, Department of Biophysics and Pharmacology, Federal University of Rio Grande do Norte, 3000 Senador Salgado Filho Ave, Lagoa Nova, Natal RN 59078-970, Brazil
| |
Collapse
|
37
|
Nanoparticle-based therapeutics of inflammatory bowel diseases: a narrative review of the current state and prospects. JOURNAL OF BIO-X RESEARCH 2020. [DOI: 10.1097/jbr.0000000000000078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
38
|
Abasian P, Shakibi S, Maniati MS, Nouri Khorasani S, Khalili S. Targeted delivery, drug release strategies, and toxicity study of polymeric drug nanocarriers. POLYM ADVAN TECHNOL 2020. [DOI: 10.1002/pat.5168] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Payam Abasian
- Department of Chemical Engineering Isfahan University of Technology Isfahan Iran
| | - Sepideh Shakibi
- Department of Textile Engineering Amirkabir University of Technology (Tehran Polytechnique) Tehran Iran
| | - Mohammad Saeed Maniati
- Cellular and Molecular Biology Research Center, Health Research Institute Babol University of Medical Sciences Babol Iran
| | | | - Shahla Khalili
- Department of Chemical Engineering Isfahan University of Technology Isfahan Iran
| |
Collapse
|
39
|
Naeem M, Lee J, Oshi MA, Cao J, Hlaing SP, Im E, Jung Y, Yoo JW. Colitis-targeted hybrid nanoparticles-in-microparticles system for the treatment of ulcerative colitis. Acta Biomater 2020; 116:368-382. [PMID: 32937207 DOI: 10.1016/j.actbio.2020.09.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/26/2022]
Abstract
Nanoparticle (NP)-based drug delivery systems accumulate in the disrupted epithelium of inflamed colon tissue in ulcerative colitis. However, premature early drug release and uptake or degradation of NPs during their passage through the harsh gastric or intestinal environment compromise their therapeutic outcomes. This study aimed to develop an advanced colitis-targeted hybrid nanoparticles-in-microparticles (NPsinMPs) drug delivery system to overcome the aforementioned challenges. First, sustained drug releasing poly(lactic-co-glycolic acid) NPs were generated and further encapsulated in pH-sensitive Eudragit FS30D MPs to ensure complete drug protection in a gastric-like pH and for selective delivery of NPs to the colon. SEM and confocal microscopy for the NPsinMPs revealed successful NP encapsulation. NPsinMPs prevented drug release in an acidic gastric-like and intestinal-like pH and presented a sustained release thereafter at an ileal and colonic pH, indicating the degradation of the outer pH-sensitive MPs and release of NPs. Furthermore, in vivo imaging of gastrointestinal tract of a colitis mouse orally administered with fluorescent NPsinMPs revealed higher fluorescence intensities selectively in the colon, demonstrating the release of loaded NPs and their concomitant accumulation at the site of colon inflammation. NPsinMPs markedly mitigated experimental colitis in mice indicated by improved histopathological analysis, decreased myeloperoxidase activity, neutrophils and macrophage infiltration, and expression of proinflammatory cytokines in colonic tissues compared with NP-treated mice. The present results show the successful formulation of an NPsinMP-based drug delivery system and provide a platform to improve NP-based colon-targeted drug delivery through improved protection of encapsulated NPs and their payload in the early small intestine.
Collapse
Affiliation(s)
- Muhammad Naeem
- College of Pharmacy, Pusan National University, Busan609-735, South Korea; Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Punjab, Pakistan
| | - Juho Lee
- College of Pharmacy, Pusan National University, Busan609-735, South Korea
| | - Murtada A Oshi
- College of Pharmacy, Pusan National University, Busan609-735, South Korea
| | - Jiafu Cao
- College of Pharmacy, Pusan National University, Busan609-735, South Korea
| | - Shwe Phyu Hlaing
- College of Pharmacy, Pusan National University, Busan609-735, South Korea
| | - Eunok Im
- College of Pharmacy, Pusan National University, Busan609-735, South Korea
| | - Yunjin Jung
- College of Pharmacy, Pusan National University, Busan609-735, South Korea
| | - Jin-Wook Yoo
- College of Pharmacy, Pusan National University, Busan609-735, South Korea.
| |
Collapse
|
40
|
Qu Z, Wong KY, Moniruzzaman M, Begun J, Santos HA, Hasnain SZ, Kumeria T, McGuckin MA, Popat A. One‐Pot Synthesis of pH‐Responsive Eudragit‐Mesoporous Silica Nanocomposites Enable Colonic Delivery of Glucocorticoids for the Treatment of Inflammatory Bowel Disease. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000165] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Zhi Qu
- School of Pharmacy The University of Queensland Brisbane QLD 4102 Australia
- Immunopathology Group Mater Research Institute –The University of Queensland Translational Research Institute Brisbane QLD 4102 Australia
| | - Kuan Yau Wong
- Immunopathology Group Mater Research Institute –The University of Queensland Translational Research Institute Brisbane QLD 4102 Australia
| | - Md. Moniruzzaman
- School of Pharmacy The University of Queensland Brisbane QLD 4102 Australia
- Inflammatory Bowel Disease Group, Mater Research Institute–The University of Queensland Translational Research Institute Brisbane QLD 4102 Australia
| | - Jakob Begun
- Inflammatory Bowel Disease Group, Mater Research Institute–The University of Queensland Translational Research Institute Brisbane QLD 4102 Australia
- Mater Hospital Brisbane Mater Health Services South Brisbane QLD 4102 Australia
| | - Hélder A Santos
- Drug Research Program Division of Pharmaceutical Chemistry and Technology Faculty of Pharmacy University of Helsinki Helsinki FI‐00014 Finland
- Helsinki Institute of Life Science (HiLIFE) University of Helsinki Helsinki FI‐00014 Finland
| | - Sumaira Z. Hasnain
- School of Pharmacy The University of Queensland Brisbane QLD 4102 Australia
- Immunopathology Group Mater Research Institute –The University of Queensland Translational Research Institute Brisbane QLD 4102 Australia
| | - Tushar Kumeria
- School of Pharmacy The University of Queensland Brisbane QLD 4102 Australia
- Immunopathology Group Mater Research Institute –The University of Queensland Translational Research Institute Brisbane QLD 4102 Australia
| | - Michael A. McGuckin
- Faculty of Medicine Dentistry and Health Sciences the University of Melbourne Melbourne VIC 3010 Australia
| | - Amirali Popat
- School of Pharmacy The University of Queensland Brisbane QLD 4102 Australia
- Immunopathology Group Mater Research Institute –The University of Queensland Translational Research Institute Brisbane QLD 4102 Australia
| |
Collapse
|
41
|
Brusini R, Varna M, Couvreur P. Advanced nanomedicines for the treatment of inflammatory diseases. Adv Drug Deliv Rev 2020; 157:161-178. [PMID: 32697950 PMCID: PMC7369016 DOI: 10.1016/j.addr.2020.07.010] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/04/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023]
Abstract
Inflammation, a common feature of many diseases, is an essential immune response that enables survival and maintains tissue homeostasis. However, in some conditions, the inflammatory process becomes detrimental, contributing to the pathogenesis of a disease. Targeting inflammation by using nanomedicines (i.e. nanoparticles loaded with a therapeutic active principle), either through the recognition of molecules overexpressed onto the surface of activated macrophages or endothelial cells, or through enhanced vasculature permeability, or even through biomimicry, offers a promising solution for the treatment of inflammatory diseases. After providing a brief insight on the pathophysiology of inflammation and current therapeutic strategies, the review will discuss, at a pre-clinical stage, the main innovative nanomedicine approaches that have been proposed in the past five years for the resolution of inflammatory disorders, finally focusing on those currently in clinical trials.
Collapse
|
42
|
Youshia J, Ali ME, Stein V, Lamprecht A. Nanoparticles' properties modify cell type-dependent distribution in immune cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 29:102244. [PMID: 32561256 DOI: 10.1016/j.nano.2020.102244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/25/2020] [Accepted: 06/08/2020] [Indexed: 10/24/2022]
Abstract
Polymeric nanoparticles can passively target inflamed tissues. How their physicochemical properties affect their distribution pattern among the infiltrating immune cells is unknown. Polyvinyl acetate nanoparticles with different particle size (100 and 300 nm) and surface charge (cationic, non-ionic, and anionic) were prepared and incubated with either LPS-activated or unactivated murine splenocytes. Nanoparticle association with macrophages, dendritic cells, neutrophils, B and T cells was investigated using flow cytometry. Cells associated with nanoparticles as follows: cationic>anionic>non-ionic and 300 nm > 100 nm. 40% of ionic nanoparticles were distributed among unactivated macrophages, reduced to 25% for activated macrophages. 60% of 100 nm and 40% of 300 nm non-ionic nanoparticles were distributed among unactivated and LPS-activated macrophages. This study highlights that particles' physicochemical properties impact the number of nanoparticles associating with immune cells more than their distribution pattern, which is principally determined by the cell activation state. This suggests a disease-dependent distribution pattern for therapeutic nanoparticles.
Collapse
Affiliation(s)
- John Youshia
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mohamed Ehab Ali
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany; Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Valentin Stein
- Institute of Physiology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Alf Lamprecht
- Department of Pharmaceutics, Institute of Pharmacy, University of Bonn, Bonn, Germany; PEPITE EA4267, University of Bourgogne Franche-Comté, Besançon, France.
| |
Collapse
|
43
|
Oral delivery of oleuropein-loaded lipid nanocarriers alleviates inflammation and oxidative stress in acute colitis. Int J Pharm 2020; 586:119515. [PMID: 32544520 DOI: 10.1016/j.ijpharm.2020.119515] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 12/22/2022]
Abstract
Inflammation and oxidative stress pathways have emerged as novel targets in the management of inflammatory bowel diseases (IBD). Targeting the drug to the inflamed colon remains a challenge. Nanostructured lipid carriers (NLCs) have been reported to accumulate in inflamed colonic mucosa. The antioxidant/antiinflamatory polyphenol oleuropein (OLE) was loaded in NLCs (NLC-OLE). NLC-OLE showed to be more effective in decreasing the TNF-α secretion and intracellular reactive oxygen species (ROS) by activated macrophages (J774) compared to the conventional form of OLE. OLE efficacy was preserved within NLC-OLE ameliorating inflammation in a murine model of acute colitis: reduced levels of TNF-α and IL-6, decreased neutrophil infiltration and improved histopathology of the colon were reported. In addition, NLC-OLE enhanced the ROS scavenging activity of OLE in the colon after oral administration. These data suggest that the proposed NLC-OLE could be a promising drug delivery system for OLE in IBD treatment.
Collapse
|
44
|
Cartaxo da Costa Urtiga S, Rodrigues Marcelino H, Sócrates Tabosa do Egito E, Eleamen Oliveira E. Xylan in drug delivery: A review of its engineered structures and biomedical applications. Eur J Pharm Biopharm 2020; 151:199-208. [DOI: 10.1016/j.ejpb.2020.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 12/15/2022]
|
45
|
Sun Y, Duan B, Chen H, Xu X. A Novel Strategy for Treating Inflammatory Bowel Disease by Targeting Delivery of Methotrexate through Glucan Particles. Adv Healthc Mater 2020; 9:e1901805. [PMID: 32092235 DOI: 10.1002/adhm.201901805] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/27/2020] [Indexed: 12/24/2022]
Abstract
Therapy of inflammatory bowel disease (IBD) has been a difficult task in the medical field. There is a great clinical need for more effective treatments for IBD. Herein, a targeted oral delivery system of yeast glucan particles (YGPs) carrying a clinically used anti-inflammatory drug methotrexate (MTX) to the inflamed sites in IBD mice for therapy is reported. In the findings, MTX is effectively loaded into YGPs through re-precipitation followed by gelation reaction of alginate to obtain the composite YGPs/MTX, which are internalized into RAW264.7 macrophage cells through dectin-1 and CR3 receptors. Furthermore, YGPs/MTX can suppress the proliferation of macrophage cells efficiently, leading to down-regulation of pro-inflammatory cytokines induced by lipopolysaccharides. Additionally, YGPs accumulate in the inflammation site of colitis mice, enabling YGPs/MTX to target the inflammatory site, significantly improve the efficacy of MTX, and reduce the cytotoxicity of MTX. Therefore, the YGPs-based drug delivery system provides a new strategy for MTX application in the clinical treatment of IBD.
Collapse
Affiliation(s)
- Ying Sun
- College of Chemistry and Molecular SciencesWuhan University Wuhan 430072 China
| | - Bingchao Duan
- College of Chemistry and Molecular SciencesWuhan University Wuhan 430072 China
| | - Huanhuan Chen
- College of Chemistry and Molecular SciencesWuhan University Wuhan 430072 China
| | - Xiaojuan Xu
- College of Chemistry and Molecular SciencesWuhan University Wuhan 430072 China
| |
Collapse
|
46
|
Sarker M, Jhung SH. Zr-MOF with free carboxylic acid for storage and controlled release of caffeine. J Mol Liq 2019. [DOI: 10.1016/j.molliq.2019.112060] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
47
|
Zhao J, Gao W, Cai X, Xu J, Zou D, Li Z, Hu B, Zheng Y. Nanozyme-mediated catalytic nanotherapy for inflammatory bowel disease. Theranostics 2019; 9:2843-2855. [PMID: 31244927 PMCID: PMC6568174 DOI: 10.7150/thno.33727] [Citation(s) in RCA: 143] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 02/27/2019] [Indexed: 12/18/2022] Open
Abstract
The overproduction of reactive oxygen species (ROS) is linked to inflammatory bowel disease (IBD) and causes oxidative damage to DNA, proteins, and lipids. These ROS promote the initiation and progression of ulcerative colitis (UC). This study proposes a unique concept of nanomaterials with intrinsic enzyme-like activity (nanozymes) to mediate catalytic nanotherapy for IBD. Methods: We first synthesized manganese Prussian blue nanozymes (MPBZs) with multi-enzyme activity. A dextran sulfate sodium (DSS)-induced mouse model of colitis was built. The ROS scavenging capacity and anti-inflammatory effects of the MPBZs were investigated. Results: As a proof of concept, MPBZs with multi-enzyme activity were constructed of variable valence elements (Mn and Fe) via a facile and efficient strategy. Due to the increased intestinal permeability and positively charged surfaces of inflamed mucosa in murine colitis, the prepared MPBZs with nanoscale sizes and negative charges preferentially accumulated at inflamed sites after oral administration. Importantly, MPBZs mediated catalytic nanotherapy for IBD in mice via a primary effect on the toll-like receptor signaling pathway without adverse side effects. Conclusion: MPBZs with multi-enzyme activity were constructed to treat IBD. This nanozyme-based approach is a promising strategy for catalytic nanotherapy in patients with colonic IBD.
Collapse
Affiliation(s)
- Jiulong Zhao
- Shanghai Institute of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233 (China)
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai 200433 (China)
| | - Wei Gao
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233 (China)
| | - Xiaojun Cai
- Shanghai Institute of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233 (China)
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233 (China)
| | - Jiajia Xu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai 200433 (China)
| | - Duowu Zou
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai 200433 (China)
| | - Zhaoshen Li
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai 200433 (China)
| | - Bing Hu
- Department of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233 (China)
| | - Yuanyi Zheng
- Shanghai Institute of Ultrasound in Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233 (China)
| |
Collapse
|
48
|
Ma P, Si X, Chen Q, Ma L, Hou M, Xu Z, Kang Y, Wang J, Xiao B. Oral Drug Delivery Systems for Ulcerative Colitis Therapy: A Comparative Study with Microparticles and Nanoparticles. Curr Cancer Drug Targets 2019; 19:304-311. [DOI: 10.2174/1568009618666181016152042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 06/28/2018] [Accepted: 07/07/2018] [Indexed: 12/31/2022]
Abstract
Background:
Oral administrations of microparticles (MPs) and nanoparticles (NPs) have
been widely employed as therapeutic approaches for the treatment of ulcerative colitis (UC). However,
no previous study has comparatively investigated the therapeutic efficacies of MPs and NPs.
Methods:
In this study, curcumin (CUR)-loaded MPs (CUR-MPs) and CUR-loaded NPs (CUR-NPs)
were prepared using a single water-in-oil emulsion solvent evaporation technique. Their therapeutic
outcomes against UC were further comparatively studied.
Results:
The resultant spherical MPs and NPs exhibited slightly negative zeta-potential with average
particle diameters of approximately 1.7 µm and 270 nm, respectively. It was found that NPs exhibited
a much higher CUR release rate than MPs within the same period of investigation. In vivo experiments
demonstrated that oral administration of CUR-MPs and CUR-NPs reduced the symptoms
of inflammation in a UC mouse model induced by dextran sulfate sodium. Importantly, CUR-NPs
showed much better therapeutic outcomes in alleviating UC compared with CUR-MPs.
Conclusion:
NPs can improve the anti-inflammatory activity of CUR by enhancing the drug release
and cellular uptake efficiency, in comparison with MPs. Thus, they could be exploited as a promising
oral drug delivery system for effective UC treatment.
Collapse
Affiliation(s)
- Panpan Ma
- Institute for Clean Energy and Advanced Materials, Faculty of Materials and Energy, Southwest University, Chongqing, China
| | - Xiaoying Si
- Institute for Clean Energy and Advanced Materials, Faculty of Materials and Energy, Southwest University, Chongqing, China
| | - Qiubing Chen
- Institute for Clean Energy and Advanced Materials, Faculty of Materials and Energy, Southwest University, Chongqing, China
| | - Lijun Ma
- Institute for Clean Energy and Advanced Materials, Faculty of Materials and Energy, Southwest University, Chongqing, China
| | - Meili Hou
- Institute for Clean Energy and Advanced Materials, Faculty of Materials and Energy, Southwest University, Chongqing, China
| | - Zhigang Xu
- Institute for Clean Energy and Advanced Materials, Faculty of Materials and Energy, Southwest University, Chongqing, China
| | - Yuejun Kang
- Institute for Clean Energy and Advanced Materials, Faculty of Materials and Energy, Southwest University, Chongqing, China
| | - Jianglin Wang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Xiao
- Institute for Clean Energy and Advanced Materials, Faculty of Materials and Energy, Southwest University, Chongqing, China
| |
Collapse
|
49
|
Shah A, Cedrone E, Sanders CE, Butcher D, DeFrancesco AM, DeGrange CL, Difilippantonio S, Edmondson EF, Dobrovolskaia M. The potential utility of iron oxide nanoparticles for the treatment of skin inflammation in a mouse model of psoriasis. PRECISION NANOMEDICINE 2019. [DOI: 10.33218/prnano2(1).181218.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Several studies have demonstrated that immunosuppressive effects accompany systemic administration of some iron oxide nanoparticles (IONPs). In our earlier study, conducted using human peripheral blood mononuclear cells, we showed that therapeutic formulation of IONPs, Feraheme, approved for clinical use in the United States for the treatment of iron deficiency in chronic kidney disease, suppresses the function of activated T-cells in vitro via a mechanism involving mitochondrial damage. Here we report an in vivo study demonstrating that topical application of Feraheme prior to disease onset decreases the development of skin lesions in the mouse model of chemically induced psoriasis.
Collapse
|
50
|
Mohan LJ, Daly JS, Ryan BM, Ramtoola Z. The future of nanomedicine in optimising the treatment of inflammatory bowel disease. Scand J Gastroenterol 2019; 54:18-26. [PMID: 30678499 DOI: 10.1080/00365521.2018.1563805] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
There have been major advancements in the treatment of inflammatory bowel disease (IBD) over the past three decades. However despite significant progress, the best available treatments continue to demonstrate variable efficacy in patients and are associated with adverse effects. Therefore there remains an unmet clinical need for ongoing therapeutic advances for IBD. In recent years nanomedicines have emerged as promising diagnostic and therapeutic tools. Nanoparticles in particular show promise to facilitate targeted oral drug delivery in IBD. Here we discuss the pitfalls of current therapies and explore the potential for nanoparticles to improve the treatment of IBD. This review examines the range of conventional and novel therapies which have benefited from nanoparticle-mediated delivery and highlights the proven therapeutic efficacy of this approach in preclinical models. These strategies under development represent a novel and innovative treatment for IBD.
Collapse
Affiliation(s)
- Lauren J Mohan
- a Division of Biology, Department of Anatomy , Royal College of Surgeons in Ireland , Dublin , Ireland.,b School of Pharmacy, Royal College of Surgeons in Ireland , Dublin , Ireland
| | - Jacqueline S Daly
- a Division of Biology, Department of Anatomy , Royal College of Surgeons in Ireland , Dublin , Ireland
| | - Barbara M Ryan
- c Department of Gastroenterology and Clinical Medicine , Tallaght Hospital and Trinity College , Dublin , Ireland
| | - Zebunnissa Ramtoola
- b School of Pharmacy, Royal College of Surgeons in Ireland , Dublin , Ireland
| |
Collapse
|