1
|
Pan X, Xian P, Li Y, Zhao X, Zhang J, Song Y, Nan Y, Ni S, Hu K. Chemotaxis-driven hybrid liposomes recover intestinal homeostasis for targeted colitis therapy. J Control Release 2025; 380:829-845. [PMID: 39961435 DOI: 10.1016/j.jconrel.2025.02.036] [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: 10/13/2024] [Revised: 02/05/2025] [Accepted: 02/14/2025] [Indexed: 02/22/2025]
Abstract
Inflammatory bowel disease (IBD) is closely linked to the dysregulation of intestinal homeostasis, accompanied by intestinal epithelial barrier destruction, dysbiosis of gut microbiota, subsequent inflammatory factor infiltration, and excessive oxidative stress. Conventional therapeutics focus on suppressing inflammation and often suffer from metabolic instability as well as limited targeting, thereby leading to suboptimal remission rates and severe side effects. Here, we designed bacterial outer membrane vesicle (OMV, from Stenotrophomonas maltophilia)-fused and borneol-modified liposomes (BO/OMV-lipo@LU) for targeted delivery of luteolin to recover intestinal homeostasis by alleviating inflammation and modulating dysregulated intestinal epithelial barrier, redox balance, and gut microbiota in IBD. In a Caco-2/HT29-MTX monolayer model, the OMV and borneol-bifunctionalized liposomes enhanced the uptake efficiency of unfunctionalized liposomes with a 2-fold increase. Owing to the chemotaxis-driven colon-targeting ability of OMVs and the ability of borneol to promote intestinal epithelial uptake, the hybrid liposomes successfully targeted the inflamed colon. In a colitis mouse model, BO/OMV-lipo@LU exhibited enhanced efficacy following oral administration. The BO/OMV-lipo@LU treatment increased the colon length and body weights of mice suffering colitis by 40 % and 15 %, respectively, with values comparable to the healthy control group. Notably, BO/OMV-lipo@LU alleviated proinflammatory markers, modulated redox balance, and restored the intestinal epithelial barrier. In addition, the formulation increased the abundance of beneficial microbiota while decreasing the abundance of harmful microbiota. These results demonstrated that this biomimetic nanoplatform could be exploited as a safe and effective gut-targeted delivery system in IBD treatment.
Collapse
Affiliation(s)
- Xier Pan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Peng Xian
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yushu Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao Zhao
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jiaxin Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yangjie Song
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yunrong Nan
- Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Shuting Ni
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Kaili Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
2
|
Chen Y, Shui M, Li H, Guo M, Yuan Q, Hao W, Wang T, Zhou H, Chen Z, Wang S. Inflammation-targeted delivery of probiotics for alleviation of colitis and associated cognitive disorders through improved vitality and colonization. Biomaterials 2025; 318:123163. [PMID: 39923539 DOI: 10.1016/j.biomaterials.2025.123163] [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: 10/20/2024] [Revised: 01/20/2025] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
Oral probiotic biotherapies hold significant promise for addressing intestinal inflammatory disorders. Nonetheless, due to the challenging pathological microenvironment of the gastrointestinal tract, it is difficult to achieve deliver probiotics in an inflammation-targeted manner while improving their intestinal colonization and minimizing the impact of gastrointestinal environment on their vitality. To address this, an innovative probiotics oral delivery system (EcN-Apt@HG) against ulcerative colitis (UC) was developed by conjugating IL-6 aptamer to the surface of EcN and subsequently encapsulating the probiotics in a hydrogel consisting of aldehyde-functionalized chondroitin sulfate (CS) and Poly(amidoamine) (PAMAM). As expected, the encapsulated EcN demonstrated resistance to gastrointestinal conditions, and the colonization duration of probiotics in the colon was enhanced via the preferential adhesion effect of IL-6 aptamer on the inflammatory site. The EcN-Apt@HG system restored the damaged mucosal layer, suppressed hyperactive immune responses, and reshaped the dysbiosis of intestinal microflora, thereby synergistically alleviating dextran sulfate sodium (DSS)-induced colitis. Notably, EcN-Apt@HG significantly alleviated depression-like behaviors and cognitive impairment in colitis mice through gut-brain axis interaction. This approach provides a simple and promising strategy for inflammation-targeted delivery of probiotics to the intestine and shows great potential for UC therapy and associated cognitive disorders.
Collapse
Affiliation(s)
- Yi Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Mingju Shui
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Hongyi Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Miao Guo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Qin Yuan
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Wei Hao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Tao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Hefeng Zhou
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, China
| | - Zhejie Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Shengpeng Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| |
Collapse
|
3
|
Zhang J, Yin YJ, Wang XW, Lu WQ, Chen ZY, Yu CH, Ren KF, Xu CF. Adhesive polyelectrolyte coating through UV-triggered polymerization on PLGA particles for enhanced drug delivery to inflammatory intestinal mucosa. J Nanobiotechnology 2025; 23:32. [PMID: 39844269 PMCID: PMC11753032 DOI: 10.1186/s12951-024-03066-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
Administering medication precisely to the inflamed intestinal sites to treat ulcerative colitis (UC), with minimized side effects, is of urgent need. In UC, the inflammation damaged mucosa contains a large number of amino groups which are positively charged, providing new opportunities for drug delivery system design. Here, we report an oral drug delivery system utilizing the tacrolimus-loaded poly (lactic-co-glycolic acid) (TAC/PLGA) particles with an adhesion coating by in situ UV-triggered polymerization of polyacrylic acid and N-hydroxysuccinimide (PAA-NHS). The negatively charged carboxyl groups effectively interact with the positively charged focal mucosa, and the NHS ester groups form the covalent bonds with the amino groups, thereby synergically enhancing the adhesion of the PLGA particles to the focal mucosa. Our findings reveal that, compared to the naked particles, the PAA-NHS coating increases the adhesion of particles to the inflammatory intestine. In a dextran sulfate sodium-induced acute colitis mouse model, the TAC/PLGA particles with PAA-NHS coating exhibits substantial retention of TAC within the inflammatory intestine, enhancing drug delivery efficiency and therapeutic effects. This approach holds promise for UC management, minimizing systemic side effects and optimizing therapeutic outcomes.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yi-Jing Yin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xing-Wang Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Wei-Qi Lu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhao-Yang Chen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China
| | - Chao-Hui Yu
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Ke-Feng Ren
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Cheng-Fu Xu
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
4
|
Kiilerich KF, Andresen T, Darbani B, Gregersen LHK, Liljensøe A, Bennike TB, Holm R, Moeller JB, Andersen V. Advancing Inflammatory Bowel Disease Treatment by Targeting the Innate Immune System and Precision Drug Delivery. Int J Mol Sci 2025; 26:575. [PMID: 39859291 PMCID: PMC11765494 DOI: 10.3390/ijms26020575] [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: 11/13/2024] [Revised: 01/04/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease and ulcerative colitis, involves chronic inflammation of the gastrointestinal tract. Current immune-modulating therapies are insufficient for 30-50% of patients or cause significant side effects, emphasizing the need for new treatments. Targeting the innate immune system and enhancing drug delivery to inflamed gut regions are promising strategies. Neutrophils play a central role in IBD by releasing reactive oxygen species (ROS) and neutrophil extracellular traps (NETs) -DNA-based structures with cytotoxic proteins-that contribute to mucosal damage and inflammation. Recent studies linking ROS production, DNA repair, and NET formation have identified NETs as potential therapeutic targets, with preclinical models showing positive outcomes from NET inhibition. Innovative oral drug delivery systems designed to target gut inflammation directly-without systemic absorption-could improve treatment precision and reduce side effects. Advanced formulations utilize properties such as particle size, surface modifications, and ROS-triggered release to selectively target the distal ileum and colon. A dual strategy that combines a deeper understanding of IBD pathophysiology to identify inflammation-related therapeutic targets with advanced drug delivery systems may offer significant promise. For instance, pairing NET inhibition with ROS-responsive nanocarriers could enhance treatment efficacy, though further research is needed. This synergistic approach has the potential to greatly improve outcomes for IBD patients.
Collapse
Affiliation(s)
- Kat F. Kiilerich
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (K.F.K.); (J.B.M.)
| | - Trine Andresen
- Department of Health Science and Technology, The Faculty of Medicine, Aalborg University, 9220 Aalborg Ø, Denmark; (T.A.); (T.B.B.)
| | - Behrooz Darbani
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
| | - Laura H. K. Gregersen
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
- Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Anette Liljensøe
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
| | - Tue B. Bennike
- Department of Health Science and Technology, The Faculty of Medicine, Aalborg University, 9220 Aalborg Ø, Denmark; (T.A.); (T.B.B.)
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
| | - René Holm
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5000 Odense, Denmark;
| | - Jesper B. Moeller
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (K.F.K.); (J.B.M.)
- Danish Institute for Advanced Study, University of Southern Denmark, 5000 Odense, Denmark
| | - Vibeke Andersen
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
- Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
5
|
Desai N, Nayi S, Khunt D, Kapoor DU, Salave S, Prajapati B, Vora C, Malviya R, Maheshwari R, Patel R. Zein: Potential biopolymer in inflammatory bowel diseases. J Biomed Mater Res A 2025; 113:e37785. [PMID: 39210660 DOI: 10.1002/jbm.a.37785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Effectively managing inflammatory bowel disease (IBD) poses difficulties due to its persistent nature and unpredictable episodes of exacerbation. There is encouraging evidence that personalized medication delivery systems can improve therapy efficacy while reducing the negative effects of standard medicines. Zein, a protein produced from corn, has garnered interest as a possible means of delivering drugs for the treatment of IBD. This review delves into Zein-based drug delivery systems, showcasing its biodegradability, controlled release capabilities, and biocompatibility. Studies have shown that Zein-based nanoparticles, microcarriers, and core-shell microparticles have the capacity to increase medication stability, enhance targeting in the intestines, and decrease toxicity in animal models of IBD. The review highlights the promise of Zein in personalized therapy for IBD and urges more study to enhance its clinical use.
Collapse
Affiliation(s)
- Nimeet Desai
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, India
| | - Smit Nayi
- Gujarat Technological University, School of Pharmacy, Gandhinagar, Gujarat, India
| | - Dignesh Khunt
- Gujarat Technological University, School of Pharmacy, Gandhinagar, Gujarat, India
| | | | - Sagar Salave
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, India
| | - Bhupendra Prajapati
- S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, India
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Chintan Vora
- WAYMADE India Pvt. Ltd., Vadodara, Gujarat, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Noida, Uttar Pradesh, India
| | - Rahul Maheshwari
- School of Pharmacy and Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), Hyderabad, India
| | - Ravi Patel
- Gujarat Technological University, School of Pharmacy, Gandhinagar, Gujarat, India
| |
Collapse
|
6
|
Zhang J, Zhao L, Liang A, Geng X, Hou M, Cheng H, Zhang S, Yang B, Li J, Chen J. Resistant starch grafted cerium-sulfasalazine infinite coordination polymers synergistically remold intestinal metabolic microenvironment for inflammatory bowel disease therapy. J Nanobiotechnology 2024; 22:785. [PMID: 39707422 DOI: 10.1186/s12951-024-03043-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic gastrointestinal disease which is closely related with the overproduced reactive oxygen species (ROS), increased pro-inflammatory cytokines and disordered intestinal microbes. However, current therapeutic methods usually ignored the interrelation among the pathogenesis, and mainly focused on a single factor, inducing clinical outcomes unsatisfied. Herein, biocompatible infinite coordination polymers of drugs (Ce-SASP-RS ICPs) composed of Ce ions, FDA-approved drug sulfasalazine (SASP) and natural ingredient resistant starch (RS) were developed for synergistic treatment of IBD. The proper Ce3+/Ce4+ ratio in Ce-SASP-RS ICPs can endow them with SOD-like activities, POD-like activities and •OH scavenging ability, which guarantee Ce-SASP-RS ICPs to simultaneously kill bacteria and maintain ROS balance through cascade reactions. Owing to the recovered redox balance microenvironment, SASP in Ce-SASP-RS ICPs can better play their anti-inflammatory function. Moreover, benefitting from the recovered metabolic balance of ROS and inflammatory cytokines in colon, resistant starch can also function better in modifying gut microbiota through generating short-chain fatty acids. Collectively, Ce-SASP-RS ICPs can synergistically restore intestinal metabolic microenvironment through modulating redox balance, attenuating inflammation and modifying intestinal flora. Hence, in view of the mutual influences among IBD pathogenesis, this work presents a synergistic intervention approach for effectively treating IBD.
Collapse
Affiliation(s)
- Jing Zhang
- School of Chemistry and Chemical Engineering, Henan University of Science and Technology, Luoyang, 471023, China
| | - Liyuan Zhao
- School of Chemistry and Chemical Engineering, Henan University of Science and Technology, Luoyang, 471023, China
| | - Along Liang
- Henan Key Laboratory of Nanocomposite and Applications, Institute of Nanostructured Functional Materials, Huanghe Science and Technology College, Zhengzhou, 450006, Henan, China
| | - Xueyan Geng
- Henan Key Laboratory of Nanocomposite and Applications, Institute of Nanostructured Functional Materials, Huanghe Science and Technology College, Zhengzhou, 450006, Henan, China
| | - Mengmeng Hou
- Henan Key Laboratory of Nanocomposite and Applications, Institute of Nanostructured Functional Materials, Huanghe Science and Technology College, Zhengzhou, 450006, Henan, China
| | - Haojie Cheng
- Henan Key Laboratory of Nanocomposite and Applications, Institute of Nanostructured Functional Materials, Huanghe Science and Technology College, Zhengzhou, 450006, Henan, China
| | - Shouren Zhang
- Henan Key Laboratory of Nanocomposite and Applications, Institute of Nanostructured Functional Materials, Huanghe Science and Technology College, Zhengzhou, 450006, Henan, China
| | - Baocheng Yang
- Henan Key Laboratory of Nanocomposite and Applications, Institute of Nanostructured Functional Materials, Huanghe Science and Technology College, Zhengzhou, 450006, Henan, China.
| | - Junbo Li
- School of Chemistry and Chemical Engineering, Henan University of Science and Technology, Luoyang, 471023, China.
| | - Jian Chen
- Henan Key Laboratory of Nanocomposite and Applications, Institute of Nanostructured Functional Materials, Huanghe Science and Technology College, Zhengzhou, 450006, Henan, China.
| |
Collapse
|
7
|
Ghosh S, Singh R, Goap TJ, Sunnapu O, Vanwinkle ZM, Li H, Nukavarapu SP, Dryden GW, Haribabu B, Vemula PK, Jala VR. Inflammation-targeted delivery of Urolithin A mitigates chemical- and immune checkpoint inhibitor-induced colitis. J Nanobiotechnology 2024; 22:701. [PMID: 39533380 PMCID: PMC11558909 DOI: 10.1186/s12951-024-02990-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Inflammatory bowel disease (IBD) treatment often involves systemic administration of anti-inflammatory drugs or biologics such as anti-TNF-α antibodies. However, current drug therapies suffer from limited efficacy due to unresponsiveness and adverse side effects. To address these challenges, we developed inflammation-targeting nanoparticles (ITNPs) using biopolymers derived from the gum kondagogu (Cochlospermum gossypium) plant. These ITNPs enable selective drug delivery to inflamed regions, offering improved therapeutic outcomes. We designed ITNPs that specifically bind to inflamed regions in both human and mouse intestines, facilitating more effective, uniform, and prolonged drug delivery within the inflamed tissues. Furthermore, we demonstrated that oral administration of ITNPs loaded with urolithin A (UroA), a microbial metabolite or its synthetic analogue UAS03 significantly attenuated chemical- and immune checkpoint inhibitor- induced colitis in pre-clinical models. In conclusion, ITNPs show great promise for delivering UroA or its analogues while enhancing therapeutic efficacy at lower doses and reduced frequency compared to free drug administration. This targeted approach offers a potential solution to enhance IBD treatment while minimizing systemic side effects.
Collapse
Affiliation(s)
- Sweta Ghosh
- Department of Microbiology and Immunology, Center for Microbiomics, Inflammation and Pathogenicity, UofL-Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Rajbir Singh
- Department of Microbiology and Immunology, Center for Microbiomics, Inflammation and Pathogenicity, UofL-Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Tanu Jain Goap
- Institute for Stem Cell Science and Regenerative Medicine (DBT-inStem), GKVK campus, Bellary Road, Bangalore, 560065, Karnataka, India
| | - Omprakash Sunnapu
- Institute for Stem Cell Science and Regenerative Medicine (DBT-inStem), GKVK campus, Bellary Road, Bangalore, 560065, Karnataka, India
| | - Zachary M Vanwinkle
- Department of Microbiology and Immunology, Center for Microbiomics, Inflammation and Pathogenicity, UofL-Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Hong Li
- UofL-Brown Cancer Cancer, University of Louisville, Louisville, KY, USA
| | - Syam P Nukavarapu
- Department of Biomedical Engineering, Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, United States of America
| | - Gerald W Dryden
- Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Bodduluri Haribabu
- Department of Microbiology and Immunology, Center for Microbiomics, Inflammation and Pathogenicity, UofL-Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Praveen Kumar Vemula
- Institute for Stem Cell Science and Regenerative Medicine (DBT-inStem), GKVK campus, Bellary Road, Bangalore, 560065, Karnataka, India.
| | - Venkatakrishna Rao Jala
- Department of Microbiology and Immunology, Center for Microbiomics, Inflammation and Pathogenicity, UofL-Brown Cancer Center, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
8
|
Seegobin N, McCoubrey LE, Vignal C, Waxin C, Abdalla Y, Fan Y, Awad A, Murdan S, Basit AW. Dual action tofacitinib-loaded PLGA nanoparticles alleviate colitis in an IBD mouse model. Drug Deliv Transl Res 2024:10.1007/s13346-024-01736-1. [PMID: 39527394 DOI: 10.1007/s13346-024-01736-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Inflammatory bowel disease (IBD) affects over 7 million people worldwide and significant side effects are associated with current therapies such as tofacitinib citrate (TFC), which is linked to increased risks of malignancy and congestive heart issues. To mitigate these systemic adverse effects, localised drug delivery via nano-sized carriers to inflamed gut tissues represents a promising approach. Herein, we aimed to optimise the synthesis of nanoparticles (NPs) using a low molecular weight grade of Poly(lactic-co-glycolic acid) (PLGA) 50:50 loaded with TFC. This approach leverages the dual anti-inflammatory action of TFC and the local production of anti-inflammatory short-chain fatty acids from the degradation of PLGA by colonic gut microbiota. NPs were produced by nanoprecipitation and characterised for their drug release profile in vitro. The efficacy of the enhanced PLGA-TFC NPs was then tested in a C57BL/6 DSS colitis mouse model. The release profile of TFC from the enhanced PLGA NPs showed a 40% burst release within the first hour, followed by up to 80% drug release in the colonic environment. Notably, the degradation of PLGA by colonic gut microbiota did not significantly influence TFC release. In the mouse model, neither PLGA NPs alone nor TFC alone showed significant effects on weight loss compared to the TFC-loaded PLGA NPs, emphasising the enhanced efficacy potential of the combined formulation. Altogether, these results suggest a promising role of NP delivery systems in enhancing TFC efficacy, marking a significant step towards reducing dosage and associated side effects in IBD treatment. This study underscores the potential of PLGA-TFC NPs in providing targeted and effective therapy for IBD.
Collapse
Affiliation(s)
- Nidhi Seegobin
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, WC1N 1AX, 29-39 Brunswick Square, London, UK
| | - Laura E McCoubrey
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, WC1N 1AX, 29-39 Brunswick Square, London, UK
- Drug Product Development, GSK R&D, Ware, SG12 0GX, UK
| | - Cécile Vignal
- Univ. Lille, Inserm, CHU Lille, UMR1286 - INFINITE - Institute for Translational Research in Inflammation, 59000, Lille, France
| | - Christophe Waxin
- Univ. Lille, Inserm, CHU Lille, UMR1286 - INFINITE - Institute for Translational Research in Inflammation, 59000, Lille, France
| | - Youssef Abdalla
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, WC1N 1AX, 29-39 Brunswick Square, London, UK
| | - Yue Fan
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, WC1N 1AX, 29-39 Brunswick Square, London, UK
| | - Atheer Awad
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, WC1N 1AX, 29-39 Brunswick Square, London, UK
- Department of Clinical, Pharmaceutical and Biological Sciences, University of Hertfordshire, College Lane, Hatfield, AL10 9AB, UK
| | - Sudaxshina Murdan
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, WC1N 1AX, 29-39 Brunswick Square, London, UK
| | - Abdul W Basit
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, WC1N 1AX, 29-39 Brunswick Square, London, UK.
| |
Collapse
|
9
|
Xi E, Zhao Y, Liu K, Ding Q, Yang F, Gao N, Sun H, Yuan Y, Zhu G. Residue-Free Orally Administered Drug Carrier Based on a Porous Aromatic Framework for Efficient Multisite Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404643. [PMID: 39016121 DOI: 10.1002/smll.202404643] [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: 06/06/2024] [Revised: 07/03/2024] [Indexed: 07/18/2024]
Abstract
Nowadays, oral medications are the primary method of treating disease due to their convenience, low cost, and safety, without the need for complex medical procedures. To maximize treatment effectiveness, almost all oral medications utilize drug carriers, such as capsules, liposomes, and sugar coatings. However, these carriers rely on dissolution or fragmentation to achieve drug release, which leads to drugs and carriers coabsorption in the body, causing unnecessary adverse drug reactions, such as nausea, vomiting, abdominal pain, and even death caused by allergy. Therefore, the ideal oral drug carrier should avoid degradation and absorption and be totally excreted after drug release at the desired location. Herein, a gastrointestinally stable oral drug carrier based on porous aromatic framework-1 (PAF-1) is constructed, and it is modified with famotidine (a well-known gastric drug) and mesalazine (a well-known ulcerative colitis drug) to verify the excellent potential of PAF-1. The results demonstrate that PAF-1 can accurately release famotidine in stomach, mesalazine in the intestine, and finally be completely excreted from the body without any residue after 12 h. The use of PAF materials for the construction of oral drug carriers with no residue in the gastrointestinal tract provides a new approach for efficient disease treatment.
Collapse
Affiliation(s)
- Enpeng Xi
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education and Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| | - Yun Zhao
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education and Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| | - Kangning Liu
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education and Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| | - Qi Ding
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education and Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| | - Fuming Yang
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education and Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| | - Nan Gao
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education and Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| | - Hanjun Sun
- School of Chemistry and Materials Science, Jiangsu Key Laboratory of New Power Batteries, Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Nanjing Normal University, Nanjing, 210023, China
| | - Ye Yuan
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education and Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| | - Guangshan Zhu
- Key Laboratory of Polyoxometalate and Reticular Material Chemistry of Ministry of Education and Faculty of Chemistry, Northeast Normal University, Changchun, 130024, China
| |
Collapse
|
10
|
Zhang X, Yi X, Gao X, Li Y, Shen X. Liver-Targeted Nanoparticles Loaded with Cannabidiol Based on Redox Response for Effective Alleviation of Acute Liver Injury. Foods 2024; 13:2464. [PMID: 39123655 PMCID: PMC11311329 DOI: 10.3390/foods13152464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 08/12/2024] Open
Abstract
The purpose of this work was to construct liver-targeted nanoparticles based on the redox response to effectively deliver cannabidiol (CBD) for the prevention of acute liver injury (ALI). CBD-loaded nanoparticles (CBD NPs) with a particle size of 126.5 ± 1.56 nm were prepared using the polymer DA-PP-LA obtained by grafting pullulan polysaccharide with deoxycholic acid (DA) and α-lipoic acid (α-LA). CBD NPs showed typical redox-response release behavior. Interestingly, CBD NPs exhibited admirable liver targeting ability, significantly accumulated in the liver, and effectively promoted the internalization of CBD in liver cells, thus effectively reducing the H2O2-induced oxidative damage of HepG2 cells and avoiding apoptosis. More importantly, CBD NPs effectively prevented CCl4-induced ALI by protecting liver function, ameliorating oxidative stress levels, inhibiting the production of inflammatory factors, and protecting the liver from histological damage. This study provides a promising strategy for achieving targeted delivery of CBD NPs in the liver, thereby effectively preventing ALI.
Collapse
Affiliation(s)
- Xuan Zhang
- School of Food Science and Engineering, Hainan University, Haikou 570228, China; (X.Z.); (X.Y.); (X.G.); (Y.L.)
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou 570228, China
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Hainan University, Haikou 570228, China
| | - Xiangzhou Yi
- School of Food Science and Engineering, Hainan University, Haikou 570228, China; (X.Z.); (X.Y.); (X.G.); (Y.L.)
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou 570228, China
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Hainan University, Haikou 570228, China
| | - Xia Gao
- School of Food Science and Engineering, Hainan University, Haikou 570228, China; (X.Z.); (X.Y.); (X.G.); (Y.L.)
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou 570228, China
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Hainan University, Haikou 570228, China
| | - Yongcheng Li
- School of Food Science and Engineering, Hainan University, Haikou 570228, China; (X.Z.); (X.Y.); (X.G.); (Y.L.)
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou 570228, China
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Hainan University, Haikou 570228, China
| | - Xuanri Shen
- School of Food Science and Engineering, Hainan University, Haikou 570228, China; (X.Z.); (X.Y.); (X.G.); (Y.L.)
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou 570228, China
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea, Hainan University, Haikou 570228, China
- College of Food Science and Engineering, Hainan Tropical Ocean University, Sanya 572022, China
| |
Collapse
|
11
|
Voci S, Gagliardi A, Ambrosio N, Zannetti A, Cosco D. Lipid- and polymer-based formulations containing TNF-α inhibitors for the treatment of inflammatory bowel diseases. Drug Discov Today 2024; 29:104090. [PMID: 38977124 DOI: 10.1016/j.drudis.2024.104090] [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: 02/23/2024] [Revised: 06/10/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024]
Abstract
Monoclonal antibodies inhibiting tumor necrosis factor-alpha (iTNF-α) have revolutionized the therapeutic regimen of inflammatory bowel disease, but their main drawback is the parenteral route of administration they require. An alternative approach lies in the delivery of these molecules to the area involved in the inflammatory process by means of innovative formulations able to promote their localization in affected tissues while also decreasing the number of administrations required. This review describes the advantages deriving from the use of lipid- and polymer-based systems containing iTNF-α, focusing on their physicochemical and technological properties and discussing the preclinical results obtained in vivo using rodent models of colitis.
Collapse
Affiliation(s)
- Silvia Voci
- Department of Health Sciences, University of Catanzaro 'Magna Græcia', 88100 Catanzaro, Italy
| | - Agnese Gagliardi
- Department of Health Sciences, University of Catanzaro 'Magna Græcia', 88100 Catanzaro, Italy
| | - Nicola Ambrosio
- Department of Health Sciences, University of Catanzaro 'Magna Græcia', 88100 Catanzaro, Italy
| | - Antonella Zannetti
- Institute of Biostructures and Bioimaging, National Research Council (IBB-CNR), Naples 80145, Italy
| | - Donato Cosco
- Department of Health Sciences, University of Catanzaro 'Magna Græcia', 88100 Catanzaro, Italy.
| |
Collapse
|
12
|
Zheng B, Wang L, Yi Y, Yin J, Liang A. Design strategies, advances and future perspectives of colon-targeted delivery systems for the treatment of inflammatory bowel disease. Asian J Pharm Sci 2024; 19:100943. [PMID: 39246510 PMCID: PMC11375318 DOI: 10.1016/j.ajps.2024.100943] [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: 03/31/2024] [Revised: 05/02/2024] [Accepted: 05/21/2024] [Indexed: 09/10/2024] Open
Abstract
Inflammatory bowel diseases (IBD) significantly contribute to high mortality globally and negatively affect patients' qualifications of life. The gastrointestinal tract has unique anatomical characteristics and physiological environment limitations. Moreover, certain natural or synthetic anti-inflammatory drugs are associated with poor targeting, low drug accumulation at the lesion site, and other side effects, hindering them from exerting their therapeutic effects. Colon-targeted drug delivery systems represent attractive alternatives as novel carriers for IBD treatment. This review mainly discusses the treatment status of IBD, obstacles to drug delivery, design strategies of colon-targeted delivery systems, and perspectives on the existing complementary therapies. Moreover, based on recent reports, we summarized the therapeutic mechanism of colon-targeted drug delivery. Finally, we addressed the challenges and future directions to facilitate the exploitation of advanced nanomedicine for IBD therapy.
Collapse
Affiliation(s)
- Baoxin Zheng
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Liping Wang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yan Yi
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jun Yin
- School of Traditional Chinese Material, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Aihua Liang
- Key Laboratory of Beijing for Identification and Safety Evaluation of Chinese Medicine, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| |
Collapse
|
13
|
Wang X, Qiu H, Chu C, Wang K, Lu B, Yang C, Liu B, Lan G, Ding W. Dual-Responsive Microsphere Based on Natural Sunflower Pollen for Hemostasis and Repair in Inflammatory Bowel Disease. ACS APPLIED MATERIALS & INTERFACES 2024; 16:30658-30670. [PMID: 38856560 DOI: 10.1021/acsami.4c02408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Noninvasive treatment of inflammatory bowel disease with lower gastrointestinal bleeding is a major clinical challenge. In this study, we designed an orally targeted microsphere based on sunflower pollen microcapsules to localize the site of inflammatory injury and promote hemostasis and tissue repair. Due to the Eudragit and ascorbate palmitate coatings, EL/AP@PS(t+Dex) demonstrates pH- and enzyme-responsive release of loaded drugs and helps to resist the harsh environment of the gastrointestinal tract. Both in vitro and in vivo experiments show the characteristics of inflammation targeting and mucosal adhesion, which reduce the systematic exposure and increase the local drug concentration. In the DSS model, orally administered EL/AP@PS(t+Dex) significantly alleviates hematochezia, inhabits intestinal inflammation, and remarkably promotes the recovery of the intestinal epithelial barrier to reduce the exposure of intestinal microvessels. Furthermore, EL/AP@PS(t+Dex) optimized the composition of intestinal microbiota, which benefits intestinal homeostasis. This finding provides a fundamental solution for the treatment of intestinal bleeding caused by inflammatory bowel disease (IBD).
Collapse
Affiliation(s)
- Xinyu Wang
- Division of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Haoyu Qiu
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
- National Engineering Research Center for Biomaterials, School of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Chengnan Chu
- Division of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Kai Wang
- Division of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Bitao Lu
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Chao Yang
- Division of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Baochen Liu
- Division of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| | - Guangqian Lan
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Weiwei Ding
- Division of Trauma and Acute Care Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, Jiangsu Province, China
| |
Collapse
|
14
|
Xu J, Zhang Y, Yao X, Wang S, Lv K, Luo G, Wang J, Li G. Intestinal Targeted Nanogel with Broad-Spectrum Autonomous ROS Scavenging Performance for Enhancing the Bioactivity of trans-Resveratrol. Int J Nanomedicine 2024; 19:5995-6014. [PMID: 38895150 PMCID: PMC11185258 DOI: 10.2147/ijn.s464849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024] Open
Abstract
Introduction To improve the bioavailability of trans-resveratrol (trans-Res), it is commonly co-delivered with antioxidant bioactives using a complex synthetic intestinal targeted carrier, however, which makes practical application challenging. Methods A nanogel (Ngel), as broad-spectrum autonomous ROS scavenger, was prepared using selenized thiolated sodium alginate (TSA-Se) and crosslinked with calcium lactate (CL) for loading trans-Res to obtain Ngel@Res, which maintained spherical morphology in the upper digestive tract but broke down in the lower digestive tract, resulting in trans-Res release. Results Under protection of Ngel, trans-Res showed enhanced stability and broad-spectrum ROS scavenging activity. The synergistic mucoadhesion of Ngel prolonged the retention time of trans-Res in the intestine. Ngel and Ngel@Res increased the lifespan of Caenorhabditis elegans to 26.00 ± 2.17 and 26.00 ± 4.27 days by enhancing the activity of antioxidases, upregulating the expression of daf-16, sod-5 and skn-1, while downregulating the expression of daf-2 and age-1. Conclusion This readily available, intestinal targeted selenized alginate-based nanogel effectively improves the bioactivity of trans-Res.
Collapse
Affiliation(s)
- Jingwen Xu
- School of Food Science and Engineering, School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi’an, 710021, People’s Republic of China
- Xi’an Key Laboratory of Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi’an, 710021, People’s Republic of China
| | - Yue Zhang
- School of Food Science and Engineering, School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi’an, 710021, People’s Republic of China
- Xi’an Key Laboratory of Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi’an, 710021, People’s Republic of China
| | - Xiaolin Yao
- School of Food Science and Engineering, School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi’an, 710021, People’s Republic of China
| | - Sijuan Wang
- School of Food Science and Engineering, School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi’an, 710021, People’s Republic of China
- Xi’an Key Laboratory of Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi’an, 710021, People’s Republic of China
| | - Kaiqiang Lv
- School of Food Science and Engineering, School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi’an, 710021, People’s Republic of China
- Xi’an Key Laboratory of Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi’an, 710021, People’s Republic of China
| | - Guangwen Luo
- School of Food Science and Engineering, School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi’an, 710021, People’s Republic of China
- Xi’an Key Laboratory of Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi’an, 710021, People’s Republic of China
| | - Jiaqi Wang
- School of Food Science and Engineering, School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi’an, 710021, People’s Republic of China
- Xi’an Key Laboratory of Antiviral and Antimicrobial-Resistant Bacteria Therapeutics Research, Xi’an, 710021, People’s Republic of China
| | - Guoliang Li
- School of Food Science and Engineering, School of Biological and Pharmaceutical Sciences, Shaanxi University of Science & Technology, Xi’an, 710021, People’s Republic of China
| |
Collapse
|
15
|
Prasad S, Cross RK, Monroe MB, Dolinger MT, Motte R, Hong S, Stidham RW, Kumar N, Levine D, Larijani A, Simone A, Chachu KA, Wyborski R, Heller CA, Moss AC, Schwerbrock NMJ, Selaru FM. Challenges in IBD Research 2024: Novel Technologies. Inflamm Bowel Dis 2024; 30:S30-S38. [PMID: 38778625 DOI: 10.1093/ibd/izae082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Indexed: 05/25/2024]
Abstract
Novel technology is one of the five focus areas of the Challenges in Inflammatory Bowel Disease (IBD) Research 2024 document. Building off the Challenges in IBD Research 2019 document, the Foundation aims to provide a comprehensive overview of current gaps in IBD research and deliver actionable approaches to address them with a focus on how these gaps can lead to advancements in interception, remission, and restoration for these diseases. The document is the result of a multidisciplinary collaboration from scientists, clinicians, patients, and funders and represents a valuable resource for patient-centric research prioritization. Specifically, the Novel Technologies section focuses on addressing key research gaps to enable interception and improve remission rates in IBD. This includes testing predictions of disease onset and progression, developing novel technologies tailored to specific phenotypes, and facilitating collaborative translation of science into diagnostics, devices, and therapeutics. Proposed priority actions outlined in the document include real-time measurement of biological changes preceding disease onset, more effective quantification of fibrosis, exploration of technologies for local treatment of fistulas, and the development of drug delivery platforms for precise, location-restricted therapies. Additionally, there is a strong emphasis on fostering collaboration between various stakeholders to accelerate progress in IBD research and treatment. Addressing these research gaps necessitates the exploration and implementation of bio-engineered novel technologies spanning a spectrum from materials to systems. By harnessing innovative ideas and technologies, there's a collective effort to enhance patient care and outcomes for individuals affected by IBD.
Collapse
Affiliation(s)
- Shalini Prasad
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Raymond K Cross
- Director of the Inflammatory Bowel Disease Program, University of Maryland School of Medicine, Maryland, MD, USA
| | - Mary Beth Monroe
- Department of Biomedical and Chemical Engineering BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Michael T Dolinger
- Icahn School of Medicine at Mount Sinai, Division of Pediatric Gastroenterology, New York, NY, USA
| | - Rachel Motte
- TISSIUM, 74 Rue du Faubourg Saint-Antoine, Paris, France
| | - Sungmo Hong
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Ryan W Stidham
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Narendra Kumar
- Department of Pharmaceutical Science, ILR-College of Pharmacy, Texas A&M University, TX, USA
| | - Deborah Levine
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Anthony Larijani
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Ashley Simone
- Department of Bioengineering, The University of Texas at Dallas, Richardson, TX, USA
| | - Karen A Chachu
- Department of Medicine, Division of Gastroenterology, Duke University School of Medicine, Durham, NC, USA
| | | | - Caren A Heller
- Members of the Crohn's & Colitis Foundation, New York, NY, USA
| | - Alan C Moss
- Members of the Crohn's & Colitis Foundation, New York, NY, USA
| | | | - Florin M Selaru
- Division of Gastroenterology, Oncology and Biomedical Engineering, Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
16
|
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
|
17
|
Zhu L, Yu T, Wang W, Xu T, Geng W, Li N, Zan X. Responsively Degradable Nanoarmor-Assisted Super Resistance and Stable Colonization of Probiotics for Enhanced Inflammation-Targeted Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308728. [PMID: 38241751 DOI: 10.1002/adma.202308728] [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: 08/28/2023] [Revised: 12/09/2023] [Indexed: 01/21/2024]
Abstract
Manipulation of the gut microbiota using oral microecological preparations has shown great promise in treating various inflammatory disorders. However, delivering these preparations while maintaining their disease-site specificity, stability, and therapeutic efficacy is highly challenging due to the dynamic changes associated with pathological microenvironments in the gastrointestinal tract. Herein, a superior armored probiotic with an inflammation-targeting capacity is developed to enhance the efficacy and timely action of bacterial therapy against inflammatory bowel disease (IBD). The coating strategy exhibits suitability for diverse probiotic strains and has negligible influence on bacterial viability. This study demonstrates that these armored probiotics have ultraresistance to extreme intraluminal conditions and stable mucoadhesive capacity. Notably, the HA-functionalized nanoarmor equips the probiotics with inflamed-site targetability through multiple interactions, thus enhancing their efficacy in IBD therapy. Moreover, timely "awakening" of ingested probiotics through the responsive transferrin-directed degradation of the nanoarmor at the site of inflammation is highly beneficial for bacterial therapy, which requires the bacterial cells to be fully functional. Given its easy preparation and favorable biocompatibility, the developed single-cell coating approach provides an effective strategy for the advanced delivery of probiotics for biomedical applications at the cellular level.
Collapse
Affiliation(s)
- Limeng Zhu
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325000, China
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
- College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tiantian Yu
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Wenchao Wang
- Department of Pain, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Tong Xu
- College of Life Science, University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
| | - Wujun Geng
- Department of Pain, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Na Li
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Xingjie Zan
- School of Ophthalmology and Optometry, Eye Hospital, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325000, China
- Wenzhou Key Laboratory of Perioperative Medicine, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| |
Collapse
|
18
|
Zhang L, Ye P, Zhu H, Zhu L, Ren Y, Lei J. Bioinspired and biomimetic strategies for inflammatory bowel disease therapy. J Mater Chem B 2024; 12:3614-3635. [PMID: 38511264 DOI: 10.1039/d3tb02995f] [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: 03/22/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic chronic inflammatory bowel disease with high morbidity and an increased risk of cancer or death, resulting in a heavy societal medical burden. While current treatment modalities have been successful in achieving long-term remission and reducing the risk of complications, IBD remains incurable. Nanomedicine has the potential to address the high toxic side effects and low efficacy in IBD treatment. However, synthesized nanomedicines typically exhibit some degree of immune rejection, off-target effects, and a poor ability to cross biological barriers, limiting the development of clinical applications. The emergence of bionic materials and bionic technologies has reshaped the landscape in novel pharmaceutical fields. Biomimetic drug-delivery systems can effectively improve biocompatibility and reduce immunogenicity. Some bioinspired strategies can mimic specific components, targets or immune mechanisms in pathological processes to produce targeting effects for precise disease control. This article highlights recent research on bioinspired and biomimetic strategies for the treatment of IBD and discusses the challenges and future directions in the field to advance the treatment of IBD.
Collapse
Affiliation(s)
- Limei Zhang
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing 100083, P. R. China.
| | - Peng Ye
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing 100083, P. R. China.
| | - Huatai Zhu
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing 100083, P. R. China.
| | - Liyu Zhu
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing 100083, P. R. China.
| | - Yuting Ren
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing 100083, P. R. China.
| | - Jiandu Lei
- Beijing Key Laboratory of Lignocellulosic Chemistry, Beijing Forestry University, Beijing 100083, P. R. China.
- MOE Engineering Research Center of Forestry Biomass Materials and Bioenergy, Beijing Forestry University, Beijing 100083, P. R. China
| |
Collapse
|
19
|
Li D, Shangguan J, Yu F, Lin G, Pan H, Zhang M, Lin H, Chen B, Xu H, Hu S. Growth Factors-Loaded Temperature-Sensitive Hydrogel as Biomimetic Mucus Attenuated Murine Ulcerative Colitis via Repairing the Mucosal Barriers. ACS APPLIED MATERIALS & INTERFACES 2024; 16:7686-7699. [PMID: 38289234 DOI: 10.1021/acsami.3c15684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2024]
Abstract
The pathogenesis of ulcerative colitis (UC) is associated with the shedding of the gut mucus. Herein, inspired by the biological functions of mucus, growth factors-loaded in situ hydrogel (PHE-EK) was designed for UC treatment by integrating dihydrocaffeic acid-modified poloxamer as a thermosensitive material with hyaluronic acid (colitis-specific adhesive), epigallocatechin-3-gallate (antibacterial agent), and bioactive factors (KPV tripeptide and epidermal growth factor). PHE-EK presented good thermosensitive properties, as a flowable liquid at room temperature and gelled within 10 s when exposed to body temperature. PHE-EK hydrogel presented good mechanical strength with a strain of 77.8%. Moreover, PHE-EK hydrogel displayed antibacterial activity against Escherichia coli. Importantly, in vitro and in vivo adhesive tests showed that the PHE-EK hydrogel could specifically adhere to the inflamed colon via electrostatic interaction. When PHE-EK as a biomimetic mucus was rectally administrated to colitis rats, it effectively hindered the body weight loss, reduced the disease activity index and improved the colonic shorting. Moreover, the expression of pro-inflammatory cytokines (e.g., IL-1β, IL-6, and TNF-α) at the laminae propria or epitheliums of the colon for colitis rats was substantially inhibited by PHE-EK. Besides, the colonic epitheliums were well rearranged, and the tight junction proteins (Zonula-1 and Claudin-5) between them were greatly upregulated after PHE-EK treatment. Collectively, PHE-EK might be a promising therapy for UC.
Collapse
Affiliation(s)
- Dingwei Li
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province 325000, China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Jianxun Shangguan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Fengnan Yu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Gaolong Lin
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Hanxiao Pan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Mengjiao Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Haoran Lin
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Ben Chen
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Helin Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Sunkuan Hu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province 325000, China
| |
Collapse
|
20
|
Liu Y, Huang J, Li S, Li Z, Chen C, Qu G, Chen K, Teng Y, Ma R, Wu X, Ren J. Advancements in hydrogel-based drug delivery systems for the treatment of inflammatory bowel disease: a review. Biomater Sci 2024; 12:837-862. [PMID: 38196386 DOI: 10.1039/d3bm01645e] [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: 01/11/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic disorder that affects millions of individuals worldwide. However, current drug therapies for IBD are plagued by significant side effects, low efficacy, and poor patient compliance. Consequently, there is an urgent need for novel therapeutic approaches to alleviate IBD. Hydrogels, three-dimensional networks of hydrophilic polymers with the ability to swell and retain water, have emerged as promising materials for drug delivery in the treatment of IBD due to their biocompatibility, tunability, and responsiveness to various stimuli. In this review, we summarize recent advancements in hydrogel-based drug delivery systems for the treatment of IBD. We first identify three pathophysiological alterations that need to be addressed in the current treatment of IBD: damage to the intestinal mucosal barrier, dysbiosis of intestinal flora, and activation of inflammatory signaling pathways leading to disequilibrium within the intestines. Subsequently, we discuss in depth the processes required to prepare hydrogel drug delivery systems, from the selection of hydrogel materials, types of drugs to be loaded, methods of drug loading and drug release mechanisms to key points in the preparation of hydrogel drug delivery systems. Additionally, we highlight the progress and impact of the hydrogel-based drug delivery system in IBD treatment through regulation of physical barrier immune responses, promotion of mucosal repair, and improvement of gut microbiota. In conclusion, we analyze the challenges of hydrogel-based drug delivery systems in clinical applications for IBD treatment, and propose potential solutions from our perspective.
Collapse
Affiliation(s)
- Ye Liu
- School of Medicine, Southeast University, Nanjing, 210009, China
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| | - Jinjian Huang
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| | - Sicheng Li
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| | - Ze Li
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| | - Canwen Chen
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| | - Guiwen Qu
- School of Medicine, Southeast University, Nanjing, 210009, China
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| | - Kang Chen
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| | - Yitian Teng
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| | - Rui Ma
- School of Medicine, Southeast University, Nanjing, 210009, China
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| | - Xiuwen Wu
- School of Medicine, Southeast University, Nanjing, 210009, China
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| | - Jianan Ren
- School of Medicine, Southeast University, Nanjing, 210009, China
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
21
|
Zhao Y, Yao Y, Fan S, Shen X, Chai X, Li Z, Zeng J, Pi J, Zhou Z, Huang G, Jin H. Oral delivery of pH-sensitive nanoparticles loaded Celastrol targeting the inflammatory colons to treat ulcerative colitis. J Tissue Eng 2024; 15:20417314241265892. [PMID: 39130681 PMCID: PMC11316965 DOI: 10.1177/20417314241265892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/18/2024] [Indexed: 08/13/2024] Open
Abstract
The incidence of ulcerative colitis (UC) is rapidly rising worldwide. Oral drug delivery system is a promising approach for treating UC, but it often fails to accumulate to the inflammatory lesions, thus, it is impressive to develop a colon-targeted oral delivery system for preventing systemic toxicity and maintaining UC therapeutics. Here, a negative-charged PLGA nanoparticle system was designed to encapsulate celastrol (Cel), and then chitosan and mannose were coated on the surface of the nanoparticles (MC@Cel-NPs) to endow these nanoparticles with the mucosal adsorption and macrophage targeting abilities. MC@Cel-NPs demonstrate excellent resist decomposition ability against the strong acidic gastrointestinal environment, and accumulates in the specific inflammatory sites through the affinity of electrostatic reaction. After releasing the payload, MC@Cel-NPs could remarkably alleviate the colon inflammation, which was evidenced by the decrease in pro-inflammatory cytokines TNF-α, IL-1β, and IL-6 in both blood and colon sections, and scavenging reactive oxygen species (ROS) in colon cells, including macrophage, neutrophil, T cell, and B cell. This nanoparticle system provided a new approach for treating UC through a Chinese herbal ingredient-related oral delivery manner.
Collapse
Affiliation(s)
- Yue Zhao
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Yinlian Yao
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Shilong Fan
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Xin Shen
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Xingxing Chai
- Laboratory Animal Center, Guangdong Medical University, Dongguan, China
| | - Zimin Li
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Jiachun Zeng
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Jiang Pi
- School of Medical Technology, Guangdong Medical University, Dongguan, China
| | - Zhikun Zhou
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Gonghua Huang
- Guangdong Provincial Key Laboratory of Medical Immunology and Molecular Diagnostics, Guangdong Medical University, Dongguan, China
| | - Hua Jin
- School of Pharmacology, Guangdong Medical University, Dongguan, China
| |
Collapse
|
22
|
Xu Y, Wang XC, Jiang W, Chen LH, Chen T, Wu D, Hu JN. Porphyra haitanensis polysaccharide-functionalized selenium nanoparticles for effective alleviation of ulcerative colitis. Int J Biol Macromol 2023; 253:127570. [PMID: 37866556 DOI: 10.1016/j.ijbiomac.2023.127570] [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: 07/06/2023] [Revised: 10/07/2023] [Accepted: 10/18/2023] [Indexed: 10/24/2023]
Abstract
Exacerbated intestinal inflammation, oxidative stress imbalance, and damage to intestinal mucosal barrier are closely related to the pathogenesis and progression of ulcerative colitis (UC). Selenium nanoparticles (Se NPs) have demonstrated promising potential to alleviate UC symptoms, however, their poor solubility and stability leading to aggregation and large precipitates have significantly limit their clinical application. In this study, we aimed to enhance the performance of Se NPs by functionalizing them with Porphyra haitanensis polysaccharide, yielding PHP-Se NPs. As expected, these PHP-Se NPs exhibited reduced particle size (70.51 ± 2.92 nm), enhanced cellular uptake compared to native Se NPs, and preferential accumulation in the colonic tissue, providing targeted UC treatment. In vivo animal experiments revealed that PHP-Se NPs significantly improved weight loss, shortened colon length, and higher disease activity index (DAI) scores in DSS-induced UC mice. Moreover, PHP-Se NPs significantly inhibited the levels of inflammatory factors in colitis tissues and oxidative stress in serum of UC mice, improved histological damage in colitis tissues, and restored the intestinal mucosal barrier. Taken together, our study offers an innovative approach to augment the bioavailability of Se NPs, presenting a promising strategy for the effective prevention and management of UC.
Collapse
Affiliation(s)
- Yu Xu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Xin-Chuang Wang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Wen Jiang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Li-Hang Chen
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Tao Chen
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Di Wu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Jiang-Ning Hu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| |
Collapse
|
23
|
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
|
24
|
Min K, Sahu A, Jeon SH, Tae G. Emerging drug delivery systems with traditional routes - A roadmap to chronic inflammatory diseases. Adv Drug Deliv Rev 2023; 203:115119. [PMID: 37898338 DOI: 10.1016/j.addr.2023.115119] [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: 09/15/2022] [Revised: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 10/30/2023]
Abstract
Inflammation is prevalent and inevitable in daily life but can generally be accommodated by the immune systems. However, incapable self-healing and persistent inflammation can progress to chronic inflammation, leading to prevalent or fatal chronic diseases. This review comprehensively covers the topic of emerging drug delivery systems (DDSs) for the treatment of chronic inflammatory diseases (CIDs). First, we introduce the basic biology of the chronic inflammatory process and provide an overview of the main CIDs of the major organs. Next, up-to-date information on various DDSs and the associated strategies for ensuring targeted delivery and stimuli-responsiveness applied to CIDs are discussed extensively. The implementation of traditional routes of drug administration to maximize their therapeutic effects against CIDs is then summarized. Finally, perspectives on future DDSs against CIDs are presented.
Collapse
Affiliation(s)
- Kiyoon Min
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Abhishek Sahu
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Hajipur, 844102, India
| | - Sae Hyun Jeon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea.
| |
Collapse
|
25
|
Wang X, Zhang M, Li Y, Cong H, Yu B, Shen Y. Research Status of Dendrimer Micelles in Tumor Therapy for Drug Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2304006. [PMID: 37635114 DOI: 10.1002/smll.202304006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/16/2023] [Indexed: 08/29/2023]
Abstract
Dendrimers are a family of polymers with highly branched structure, well-defined composition, and extensive functional groups, which have attracted great attention in biomedical applications. Micelles formed by dendrimers are ideal nanocarriers for delivering anticancer agents due to the explicit study of their characteristics of particle size, charge, and biological properties such as toxicity, blood circulation time, biodistribution, and cellular internalization. Here, the classification, preparation, and structure of dendrimer micelles are reviewed, and the specific functional groups modified on the surface of dendrimers for tumor active targeting, stimuli-responsive drug release, reduced toxicity, and prolonged blood circulation time are discussed. In addition, their applications are summarized as various platforms for biomedical applications related to cancer therapy including drug delivery, gene transfection, nano-contrast for imaging, and combined therapy. Other applications such as tissue engineering and biosensor are also involved. Finally, the possible challenges and perspectives of dendrimer micelles for their further applications are discussed.
Collapse
Affiliation(s)
- Xijie Wang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Min Zhang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Yanan Li
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
- School of Materials Science and Engineering, Shandong University of Technology, Zibo, 255000, China
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
| | - Youqing Shen
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, and Department of, Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| |
Collapse
|
26
|
Zhang X, Yuan Z, Wu J, He Y, Lu G, Zhang D, Zhao Y, Wu R, Lv Y, Cai K, He S. An Orally-Administered Nanotherapeutics with Carbon Monoxide Supplying for Inflammatory Bowel Disease Therapy by Scavenging Oxidative Stress and Restoring Gut Immune Homeostasis. ACS NANO 2023; 17:21116-21133. [PMID: 37843108 DOI: 10.1021/acsnano.3c04819] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Traditional drug-based treatments for inflammatory bowel disease (IBD) have significant limitations due to their potential off-target systemic side-effects. Currently, there is a lack of understanding on how to effectively address excessive oxidative stress, dysregulated immune homeostasis, and microbiota dysbiosis within the IBD microenvironment. Herein, we introduce a nanotherapeutic approach, named LBL-CO@MPDA, for IBD treatment. LBL-CO@MPDA is an orally administered formulation that supplies carbon monoxide (CO) for therapeutic purposes. To create the LBL-CO@MPDA nanocomposite, we developed a layer by layer (LBL) self-assembly strategy where we coated chitosan/alginate polyelectrolytes onto the surface of CO prodrug-loaded mesoporous polydopamine nanoparticles (CO@MPDA). Benefiting from the negatively charged surface of the LBL coating, it allows for targeted accumulation of LBL-CO@MPDA specifically onto the positively charged inflamed colon lesions through electrostatic interactions. Furthermore, in the oxidative microenvironment of the inflamed colon, the nanotherapeutic system releases CO in a responsive manner. Interestingly, CO@MPDA ameliorates inflammatory conditions by MPDA-mediated ROS-scavenging and CO-mediated immunomodulation. CO-supplying activates heme oxygenase-1, leading to macrophage M2 polarization via the Notch/Hes1/Stat3 signaling pathway, while suppressing the inflammatory response by down-regulating the p38 MAPK and NF-κB (p50/p65) signaling pathways. In the mice model of dextran sulfate sodium (DSS)-induced IBD, LBL-CO@MPDA effectively reverses the pro-inflammatory microenvironment and restores gut barrier functions through multiple mechanisms, including scavenging oxidative stress, restoring immune homeostasis, and modulating the gut microbiota. Collectively, our findings highlight the promising potential of this innovative nanotherapeutic strategy for the targeted treatment of IBD.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Zhang Yuan
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Jianshuang Wu
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Ye He
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
| | - Guifang Lu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Dan Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Yan Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Rongqian Wu
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Yi Lv
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an 710061, P. R. China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, 174 Shazheng Street, Shapingba District, Chongqing 400044, P.R. China
| | - Shuixiang He
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| |
Collapse
|
27
|
Fu W, Xu L, Chen Z, Kan L, Ma Y, Qian H, Wang W. Recent advances on emerging nanomaterials for diagnosis and treatment of inflammatory bowel disease. J Control Release 2023; 363:149-179. [PMID: 37741461 DOI: 10.1016/j.jconrel.2023.09.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 09/25/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic idiopathic inflammatory disorder that affects the entire gastrointestinal tract and is associated with an increased risk of colorectal cancer. Mainstream clinical testing methods are time-consuming, painful for patients, and insufficiently sensitive to detect early symptoms. Currently, there is no definitive cure for IBD, and frequent doses of medications with potentially severe side effects may affect patient response. In recent years, nanomaterials have demonstrated considerable potential for IBD management due to their diverse structures, composition, and physical and chemical properties. In this review, we provide an overview of the advances in nanomaterial-based diagnosis and treatment of IBD in recent five years. Multi-functional bio-nano platforms, including contrast agents, near-infrared (NIR) fluorescent probes, and bioactive substance detection agents have been developed for IBD diagnosis. Based on a series of pathogenic characteristics of IBD, the therapeutic strategies of antioxidant, anti-inflammatory, and intestinal microbiome regulation of IBD based on nanomaterials are systematically introduced. Finally, the future challenges and prospects in this field are presented to facilitate the development of diagnosis and treatment of IBD.
Collapse
Affiliation(s)
- Wanyue Fu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, China
| | - Lingling Xu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, China
| | - Zetong Chen
- School of Stomatology, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Lingling Kan
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, China
| | - Yan Ma
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, China.
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, China.
| | - Wanni Wang
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, PR China; Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, Anhui 230012, China.
| |
Collapse
|
28
|
Kotla NG, Langlois JB, Fisch A, Kramer I, Halleux C. Surface modified cationic PLGA microparticles as long-acting injectable carriers for intra-articular small molecule drug delivery. Eur J Pharm Biopharm 2023; 193:S0939-6411(23)00286-2. [PMID: 39491137 DOI: 10.1016/j.ejpb.2023.10.020] [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: 09/18/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2024]
Abstract
Controlled local delivery of therapeutics (small molecule drug crystals or biologics) for knee-associated diseases such as osteoarthritis necessitates patient compliance, ensuring that the injected depot does not trigger local tissue inflammation and immune responses. A local drug delivery strategy that releases drug at a controlled rate while ensuring minimal tolerability issues at the injection site would be an appealing paradigm in intra-articular (IA) therapies. Herein, we report the formulation development and characterization of surface modified PLGA microparticles (MPs) through the surface integration of a cationic lipid, DOTAP (1,2-Dioleoyl-3-trimethylammonium propane). Following IA administration, these particles are able to interact with anionic synovial fluid glycosaminoglycans (GAGs) to form an in-situ surface coating in the knee joint, thereby reducing the depot-associated local inflammatory response. The formulated microparticles were about 10-40 µm in size range, with a +19 to +33 mV overall surface charge after DOTAP lipid surface integration. These particles showed preferential surface adhesion with endogenous anionic GAGs (e.g., hyaluronic acid) due to electrostatic interactions in vitro, and approximately 65% of the model drug triamcinolone acetonide (TCA) was released after 10 weeks in simulated synovial fluid. The uncoated and DOTAP-coated PLGA microparticles had no effect on mouse osteoblast MC3T3 cell viability and human macrophage inflammatory response. Further, DOTAP-coated particles showed a marginal decrease in pro-inflammatory cytokines in naïve rats following knee injection. Together, the results suggest that surface-modified PLGA particles may have promising potential as delivery carriers for long-acting injectables.
Collapse
Affiliation(s)
- Niranjan G Kotla
- Diseases of Aging and Regenerative Medicine, Novartis Institute for BioMedical Research (NIBR), Novartis Pharma AG, Basel 4056, Switzerland.
| | - Jean-Baptiste Langlois
- Diseases of Aging and Regenerative Medicine, Novartis Institute for BioMedical Research (NIBR), Novartis Pharma AG, Basel 4056, Switzerland
| | - Andreas Fisch
- Technical Research and Development (TRD), Novartis Pharma AG, Basel 4056, Switzerland
| | - Ina Kramer
- Diseases of Aging and Regenerative Medicine, Novartis Institute for BioMedical Research (NIBR), Novartis Pharma AG, Basel 4056, Switzerland
| | - Christine Halleux
- Diseases of Aging and Regenerative Medicine, Novartis Institute for BioMedical Research (NIBR), Novartis Pharma AG, Basel 4056, Switzerland
| |
Collapse
|
29
|
Lei P, Yu H, Ma J, Du J, Fang Y, Yang Q, Zhang K, Luo L, Jin L, Wu W, Sun D. Cell membrane nanomaterials composed of phospholipids and glycoproteins for drug delivery in inflammatory bowel disease: A review. Int J Biol Macromol 2023; 249:126000. [PMID: 37532186 DOI: 10.1016/j.ijbiomac.2023.126000] [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: 05/30/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
Inflammatory bowel disease (IBD) is a serious chronic intestinal disorder with an increasing global incidence. However, current treatment strategies, such as anti-inflammatory drugs and probiotics, have limitations in terms of safety, stability, and effectiveness. The emergence of targeted nanoparticles has revolutionized IBD treatment by enhancing the biological properties of drugs and promoting efficiency and safety. Unlike synthetic nanoparticles, cell membrane nanomaterials (CMNs) consist primarily of biological macromolecules, including phospholipids, proteins, and sugars. CMNs include red blood cell membranes, macrophage membranes, and leukocyte membranes, which possess abundant glycoprotein receptors and ligands on their surfaces, allowing for the formation of cell-to-cell connections with other biological macromolecules. Consequently, they exhibit superior cell affinity, evade immune responses, and target inflammation effectively, making them ideal material for targeted delivery of IBD therapies. This review explores various CMNs delivery systems for IBD treatment. However, due to the complexity and harsh nature of the intestinal microenvironment, the lack of flexibility or loss of selectivity poses challenges in designing single CMNs delivery strategies. Therefore, we propose a hierarchically programmed delivery modality that combines CMNs with pH, charge, ROS and ligand-modified responsive nanoparticles. This approach significantly improves delivery efficiency and points the way for future research in this area.
Collapse
Affiliation(s)
- Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Haiyang Yu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiao Du
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Li Luo
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523059, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
30
|
Nyandoro VO, Omolo CA, Ismail EA, Yong L, Govender T. Inflammation-responsive drug delivery nanosystems for treatment of bacterial-induced sepsis. Int J Pharm 2023; 644:123346. [PMID: 37633537 DOI: 10.1016/j.ijpharm.2023.123346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Sepsis, a complication of dysregulated host immune systemic response to an infection, is life threatening and causes multiple organ injuries. Sepsis is recognized by WHO as a big contributor to global morbidity and mortality. The heterogeneity in sepsis pathophysiology, antimicrobial resistance threat, the slowdown in the development of antimicrobials, and limitations of conventional dosage forms jeopardize the treatment of sepsis. Drug delivery nanosystems are promising tools to overcome some of these challenges. Among the drug delivery nanosystems, inflammation-responsive nanosystems have attracted considerable interest in sepsis treatment due to their ability to respond to specific stimuli in the sepsis microenvironment to release their payload in a precise, targeted, controlled, and rapid manner compared to non-responsive nanosystems. These nanosystems posit superior therapeutic potential to enhance sepsis treatment. This review critically evaluates the recent advances in the design of drug delivery nanosystems that are inflammation responsive and their potential in enhancing sepsis treatment. The sepsis microenvironment's unique features, such as acidic pH, upregulated receptors, overexpressed enzymes, and enhanced oxidative stress, that form the basis for their design have been adequately discussed. These inflammation-responsive nanosystems have been organized into five classes namely: Receptor-targeted nanosystems, pH-responsive nanosystems, redox-responsive nanosystems, enzyme-responsive nanosystems, and multi-responsive nanosystems. Studies under each class have been thematically grouped and discussed with an emphasis on the polymers used in their design, nanocarriers, key characterization, loaded actives, and key findings on drug release and therapeutic efficacy. Further, this information is concisely summarized into tables and supplemented by inserted figures. Additionally, this review adeptly points out the strengths and limitations of the studies and identifies research avenues that need to be explored. Finally, the challenges and future perspectives on these nanosystems have been thoughtfully highlighted.
Collapse
Affiliation(s)
- Vincent O Nyandoro
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa; Department of Pharmaceutical Chemistry and Pharmaceutics, School of Pharmacy, Kabarak University, Nakuru, Kenya
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa; Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, Nairobi, Kenya.
| | - Eman A Ismail
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Liu Yong
- Wenzhou Institute, University of Chinese Academy of Sciences (WIUCAS), China
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa.
| |
Collapse
|
31
|
Sun LF, Li MM, Chen Y, Lu WJ, Zhang Q, Wang N, Fang WY, Gao S, Chen SQ, Hu RF. pH/enzyme dual sensitive Gegenqinlian pellets coated with Bletilla striata polysaccharide membranes for the treatment of ulcerative colitis. Colloids Surf B Biointerfaces 2023; 229:113453. [PMID: 37454443 DOI: 10.1016/j.colsurfb.2023.113453] [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: 04/24/2023] [Revised: 06/28/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Gegen Qinlian Decoction, derived from Zhang Zhongjing's Treatise on Typhoid Fever, has been widely used in the treatment of various common diseases, frequently-occurring diseases and difficult and complicated diseases, such as ulcerative colitis. In this study, Bletilla striata polysaccharide (BSP) was innovatively used as a film coating material to prepare Gegen Qinlian pellets with dual sensitivity of pH enzyme for the treatment of ulcerative colitis. BSP has the ability to repair the inflamed colon mucosa and can produce synergistic effects, while avoiding the adverse therapeutic effects caused by the early release of drugs from a single pH-sensitive pellets in the small intestine. The prepared pellets have a uniform particle size, good roundness, a particle size range from 0.8 mm to 1.0 mm, and a particle yield is 85.6 %. The results of in vitro release showed that ES-BSP pellets hardly released drugs in the pH range of 1.2-6.8. However, in the colon mimic fluid containing specific enzymes, the drug release was significantly accelerated, demonstrating the sensitivity of the pellets to pH enzymes. In vivo and ex vivo fluorescence imaging of small animals showed that Gegen Qinlian pellets with dual sensitivity of pH enzyme remained longer in the colon compared with pH-sensitive pellets. In vivo pharmacodynamics study showed that the Gegen Qinlian pellets with dual sensitivity of pH enzyme had a better therapeutic effect in the rat model of the ulcerative colon than the commercially available Gegenqinlian pellets in the control group.
Collapse
Affiliation(s)
- Ling Feng Sun
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application,MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials,Key Laboratory of Xin'an Medicine ,the Ministry of Education Anhui Province Key Laboratory of Chinese Medicinal Formula,Anhui University of Chinese Medicine, Hefei, Anhui 230038, China.; Plant Active Peptide Function Food Innovative Manufacturing Industry Innovation Team, Hefei, Anhui 230038, China
| | - Man Man Li
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application,MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials,Key Laboratory of Xin'an Medicine ,the Ministry of Education Anhui Province Key Laboratory of Chinese Medicinal Formula,Anhui University of Chinese Medicine, Hefei, Anhui 230038, China.; Plant Active Peptide Function Food Innovative Manufacturing Industry Innovation Team, Hefei, Anhui 230038, China
| | - Yuan Chen
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application,MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials,Key Laboratory of Xin'an Medicine ,the Ministry of Education Anhui Province Key Laboratory of Chinese Medicinal Formula,Anhui University of Chinese Medicine, Hefei, Anhui 230038, China.; Plant Active Peptide Function Food Innovative Manufacturing Industry Innovation Team, Hefei, Anhui 230038, China
| | - Wen Jie Lu
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Qing Zhang
- Department of Pharmacy, School of Pharmacy, Nanjing Medical University Nanjing, Jiangsu, 210009, China
| | - Nan Wang
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application,MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials,Key Laboratory of Xin'an Medicine ,the Ministry of Education Anhui Province Key Laboratory of Chinese Medicinal Formula,Anhui University of Chinese Medicine, Hefei, Anhui 230038, China.; Plant Active Peptide Function Food Innovative Manufacturing Industry Innovation Team, Hefei, Anhui 230038, China
| | - Wen You Fang
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application,MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials,Key Laboratory of Xin'an Medicine ,the Ministry of Education Anhui Province Key Laboratory of Chinese Medicinal Formula,Anhui University of Chinese Medicine, Hefei, Anhui 230038, China.; Plant Active Peptide Function Food Innovative Manufacturing Industry Innovation Team, Hefei, Anhui 230038, China
| | - Song Gao
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application,MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials,Key Laboratory of Xin'an Medicine ,the Ministry of Education Anhui Province Key Laboratory of Chinese Medicinal Formula,Anhui University of Chinese Medicine, Hefei, Anhui 230038, China.; Plant Active Peptide Function Food Innovative Manufacturing Industry Innovation Team, Hefei, Anhui 230038, China.
| | - Sheng Qi Chen
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application,MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials,Key Laboratory of Xin'an Medicine ,the Ministry of Education Anhui Province Key Laboratory of Chinese Medicinal Formula,Anhui University of Chinese Medicine, Hefei, Anhui 230038, China.; Plant Active Peptide Function Food Innovative Manufacturing Industry Innovation Team, Hefei, Anhui 230038, China.
| | - Rong Feng Hu
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application,MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials,Key Laboratory of Xin'an Medicine ,the Ministry of Education Anhui Province Key Laboratory of Chinese Medicinal Formula,Anhui University of Chinese Medicine, Hefei, Anhui 230038, China.; Plant Active Peptide Function Food Innovative Manufacturing Industry Innovation Team, Hefei, Anhui 230038, China.
| |
Collapse
|
32
|
Li D, Li J, Chen T, Qin X, Pan L, Lin X, Liang W, Wang Q. Injectable Bioadhesive Hydrogels Scavenging ROS and Restoring Mucosal Barrier for Enhanced Ulcerative Colitis Therapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:38273-38284. [PMID: 37530040 DOI: 10.1021/acsami.3c06693] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
Despite the progress in the therapy of ulcerative colitis (UC), long-lasting UC remission can hardly be achieved in the majority of UC patients. The key pathological characteristics of UC include an impaired mucosal barrier and local inflammatory infiltration. Thus, a two-pronged approach aiming at repairing damaged mucosal barrier and scavenging inflammatory mediators simultaneously might hold great potential for long-term remission of UC. A rectal formulation can directly offer preferential and effective drug delivery to inflamed colon. However, regular intestinal peristalsis and frequent diarrhea in UC might cause transient drug retention. Therefore, a bioadhesive hydrogel with strong interaction with intestinal mucosa might be preferable for rectal administration to prolong drug retention. Here, we designed a bioadhesive hydrogel formed by the cross-linking of sulfhydryl chondroitin sulfate and polydopamine (CS-PDA). The presence of PDA would ensure the mucosa-adhesive behavior, and the addition of CS in the hydrogel network was expected to achieve the restoration of the intestinal epithelial barrier. To scavenge the key player (excessive reactive oxygen species, ROS) in inflamed colon, sodium ferulic (SF), a potent ROS inhibitor, was incorporated into the CS-PDA hydrogel. After rectal administration, the SF-loaded CS-PDA hydrogel could adhere to the colonic mucosa to allow prolonged drug retention. Subsequently, sustained SF release could be achieved to persistently scavenge ROS in inflammatory areas. Meanwhile, the presence of CS would promote the restoration of the mucosal barrier. Ultimately, scavenging ROS and restoring the mucosal barrier could be simultaneously achieved via this SF-loaded bioadhesive hydrogel scaffold. Our two-pronged approach might provide new insight for effective UC treatment.
Collapse
Affiliation(s)
- Daming Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jiao Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Tao Chen
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xianyan Qin
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Lihua Pan
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xin Lin
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education and School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Wenlang Liang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Qin Wang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
33
|
Wu C, Lu N, Peng L, Lin M, Bai Y, Lu M, Deng J, Wang J. Regulation of inflammatory macrophages by oral mineralized metal-organic framework nanoparticles for the synergistic treatment of ulcerative colitis and liver injury. CHEMICAL ENGINEERING JOURNAL 2023; 468:143655. [DOI: 10.1016/j.cej.2023.143655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
|
34
|
Swastha D, Varsha N, Aravind S, Samyuktha KB, Yokesh MM, Balde A, Ayilya BL, Benjakul S, Kim SK, Nazeer RA. Alginate-based drug carrier systems to target inflammatory bowel disease: A review. Int J Biol Macromol 2023:125472. [PMID: 37336375 DOI: 10.1016/j.ijbiomac.2023.125472] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/06/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023]
Abstract
Inflammatory bowel disease (IBD) is an inflammatory disorder that affects the gastrointestinal tract. IBD has become an increasingly common condition in both developed and developing nations over the last few decades, owing to a variety of factors like a rising population and diets packed with processed and junk foods. While the root pathophysiology of IBD is unknown, treatments are focused on medications aimed to mitigate symptoms. Alginate (AG), a marine-derived polysaccharide, is extensively studied for its biocompatibility, pH sensitivity, and crosslinking nature. This polymer is thoroughly researched in drug delivery systems for IBD treatment, as it is naturally available, non-toxic, cost effective, and can be easily and safely cross-linked with other polymers to form an interconnected network, which helps in controlling the release of drugs over an extended period. There are various types of drug delivery systems developed from AG to deliver therapeutic agents; among them, nanotechnology-based systems and hydrogels are popular due to their ability to facilitate targeted drug delivery, reduce dosage, and increase the therapeutic efficiency. AG-based carrier systems are not only used for the sustained release of drug, but also used in the delivery of siRNA, interleukins, and stem cells for site directed drug delivery and tissue regenerating ability respectively. This review is focussed on pathogenesis and currently studied medications for IBD, AG-based drug delivery systems and their properties for the alleviation of IBD. Moreover, future challenges are also be discoursed to improve the research of AG in the field of biopharmaceuticals and drug delivery.
Collapse
Affiliation(s)
- Dinakar Swastha
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India
| | - Nambolan Varsha
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India
| | - Suresh Aravind
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India
| | - Kavassery Balasubramanian Samyuktha
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India
| | - Muruganandam Mohaneswari Yokesh
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India
| | - Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India
| | - Bakthavatchalam Loganathan Ayilya
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India
| | - Soottawat Benjakul
- Department of Food Technology, Faculty of Agro-Industry, Prince of Songkhla University, 90112 Hat Yai, Songkhla, Thailand
| | - Se-Kwon Kim
- Department of Marine Science and Convergence Engineering, Hanyang University, Ansan, 11558, Gyeonggi-do, South Korea
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRMInstitute of Science and Technology, Kattankulathur, Chennai, 603203, Tamilnadu, India.
| |
Collapse
|
35
|
Li J, Pu Y, Li S, He B, Chen J. Orally Administrated Olsalazine-Loaded Multilayer Pectin/Chitosan/Alginate Composite Microspheres for Ulcerative Colitis Treatment. Biomacromolecules 2023; 24:2250-2263. [PMID: 37068182 DOI: 10.1021/acs.biomac.3c00146] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2023]
Abstract
The pathogenesis of inflammatory bowel diseases (IBDs) including ulcerative colitis (UC) and Crohn's disease is extremely cloudy. Maintaining the level of remission lesions in colitis is the default treatment attitude at present. Epithelial barrier restoration is considered as the same important strategy as colonic targeted drug delivery in UC treatment. In this paper, we developed a multilayer natural polysaccharide microsphere (pectin/chitosan/alginate) with pH and enzyme dual sensitivity to reduce the loss of medication in the upper digestive tract and preferentially adhere to exposed epithelial cells in colonic tissues by electrostatic forces for efficiently targeted UC treatment. Olsalazine as an inflammatory drug was efficiently loaded in the chitosan layer and realized a colonic pH-responsive drug release. Furthermore, the multilayer microspheres exhibited excellent capability in suppressing harmful flora and a bio-adhesion effect to extend the duration of local medicine. In the in vivo anti-colitis study, the downregulated levels of pro-inflammatory factors and the increase of tight junction protein indicated the excellent anti-inflammation effect of the olsalazine-loaded microspheres. In summary, these results showed that the multilayer natural polysaccharide microspheres could be a powerful candidate in the targeted drug delivery system for UC therapy.
Collapse
Affiliation(s)
- Jiaying Li
- College of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Sai Li
- College of Chemical Engineering, Sichuan University, Chengdu 610065, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Jianlin Chen
- School of Laboratory Medicine, Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu 610500, China
| |
Collapse
|
36
|
Sun Q, Chen J, Zhao Q, He Z, Tang L, Pu Y, He B. Bio-adhesive and ROS-scavenging hydrogel microspheres for targeted ulcerative colitis therapy. Int J Pharm 2023; 639:122962. [PMID: 37068716 DOI: 10.1016/j.ijpharm.2023.122962] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/24/2023] [Accepted: 04/11/2023] [Indexed: 04/19/2023]
Abstract
Ulcerative colitis (UC) as an important type of inflammatory bowel disease is a chronic disease characterized by intestinal dyshomeostasis. The UC treatment is challenged by the insufficiency of drug delivery and retention. Herein, we fabricated an intrarectal formulation of olsalazine (Olsa)-loaded hydrogel microspheres (LDKT/Olsa) with good bio-adhesiveness and reactive oxygen species (ROS)-scavenging ability to enhance drug retention and therapeutic effect. Low methoxy pectin-dopamine conjugate/konjac glucomannan composite hydrogel microspheres (LDKT) with a size ranging from 10 to 100 μm were prepared by using Zn2+ and ROS-sensitive thioketal as crosslinkers. Upon intrarectal administration, the negatively charged and dopamine-functionalized hydrogel microspheres efficiently adhered to cationic surface of inflammatory mucosa, scavenging ROS and releasing Zn2+ and Olsa for antibacterial and anti-inflammatory effects. In the dextran sodium sulfate (DSS)-induced mouse UC model, the microspheres significantly reduced the levels of colonic ROS and pro-inflammatory cytokines, improved gut mucosal barrier integrity, and remarkably relieved colitis. Overall, the LDKT microspheres are promising carriers to deliver drugs for UC treatment.
Collapse
Affiliation(s)
- Qiqi Sun
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Jun Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Quan Zhao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Ziyun He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Lei Tang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
37
|
Sun W, Chen Y, Wang L, Wang Z, Liu S, Zhang M, Liu Y, Li Q, Zhang H. Gram-scale preparation of quercetin supramolecular nanoribbons for intestinal inflammatory diseases by oral administration. Biomaterials 2023; 295:122039. [PMID: 36791522 DOI: 10.1016/j.biomaterials.2023.122039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 01/20/2023] [Accepted: 02/08/2023] [Indexed: 02/11/2023]
Abstract
Gastrointestinal (GI) tract, which possesses the largest surface area of mucosa in the body, is easily suffered from inflammatory damages under the exposure of external stimulations. Excessive reactive oxygen species (ROS) production and continuous oxidative stress in intestines can elicit local mucosal injury, accelerate mucosal ulceration, and amplify the inflammatory response. Thereby, antioxidant therapy is a potential strategy against intestinal inflammatory diseases. Herein, we demonstrate the gram-scale preparation of quercetin supramolecular nanoribbons (SNRs) by using free quercetin molecules as the sole building block for preventing and treating intestinal inflammatory diseases. Unlike current clinical medicines, which mainly confront with poor response and severe adverse effects via bloodstream delivery, our quercetin SNRs possess an excellent antioxidant activity in the harsh environments of GI tract, a relative long retention time in GI tract, an admirable metabolism in GI tract without burdening other organs, and a specific adhesion to the inflamed intestinal epithelium via electrostatic interactions. These advantages strongly guarantee the applications of quercetin SNRs as oral medicines for intestinal inflammatory diseases. After establishing the models of intestinal inflammatory diseases caused by irradiation and drug stimulations, our quercetin SNRs exhibit the promising protective and therapeutic effects for radiation-induced acute enteritis and dextran sulfate sodium (DSS)-induced acute colitis. Because the super easy and fast preparation procedure and the nearly 100% loading capacity of quercetin SNRs, the current work provides a supramolecular nanomedicine with great clinical translation potential against intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Wei Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Yingxuan Chen
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130021, PR China
| | - Liang Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Ze Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Shuwei Liu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130021, PR China
| | - Mengzhu Zhang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130021, PR China
| | - Yi Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China.
| | - Quanshun Li
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun, 130021, PR China.
| | - Hao Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, 130012, PR China; Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Changchun, 130021, PR China; Green Catalysis Center, College of Chemistry, Zhengzhou University, Zhengzhou, 450001, PR China
| |
Collapse
|
38
|
Kotla NG, Rochev Y. IBD disease-modifying therapies: insights from emerging therapeutics. Trends Mol Med 2023; 29:241-253. [PMID: 36720660 DOI: 10.1016/j.molmed.2023.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/19/2022] [Accepted: 01/05/2023] [Indexed: 02/01/2023]
Abstract
Inflammatory bowel disease (IBD) pathogenesis is associated with gut mucosal inflammation, epithelial damage, and dysbiosis leading to a dysregulated gut mucosal barrier. However, the extent and underlying mechanisms remain largely unknown. Current treatment regimens have focused mainly on treating IBD symptoms; however, such treatment strategies do not address mucosal epithelial repair, barrier homeostasis, or intestinal dysbiosis. Although attempts have been made to identify new therapeutic modalities to enhance gut barrier functions, these are at an early developmental stage and have not been wholly successful. We review conventional therapies, the possible relevant role of gut barrier-protecting agents, and biomaterial strategies relating to combination therapies that may pave the way towards developing new therapeutic approaches for IBD.
Collapse
Affiliation(s)
- Niranjan G Kotla
- CÚRAM, Science Foundation Ireland (SFI) Research Centre for Medical Devices, University of Galway, Galway, Ireland.
| | - Yury Rochev
- CÚRAM, Science Foundation Ireland (SFI) Research Centre for Medical Devices, University of Galway, Galway, Ireland.
| |
Collapse
|
39
|
Liu J, Lu R, Zheng X, Hou W, Wu X, Zhao H, Wang G, Tian T. Establishment of a gut-on-a-chip device with controllable oxygen gradients to study the contribution of Bifidobacterium bifidum to inflammatory bowel disease. Biomater Sci 2023; 11:2504-2517. [PMID: 36779280 DOI: 10.1039/d2bm01490d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Supplemental Bifidobacterium has been shown to aid in the prevention, alleviation, and treatment of inflammatory bowel disease (IBD), but the progression and mechanisms are largely unstudied, partly because of a lack of appropriate models. In vitro human gut models must accurately recreate oxygen concentration gradients consistent with those in vivo to mimic gene expression, metabolism, and host-microbiome interactions. A non-equipment-intensive and inexpensive method for constructing the gut-on-a-chip with physiological oxygen concentration gradients remains challenging. Here, we propose a simple strategy using numerical simulations in a dual-channel gut-on-a-chip to guide chip design and achieve controllable oxygen gradients. By varying the size of microchannels, blocking the oxygen penetration of the polydimethylsiloxane layer at a given location, and controlling the flow of hypoxic/aerobic media, this strategy creates steep gradients across the intestinal epithelium. IBD symptoms were induced on the chip by tumor necrosis factor-α and lipopolysaccharide treatment. Bifidobacterium bifidum has been validated to contribute to the stability of the intestinal epithelial barrier, including preventing epithelial barrier disruption and promoting the repair of damaged intestinal epithelial cell monolayers. These effects may be associated with the co-localization of Bifidobacterium bifidum and ZO-1. This simple but robust approach for designing microfluidic devices is applicable to various organs-on-chips in which fluid dynamics and concentration profiles between different media must be considered. With the customized chip, the integration of activated Bifidobacterium bifidum provides an initial step toward developing a multi-factorial IBD platform. The approach could be scaled up for disease modeling, high-throughput drug screening and personalized medicine.
Collapse
Affiliation(s)
- Jun Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Ronghao Lu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Xiaolin Zheng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Wensheng Hou
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Xiaoying Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Hezhao Zhao
- Department of Gastrointestinal Surgery, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China
| | - Guixue Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| | - Tian Tian
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
40
|
Sardou HS, Vosough PR, Abbaspour M, Akhgari A, Sathyapalan T, Sahebkar A. A review on curcumin colon-targeted oral drug delivery systems for the treatment of inflammatory bowel disease. Inflammopharmacology 2023; 31:1095-1105. [PMID: 36757584 DOI: 10.1007/s10787-023-01140-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 01/19/2023] [Indexed: 02/10/2023]
Abstract
Synthetic drugs and monoclonal antibodies are the typical treatments to combat inflammatory bowel disease (IBD). However, side effects are present when these treatments are used, and their continued application could be restricted by the high relapse rate of the disease. One potential alternative to these treatments is the use of plant-derived products. The use curcumin is one such treatment option that has seen an increase in usage in treating IBD. Curcumin is derived from a rhizome of turmeric (Curcuma longa), and the results of studies on the use of curcumin to treat IBD are promising. These studies suggest that curcumin interacts with cellular targets such as NF-κB, JAKs/STATs, MAPKs, TNF-α, IL-6, PPAR, and TRPV1 and may reduce the progression of IBD. Potentially, curcumin can be used as a therapeutic agent for patients with IBD when it reduces the incidence of clinical relapse. This review discusses the strategies utilized in designing and developing an oral colonic delivery dosage form of curcumin.
Collapse
Affiliation(s)
- Hossein Shahdadi Sardou
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Paria Rahnama Vosough
- Food Science and Technology Department, Agriculture Faculty, Ferdowsi University of Mashhad (FUM), Mashhad, Iran
| | - Mohammadreza Abbaspour
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abbas Akhgari
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Thozhukat Sathyapalan
- Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran. .,Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
41
|
Kotla NG, Pandey A, Vijaya Kumar Y, Ramazani F, Fisch A. Polyester-based long acting injectables: Advancements in molecular dynamics simulation and technological insights. Drug Discov Today 2023; 28:103463. [PMID: 36481584 DOI: 10.1016/j.drudis.2022.103463] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/21/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Long-acting injectable (LAI) delivery technologies have enabled the development of several pharmaceutical products that improve patient health by delivering therapeutics from weeks to months. Over the last decade, due to its good biocompatibility, formulation tunability, wide range of degradation rates, and extensive clinical studies, polyester-based LAI technologies including poly(lactic-co-glycolic acid) (PLGA) have made substantial progress. Herein, we discuss PLGA properties with seminal approaches in the development of LAIs, the role of molecular dynamic simulations of polymer-drug interactions, and their effects on quality attributes. We also outline the landscape of various advanced PLGA-based and a few non-PLGA LAI technologies; their design, delivery, and challenges from laboratory scale to preclinical and clinical use; and commercial products incorporating the importance of end-user preferences.
Collapse
Affiliation(s)
- Niranjan G Kotla
- Novartis Institutes for Biomedical Research (NIBR), Novartis Pharma AG, Basel 4002, Switzerland.
| | - Abhijeet Pandey
- Technical Research and Development, Novartis Pharma AG, Hyderabad 500081, India.
| | - Y Vijaya Kumar
- Technical Research and Development, Novartis Pharma AG, Hyderabad 500081, India
| | - Farshad Ramazani
- Technical Research and Development (TRD), Novartis Pharma AG, Basel 4002, Switzerland
| | - Andreas Fisch
- Technical Research and Development (TRD), Novartis Pharma AG, Basel 4002, Switzerland
| |
Collapse
|
42
|
Pu Y, Fan X, Zhang Z, Guo Z, Pan Q, Gao W, Luo K, He B. Harnessing polymer-derived drug delivery systems for combating inflammatory bowel disease. J Control Release 2023; 354:1-18. [PMID: 36566845 DOI: 10.1016/j.jconrel.2022.12.044] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
The inflammatory bowel disease (IBD) is incurable, chronic, recrudescent disorders in the inflamed intestines. Current clinic treatments are challenged by systemic exposure-induced severe side effects, inefficiency after long-term treatment, and increased risks of infection and malignancy due to immunosuppression. Fortunately, naturally bioactive small molecules, reactive oxygen species scavengers (or antioxidants), and gut microbiota modulators have emerged as promising candidates for the IBD treatment. Polymeric systems have been engineered as a delivery vehicle to improve the bioavailability and efficacy of these therapeutic agents through targeting the mucosa and enhancing intestinal adhesion and retention, and reduce their systemic toxicity. Herein we survey polymer-derived drug delivery systems for combating the IBD. Advanced delivery technologies, therapeutic intervention strategies, and the principles for the construction of hierarchical, mucosa-targeting, and bioresponsive systems are elaborated, providing insights into design and development of from-bench-to-bedside drug delivery polymeric systems for the IBD treatment.
Collapse
Affiliation(s)
- Yuji Pu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xi Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Zhuangzhuang Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Zhaoyuan Guo
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Qingqing Pan
- School of Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Wenxia Gao
- College of Chemistry & Materials Engineering, Wenzhou University, Wenzhou 325027, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital, Functional and Molecular Imaging Key Laboratory of Sichuan Province, Sichuan University, Chengdu 610041, China
| | - Bin He
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China.
| |
Collapse
|
43
|
Li R, Fan Y, Liu L, Ma H, Gong D, Miao Z, Wang H, Zha Z. Ultrathin Hafnium Disulfide Atomic Crystals with ROS-Scavenging and Colon-Targeting Capabilities for Inflammatory Bowel Disease Treatment. ACS NANO 2022; 16:15026-15041. [PMID: 36037406 DOI: 10.1021/acsnano.2c06151] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The exciting success of NBTXR3 in the clinic has triggered a tumult of activities in the design and development of hafnium-based nanoparticles. However, due to the concerns of nondegradation and limited functions, the biomedical applications of Hf-based nanoparticles mainly focus on tumors. Herein, tannic acid capped hafnium disulfide (HfS2@TA) nanosheets, a 2D atomic crystal of hafnium-based materials prepared by liquid-phase exfoliation, were explored as high-performance anti-inflammatory nanoagents for the targeted therapy of inflammatory bowel disease (IBD). Benefiting from the transformation of the S2-/S6+ valence state and huge specific surface area, the obtained HfS2@TA nanosheets were not only capable of effectively eliminating reactive oxygen species/reactive nitrogen species and downregulating pro-inflammatory factors but also could be excreted via kidney and hepatointestinal systems. Unexpectedly, HfS2@TA maintained excellent targeting capability to an inflamed colon even in the harsh digestive tract environment, mainly attributed to the electrostatic interactions between negatively charged tannic acid and positively charged inflamed epithelium. Encouragingly, upon oral or intravenous administration, HfS2@TA quickly inhibited inflammation and repaired the intestinal mucosa barrier in both dextran sodium sulfate and 2,4,6-trinitrobenzenesulfonic acid induced IBD models. This work demonstrated that ultrathin HfS2@TA atomic crystals with enhanced colon accumulation were promising for the targeted therapy of IBD.
Collapse
Affiliation(s)
- Ruoyao Li
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China
| | - Yuanyuan Fan
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230036, People's Republic of China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, People's Republic of China
| | - Lulu Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China
| | - Hongna Ma
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China
| | - Deyan Gong
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China
| | - Zhaohua Miao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China
- Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230026, Anhui, People's Republic of China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei 230036, People's Republic of China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, People's Republic of China
| | - Zhengbao Zha
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230009, People's Republic of China
| |
Collapse
|
44
|
Tian W, Wang H, Zhu Y, Wang Q, Song M, Cao Y, Xiao J. Intervention effects of delivery vehicles on the therapeutic efficacy of 6-gingerol on colitis. J Control Release 2022; 349:51-66. [DOI: 10.1016/j.jconrel.2022.06.058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 10/17/2022]
|