1
|
Chen J, Long Y, Zhou X, Yin Z. AG86 Peptide-modified Reactive Oxygen Species-responsive Polymer Nanovesicles for Transdermal Delivery of Hydrophilic Ergothioneine to Basal Keratinocytes. AAPS PharmSciTech 2025; 26:128. [PMID: 40341492 DOI: 10.1208/s12249-025-03122-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/17/2025] [Indexed: 05/10/2025] Open
Abstract
This study aims to develop a multifunctional polymer nanovesicle system, AG86-β-glucan-poly(methionine-b-carboxybetaine) (AGPMC), which integrates AG86 peptide (a targeting ligand for the α6β4 integrin on basal keratinocytes) and reactive oxygen species (ROS) responsiveness to enhance the transdermal delivery of ergothioneine (EGT). The objective is to overcome the limitations of hydrophilic drug delivery and provide a targeted, ROS-triggered strategy for treating skin lipofuscin. AGPMC was synthesized via RAFT and ROP polymerization and self-assembles into stable nanovesicles (ANVs). Physicochemical characterization confirmed that the unloaded ANVs have an average size of 106.17 ± 3.26 nm and a polydispersity index (PDI) of 0.261 ± 0.027, with excellent stability and clear ROS-triggered structural responsiveness. After EGT loading, the nanovesicles exhibited a size of 140.10 ± 3.33 nm and a PDI of 0.217 ± 0.008. In vitro studies using HaCaT cells demonstrated a significant enhancement in cellular uptake, with an increase of up to 2.2-fold compared to free FSS (p < 0.0001), as well as effective ROS scavenging and minimal cytotoxicity. The endocytosis mechanism study revealed that ANVs are primarily taken up via energy-dependent active transport, with caveolae-mediated endocytosis being the main pathway. Moreover, transdermal delivery studies confirmed that EGT-loaded nanovesicles significantly improved drug penetration through the stratum corneum. This work represents the first application of the AG86-conjugated nanovesicles for keratinocyte-targeted delivery, combined with ROS responsiveness. The "Loading-Targeting-Responding " strategy holds significant potential for enhancing hydrophilic drug delivery and offers a promising approach for treating age-related skin lipofuscin accumulation.
Collapse
Affiliation(s)
- Junlong Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yiqing Long
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Xiaozeliang Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Zongning Yin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
2
|
Ding S, Alexander E, Liang H, Kulchar RJ, Singh R, Herzog RW, Daniell H, Leong KW. Synthetic and Biogenic Materials for Oral Delivery of Biologics: From Bench to Bedside. Chem Rev 2025; 125:4009-4068. [PMID: 40168474 DOI: 10.1021/acs.chemrev.4c00482] [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: 04/03/2025]
Abstract
The development of nucleic acid and protein drugs for oral delivery has lagged behind their production for conventional nonoral routes. Over the past decade, the evolution of DNA- and RNA-based technologies combined with the innovation of state-of-the-art delivery vehicles for nucleic acids has brought rapid advancements to the biopharmaceutical field. Nucleic acid therapies have the potential to achieve long-lasting effects, or even cures, by inhibiting or editing genes, which is not possible with conventional small-molecule drugs. However, challenges and limitations must be addressed before these therapies can provide cures for chronic conditions and rare diseases, rather than only offering temporary relief. Nucleic acids and proteins face premature degradation in the acidic, enzyme-rich stomach environment and are rapidly cleared by the liver. To overcome these challenges, various delivery vehicles have been developed to transport therapeutic compounds to the intestines, where the active compounds are released and gut microbiota and mucosal immune system also play an important role. This review provides a comprehensive overview of the promises and pitfalls associated with the oral route of administration of biologics, current delivery systems, applications of orally delivered therapeutics, and the challenges and considerations for translation of nucleic acid and protein therapeutics into clinical practice.
Collapse
Affiliation(s)
- Suwan Ding
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Elena Alexander
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Huiyi Liang
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Rachel J Kulchar
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Rahul Singh
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Roland W Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| |
Collapse
|
3
|
Luo J, Shi B, Hao C, Sun M, Xu L, Kuang H, Qu A. Chiral Zinc Sulfide Nanoparticles Scavenging Reactive Oxygen Species for Remodeling Intestinal Homeostasis. Angew Chem Int Ed Engl 2025:e202503654. [PMID: 40170506 DOI: 10.1002/anie.202503654] [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: 02/13/2025] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/03/2025]
Abstract
Elevated levels of reactive oxygen species (ROS) and gut microbiota dysbiosis are crucial factors that exacerbate inflammatory bowel disease (IBD). To address this, we successfully synthesized zinc sulfide nanoparticles (ZnS NPs) with a particle size of approximately 500 nm and a maximum g-factor of 0.07, utilizing l-/d-cysteine as chiral ligands. Chirality gives NPs unique bioactivity. These chiral ZnS NPs could enter macrophages through the CD44 and clathrin pathways, which enhanced the ability to scavenge ROS, in turn significantly inhibited the NF-κB and NLRP3 signaling pathways, thereby reducing the secretion of TNF-α, IL-6, and IL-1β, while upregulating IL-10. In vivo experimental data showed that l-ZnS NPs outperformed 5-aminosalicylic acid, significantly improving body weight, reducing the IBD activity index, and attenuating tissue damage. Concurrently, l-ZnS NPs exhibited a marked prophylactic effect. The benchmark studies verified that l-ZnS NPs increased the abundance of the beneficial Lachnospiraceae NK4A136 by 10.55-fold and decreased harmful Enterobacter by 2914.00-fold, thereby reshaping the intestinal microecological balance. Pharmacokinetic and biosafety assessments confirmed the safety of l-ZnS NPs. Our findings indicate that chiral ZnS NPs hold great potential as nanodrugs for the treatment and prevention of IBD, providing an important foundation for the development of IBD therapeutic strategies.
Collapse
Affiliation(s)
- Jun Luo
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Baimei Shi
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Changlong Hao
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Maozhong Sun
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Liguang Xu
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Hua Kuang
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Aihua Qu
- State Key Laboratory of Food Science and Technology, International Joint Research Laboratory for Biointerface and Biodetection, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
4
|
Gong H, Hua Y, Wang Y, Zhang X, Wang H, Zhao Z, Zhang Y. Fabrication of a novel macrophage-targeted biomimetic delivery composite hydrogel with multiple-sensitive properties for tri-modal combination therapy of rheumatoid arthritis. Int J Pharm 2024; 665:124708. [PMID: 39284423 DOI: 10.1016/j.ijpharm.2024.124708] [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: 05/27/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024]
Abstract
In this study, a porous polydopamine (PDA) nanoparticle-decorated β-glucan microcapsules (GMs) nanoplatform (PDA/GMs) were developed with macrophage-targeted biomimetic features and a carriers-within-carriers structure. Indocyanine green (ICG) and catalase (CAT) were subsequently co-encapsulated within the PDA/GMs to create a multifunctional nanotherapeutic agent, termed CIPGs. Furthermore, CIPGs and sinomenine (SIN) were co-loaded within a thermo-sensitive hydrogel to design an injectable delivery system, termed CIPG/SH, with potential for multi-modal therapy of rheumatoid arthritis (RA). Photothermal studies indicated that the CIPGs hold excellent photothermal conversion ability and thermal stability, as they combined the photothermal performance of both PDA and ICG. Meanwhile, the CIPGs displayed favorable oxygen self-supplying and photodynamic performance. The CIPGs showed near-infrared (NIR)-induced phototoxicity, effectively inhibiting macrophage proliferation and displaying remarkable antibacterial activity. In vitro drug release from the prepared CIPG/SH showed a controlled release pattern. Animal experiments conducted on an RA mice model confirmed that the formulated CIPG/SH exhibited significant therapeutic effects. By integrating the biological advantages, photothermal/photodynamic performance of the CIPGs, and controlled drug release performance of the thermo-sensitive hydrogels in a single delivery system, the prepared injectable CIPG/SH represents a novel versatile delivery system with great potential for multi-modal combination targeting therapy in RA.
Collapse
Affiliation(s)
- Haoyang Gong
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Yabing Hua
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Yicheng Wang
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Xinyi Zhang
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Hui Wang
- Department of Pharmacy, Xuzhou Hospital of TCM, Xuzhou 221000, China.
| | - Ziming Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Yanzhuo Zhang
- School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China.
| |
Collapse
|
5
|
Li Y, Liu W, Wang Y, Liu T, Feng Y. Nanotechnology-Mediated Immunomodulation Strategy for Inflammation Resolution. Adv Healthc Mater 2024; 13:e2401384. [PMID: 39039994 DOI: 10.1002/adhm.202401384] [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: 04/16/2024] [Revised: 07/02/2024] [Indexed: 07/24/2024]
Abstract
Inflammation serves as a common characteristic across a wide range of diseases and plays a vital role in maintaining homeostasis. Inflammation can lead to tissue damage and the onset of inflammatory diseases. Although significant progress is made in anti-inflammation in recent years, the current clinical approaches mainly rely on the systemic administration of corticosteroids and antibiotics, which only provide short-term relief. Recently, immunomodulatory approaches have emerged as promising strategies for facilitating the resolution of inflammation. Especially, the advanced nanosystems with unique biocompatibility and multifunctionality have provided an ideal platform for immunomodulation. In this review, the pathophysiology of inflammation and current therapeutic strategies are summarized. It is mainly focused on the nanomedicines that modulate the inflammatory signaling pathways, inflammatory cells, oxidative stress, and inflammation targeting. Finally, the challenges and opportunities of nanomaterials in addressing inflammation are also discussed. The nanotechnology-mediated immunomodulation will open a new treatment strategy for inflammation therapy.
Collapse
Affiliation(s)
- Ying Li
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
| | - Wen Liu
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
| | - Yuanchao Wang
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
| | - Taotao Liu
- Department of Gastroenterology and Hepatology, Characteristic Medical Center of the Chinese People's Armed Police Force, Tianjin Key Laboratory of Hepatopancreatic Fibrosis and Molecular Diagnosis & Treatment, Tianjin, 300162, China
| | - Yakai Feng
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin, 300350, P. R. China
- Frontiers Science Center for Synthetic Biology, Tianjin University, Weijin Road 92, Tianjin, 300072, P. R. China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Weijin Road 92, Tianjin, 300072, P. R. China
| |
Collapse
|
6
|
Song C, Wu J, Wu J, Wang F. MnO 2 and roflumilast-loaded probiotic membrane vesicles mitigate experimental colitis by synergistically augmenting cAMP in macrophage. J Nanobiotechnology 2024; 22:294. [PMID: 38807127 PMCID: PMC11131305 DOI: 10.1186/s12951-024-02558-6] [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: 02/22/2024] [Accepted: 05/16/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is one chronic and relapsing inflammatory bowel disease. Macrophage has been reputed as one trigger for UC. Recently, phosphodiesterase 4 (PDE4) inhibitors, for instance roflumilast, have been regarded as one latent approach to modulating macrophage in UC treatment. Roflumilast can decelerate cyclic adenosine monophosphate (cAMP) degradation, which impedes TNF-α synthesis in macrophage. However, roflumilast is devoid of macrophage-target and consequently causes some unavoidable adverse reactions, which restrict the utilization in UC. RESULTS Membrane vesicles (MVs) from probiotic Escherichia coli Nissle 1917 (EcN 1917) served as a drug delivery platform for targeting macrophage. As model drugs, roflumilast and MnO2 were encapsulated in MVs (Rof&MnO2@MVs). Roflumilast inhibited cAMP degradation via PDE4 deactivation and MnO2 boosted cAMP generation by activating adenylate cyclase (AC). Compared with roflumilast, co-delivery of roflumilast and MnO2 apparently produced more cAMP and less TNF-α in macrophage. Besides, Rof&MnO2@MVs could ameliorate colitis in mouse model and regulate gut microbe such as mitigating pathogenic Escherichia-Shigella and elevating probiotic Akkermansia. CONCLUSIONS A probiotic-based nanoparticle was prepared for precise codelivery of roflumilast and MnO2 into macrophage. This biomimetic nanoparticle could synergistically modulate cAMP in macrophage and ameliorate experimental colitis.
Collapse
Affiliation(s)
- Chengjun Song
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Jiamin Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China
- Institution of Drug R&D, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, China
| | - Fangyu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, 210093, China.
- Department of Gastroenterology and Hepatology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Fei C, Liu L, Qi H, Peng Y, Han J, Wang C, Li X. Curdlan-Decorated Fullerenes Mitigate Immune-Mediated Hepatic Injury for Autoimmune Hepatitis Therapeutics via Reducing Macrophage Infiltration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:5536-5547. [PMID: 38267397 PMCID: PMC10860698 DOI: 10.1021/acsami.3c16168] [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: 10/29/2023] [Revised: 12/23/2023] [Accepted: 01/11/2024] [Indexed: 01/26/2024]
Abstract
Autoimmune hepatitis (AIH) is a severe immune-mediated inflammatory liver disease whose standard of care is immunosuppressive treatment with inevitable undesired outcomes. Macrophage is acknowledged to aggravate liver damage, providing a promising AIH therapeutic target. Accordingly, in this study, a kind of curdlan-decorated fullerene nanoparticle (Cur-F) is fabricated to alleviate immune-mediated hepatic injury for treating AIH via reducing macrophage infiltration in a concanavalin A (Con A)-induced AIH mouse model. After intravenous administration, Cur-F primarily distributes in liver tissues, efficiently eliminates the excessive reactive oxygen species, significantly attenuates oxidative stress, and subsequently suppresses the nuclear factor kappa-B-gene binding (NF-κB) signal pathway, resulting in the lowered production of pro-inflammatory cytokines and the balancing of the immune homeostasis with the prevention of macrophage infiltration in the liver. The regulation of hepatic inflammation contributes to inhibiting inflammatory cytokines-induced hepatocyte apoptosis, decreasing the serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) contents and thus ameliorating immune-mediated hepatic injury. Notably, there is no detectable toxicity to the body. Our findings may open up novel avenues for AIH based on curdlan and fullerene materials.
Collapse
Affiliation(s)
- Chenglong Fei
- Key
Laboratory of Molecular Nanostructure and Nanotechnology, Beijing
National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- School
of Chemistry and Chemical Engineering, Inner
Mongolia University, Inner
Mongolia 010021, China
| | - Lei Liu
- Key
Laboratory of Molecular Nanostructure and Nanotechnology, Beijing
National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Hedong Qi
- Key
Laboratory of Molecular Nanostructure and Nanotechnology, Beijing
National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuyang Peng
- Key
Laboratory of Molecular Nanostructure and Nanotechnology, Beijing
National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingfen Han
- School
of Chemistry and Chemical Engineering, Inner
Mongolia University, Inner
Mongolia 010021, China
| | - Chunru Wang
- Key
Laboratory of Molecular Nanostructure and Nanotechnology, Beijing
National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Xue Li
- Key
Laboratory of Molecular Nanostructure and Nanotechnology, Beijing
National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
10
|
Yang F, Shang S, Qi M, Xiang Y, Wang L, Wang X, Lin T, Hao D, Chen J, Liu J, Wu Q. Yeast glucan particles: An express train for oral targeted drug delivery systems. Int J Biol Macromol 2023; 253:127131. [PMID: 37776921 DOI: 10.1016/j.ijbiomac.2023.127131] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/17/2023] [Accepted: 09/27/2023] [Indexed: 10/02/2023]
Abstract
As an emerging drug delivery vehicle, yeast glucan particles (YGPs) derived from yeast cells could be specifically taken up by macrophages. Therefore, these vehicles could rely on the recruitment of macrophages at the site of inflammation and tumors to enable targeted imaging and drug delivery. This review summarizes recent advances in the application of YGPs in oral targeted delivery systems, covering the basic structure of yeast cells, methods for pre-preparation, drug encapsulation and characterization. The mechanism and validation of the target recognition interaction of YGPs with macrophages are highlighted, and some inspiring cases are presented to show that yeast cells have promising applications. The future chances and difficulties that YGPs will confront are also emphasized throughout this essay. YGPs are not only the "armor" but also the "compass" of drugs in the process of targeted drug transport. This system is expected to provide a new idea about the oral targeted delivery of anti-inflammatory and anti-tumor drugs, and furthermore offer an effective delivery strategy for targeted therapy of other macrophage-related diseases.
Collapse
Affiliation(s)
- Fan Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shang Shang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Mengfei Qi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Yajinjing Xiang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Lingmin Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xinyi Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Tao Lin
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Doudou Hao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jiajia Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Jia Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Qing Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| |
Collapse
|
11
|
Guo J, Zhou B, Niu Y, Liu L, Yang L. Engineered probiotics introduced to improve intestinal microecology for the treatment of chronic diseases: present state and perspectives. J Diabetes Metab Disord 2023; 22:1029-1038. [PMID: 37975092 PMCID: PMC10638336 DOI: 10.1007/s40200-023-01279-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/05/2023] [Indexed: 11/19/2023]
Abstract
Purpose Correcting intestinal microecological imbalance has become one of the core strategies to treat chronic diseases. Some traditional microecology-based therapies targeting intestine, such as prebiotic therapy, probiotic therapy and fecal microbiota transplantation therapy, have been used in the prevention and treatment of clinical chronic diseases, which still facing low safety and poor controllability problems. The development of synthetic biology technology has promoted the development of intestinal microecology-based therapeutics for chronic diseases, which exhibiting higher robustness and controllability, and become an important part of the next generation of microecological therapy. The purpose of this review is to summarize the application of synthetic biology in intestinal microecology-based therapeutics for chronic diseases. Methods The available literatures were searched to find out experimental studies and relevant review articles on the application of synthetic biology in intestinal microecology-based therapeutics for chronic diseases from year 1990 to 2023. Results Evidence proposed that synthetic biology has been applied in the intestinal microecology-based therapeutics for chronic diseases, covering metabolic diseases (e.g. diabetes, obesity, nonalcoholic fatty liver disease and phenylketonuria), digestive diseases (e.g. inflammatory bowel disease and colorectal cancer), and neurodegenerative diseases (e.g. Alzheimer's disease and Parkinson's disease). Conclusion This review summarizes the application of synthetic biology in intestinal microecology-based therapeutics for major chronic diseases and discusses the opportunities and challenges in the above process, providing clinical possibilities of synthetic biology technology applied in microecological therapies.
Collapse
Affiliation(s)
- Jianquan Guo
- Key Laboratory of Coal Environmental Pathogenicity and Prevention, (Shanxi Medical University), Ministry of Education, Taiyuan, PR China
- School of Public Health, Shanxi Medical University, Taiyuan, 030001 Shanxi PR China
| | - Bangyuan Zhou
- School of Public Health, Shanxi Medical University, Taiyuan, 030001 Shanxi PR China
| | - Yali Niu
- School of Public Health, Shanxi Medical University, Taiyuan, 030001 Shanxi PR China
| | - Liangpo Liu
- School of Public Health, Shanxi Medical University, Taiyuan, 030001 Shanxi PR China
| | - Liyang Yang
- School of Basic Medical Sciences, Shanxi University of Chinese Medicine, 030619 Jinzhong, PR China
| |
Collapse
|
12
|
Liu J, Ren H, Zhang C, Li J, Qiu Q, Zhang N, Jiang N, Lovell JF, Zhang Y. Orally-Delivered, Cytokine-Engineered Extracellular Vesicles for Targeted Treatment of Inflammatory Bowel Disease. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2304023. [PMID: 37728188 DOI: 10.1002/smll.202304023] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 09/11/2023] [Indexed: 09/21/2023]
Abstract
The use of orally-administered therapeutic proteins for treatment of inflammatory bowel disease (IBD) has been limited due to the harsh gastrointestinal environment and low bioavailability that affects delivery to diseased sites. Here, a nested delivery system, termed Gal-IL10-EVs (C/A) that protects interleukin 10 (IL-10) from degradation in the stomach and enables targeted delivery of IL-10 to inflammatory macrophages infiltrating the colonic lamina propria, is reported. Extracellular vesicles (EVs) carrying IL-10 are designed to be secreted from genetically engineered mammalian cells by a plasmid system, and EVs are subsequently modified with galactose, endowing the targeted IL-10 delivery to inflammatory macrophages. Chitosan/alginate (C/A) hydrogel coating on Gal-IL10-EVs enables protection from harsh conditions in the gastrointestinal tract and favorable delivery to the colonic lumen, where the C/A hydrogel coating is removed at the diseased sites. Gal-IL10-EVs control the production of reactive oxygen species (ROS) and inhibit the expression of proinflammatory cytokines. In a murine model of colitis, Gal-IL10-EVs (C/A) alleviate IBD symptoms including inflammatory responses and disrupt colonic barriers. Taken together, Gal-IL10-EVs (C/A) features biocompatibility, pH-responsive drug release, and macrophage-targeting as a therapeutic platform for oral delivery of bioactive proteins for treating intestinal diseases.
Collapse
Affiliation(s)
- Jingang Liu
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| | - He Ren
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| | - Chen Zhang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| | - Jiexin Li
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| | - Qian Qiu
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| | - Nan Zhang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| | - Ning Jiang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Jonathan F Lovell
- Department of Biomedical Engineering, The State University of New York at Buffalo, Buffalo, NY, 14260, USA
| | - Yumiao Zhang
- School of Chemical Engineering and Technology, Key Laboratory of Systems Bioengineering (Ministry of Education), Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300350, P. R. China
| |
Collapse
|
13
|
He F, Xie C, Xu X. Hyaluronic acid-modified yeast β-glucan particles delivering doxorubicin for treatment of breast cancer. Carbohydr Polym 2023; 314:120907. [PMID: 37173014 DOI: 10.1016/j.carbpol.2023.120907] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/08/2023] [Accepted: 04/10/2023] [Indexed: 05/15/2023]
Abstract
Breast cancer is one of the most threatening cancers that poses a great risk to women's health. The anti-tumor drug doxorubicin (DOX) is one of commonly used drugs in the treatment of breast cancer. However, the cytotoxicity of DOX has always been an urgent challenge to be solved. In this study, we report an alternative drug delivery system delivering DOX for reducing its physiological toxicity by using the yeast β-glucan particle (YGP) with a hollow and porous vesicle structure. Briefly, amino groups were grafted onto the surface of YGP with the silane coupling agent, then the oxidized hyaluronic acid (OHA) was attached by Schiff base reaction to get HA-modified YGP (YGP@N=C-HA), finally DOX was encapsulated into YGP@N=C-HA to get DOX-loaded YGP@N=C-HA (YGP@N=C-HA/DOX). In vitro release experiments exhibited the pH-responsive DOX release from YGP@N=C-HA/DOX. Cell experiments displayed that YGP@N=C-HA/DOX had good killing effect on both MCF-7 and 4T1 cells and could be internalized into these cells through CD44 receptors, showing targetability to cancer cells. Furthermore, YGP@N=C-HA/DOX could effectively inhibit tumor growth and reduce the physiological toxicity of DOX. Thus, the YGP-based vesicle provides an alternative strategy for lowering the physiological toxicity of DOX in the medical treatment of breast cancer.
Collapse
Affiliation(s)
- Fangzhou He
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430062, China
| | - Xiaojuan Xu
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan 430072, China; Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430062, China; Hubei Engineering Center of Natural Polymer-Based Medical Materials, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
14
|
Wang W, Wu Y, Wang Y, Wang R, Deng C, Yi L, Wang L, He M, Zhou W, Xie Y, Jin Q, Chen Y, Gao T, Zhang L, Xie M. Orally Administrable Aggregation-Induced Emission-Based Bionic Probe for Imaging and Ameliorating Dextran Sulfate Sodium-Induced Inflammatory Bowel Diseases. Adv Healthc Mater 2023; 12:e2202420. [PMID: 36575111 DOI: 10.1002/adhm.202202420] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/12/2022] [Indexed: 12/29/2022]
Abstract
As macrophage infiltration is significantly related to the progression of inflammatory bowel disease (IBD), monitoring the macrophages is a valuable strategy for IBD diagnosis. However, owing to the harsh physiological environment of the gastrointestinal tract and enzymatic degradation, the development of orally administrable imaging probes for tracking macrophages remains a considerable challenge. Accordingly, herein, an orally administrable aggregation-induced emission biomimetic probe (HBTTPIP/β-glucan particles [GPs]) is developed for tracing macrophages; HBTTPIP/GPs can diagnose and alleviate dextran sulfate sodium (DSS)-induced colonic inflammation and self-report the treatment efficiency. The fluorophore HBTTPIP can effectively aggregate in GPs, restricting intramolecular rotation and activating the fluorescence of HBTTPIP. After being orally administrated, HBTTPIP/GPs are phagocytosed by intestinal macrophages, which then migrate to colonic lesions, enabling non-invasive monitoring of the severity of IBD via in vivo fluorescence imaging. Notably, oral HBTTPIP/GPs ameliorate DSS-induced IBD by inhibiting the expressions of pro-inflammatory factors and improving colonic mucosal barrier function. Furthermore, these HBTTPIP/GPs realize self-feedback of the therapeutic effects of GPs on DSS-induced colitis. The oral biomimetic probe HBTTPIP/GPs reported herein provide a novel theranostic platform for IBD, integrating non-invasive diagnosis of IBD in situ and the corresponding treatment.
Collapse
Affiliation(s)
- Wenyuan Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Ya Wu
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Vascular Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yihui Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Rui Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Cheng Deng
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Luyang Yi
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Lufang Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Mengrong He
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Wuqi Zhou
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yuji Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Tang Gao
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| |
Collapse
|
15
|
Colon-specific delivery of methotrexate using hyaluronic acid modified pH-responsive nanocarrier for the therapy of colitis in mice. Int J Pharm 2023; 635:122741. [PMID: 36804523 DOI: 10.1016/j.ijpharm.2023.122741] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Oral immunosuppressant methotrexate (MTX) is an effective method for the treatment of inflammatory bowel disease (IBD). To overcome the defects of clinical application of MTX, poly (lactic-co-glycolic acid) (PLGA), Eudragits® S100 (ES100), chitosan (CS) and hyaluronic acid (HA) were used to structure the MTX-loaded HA-CS/ES100/PLGA nanoparticles (MTX@hCEP). MTX@hCEP had a hydrodynamic particle size of approximately 202.5 nm, narrow size distribution, negative zeta potential (-18.7 mV), and smooth surface morphology. In vitro drug release experiments under simulated gastrointestinal conditions indicated that MTX@hCEP exhibited colonic pH-sensitive drug release properties. The cellular uptake capacity of hCEP nanoparticles was significantly enhanced in RAW 264.7 macrophages. Moreover, we further found that the MTX@hCEP also inhibited the proliferation and the secretion of pro-inflammatory cytokines in the LPS-stimulated macrophages. In vivo imaging results not only demonstrated that the accumulated in the colon of colitis mice, but also indicated the extended retention time of MTX in the colon. Additionally, MTX@hCEP alleviated inflammatory symptoms via decreasing the activities of myeloperoxidase and pro-inflammatory factors, promoting mucosal repair in vivo. Collectively, these results clearly demonstrated that MTX@hCEP with properties of colon-specific and macrophages targeting can be exploited as an efficient nanotherapeutic for IBD therapy.
Collapse
|
16
|
The Effect of Curcumin-Loaded Glucan Nanoparticles on Immune Cells: Size as a Critical Quality Attribute. Pharmaceutics 2023; 15:pharmaceutics15020623. [PMID: 36839945 PMCID: PMC9959491 DOI: 10.3390/pharmaceutics15020623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/08/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Curcumin is known for its multiple health benefits, largely due to its antioxidant and anti-inflammatory properties. It has been extensively studied as a therapeutic agent, however, it does not have good clinical efficacy due to its poor water solubility and bioavailability. Despite accepting the encapsulation of this compound in polymeric particles as one of the most promising strategies to increase its therapeutic value, these nanoparticles have fallen short of expectations due to a lack of assessment of their possible adverse effects on the immune system. Therefore, in this work, we report on a new method to encapsulate curcumin into glucan nanoparticles and their effects on cells of the immune system were evaluated. Two different-sized curcumin-loaded glucan NPs (GluCur 100 and GluCur 380) were produced, each with an encapsulation efficiency close to 100%, and were characterized regarding their size distribution, surface properties, and morphology. The results revealed the greatest hemolytic effect and cytotoxicity for the smallest particles (100 nm) tested in human PBMCs and RAW 264.7 cells. Although GluCur 380 NPs showed a weaker ROS production, they were able to inhibit the production of NO by macrophages. Furthermore, we found that the coagulation time was not affected by both sized-particles as well as platelet function. Additionally, both nanoparticles induced lymphocyte proliferation and TNF-α secretion by Mo-DCs. In conclusion, this report emphasizes the importance of the immunotoxicity assessment and how this is dependent on the intrinsic properties of nanomaterials, hopefully contributing to increasing the safety of nanomedicines.
Collapse
|
17
|
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
|
18
|
Reis SF, Martins VJ, Bastos R, Lima T, Correia VG, Pinheiro BA, Silva LM, Palma AS, Ferreira P, Vilanova M, Coimbra MA, Coelho E. Feasibility of Brewer's Spent Yeast Microcapsules as Targeted Oral Carriers. Foods 2023; 12:246. [PMID: 36673340 PMCID: PMC9857821 DOI: 10.3390/foods12020246] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/15/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Brewer's spent yeast (BSY) microcapsules have a complex network of cell-wall polysaccharides that are induced by brewing when compared to the baker's yeast (Saccharomyces cerevisiae) microcapsules. These are rich in (β1→3)-glucans and covalently linked to (α1→4)- and (β1→4)-glucans in addition to residual mannoproteins. S. cerevisiae is often used as a drug delivery system due to its immunostimulatory potential conferred by the presence of (β1→3)-glucans. Similarly, BSY microcapsules could also be used in the encapsulation of compounds or drug delivery systems with the advantage of resisting digestion conferred by (β1→4)-glucans and promoting a broader immunomodulatory response. This work aims to study the feasibility of BSY microcapsules that are the result of alkali and subcritical water extraction processes, as oral carriers for food and biomedical applications by (1) evaluating the resistance of BSY microcapsules to in vitro digestion (IVD), (2) their recognition by the human Dectin-1 immune receptor after IVD, and (3) the recognition of IVD-solubilized material by different mammalian immune receptors. IVD digested 44-63% of the material, depending on the extraction process. The non-digested material, despite some visible agglutination and deformation of the microcapsules, preserved their spherical shape and was enriched in (β1→3)-glucans. These microcapsules were all recognized by the human Dectin-1 immune receptor. The digested material was differentially recognized by a variety of lectins of the immune system related to (β1→3)-glucans, glycogen, and mannans. These results show the potential of BSY microcapsules to be used as oral carriers for food and biomedical applications.
Collapse
Affiliation(s)
- Sofia F. Reis
- REQUIMTE-LAQV, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Vitor J. Martins
- REQUIMTE-LAQV, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Rita Bastos
- REQUIMTE-LAQV, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Tânia Lima
- I3S, Institute for Research in Innovation in Health, University of Porto, 4200-135 Porto, Portugal
| | - Viviana G. Correia
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Benedita A. Pinheiro
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Lisete M. Silva
- REQUIMTE-LAQV, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Angelina S. Palma
- UCIBIO—Applied Molecular Biosciences Unit, Department of Chemistry, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, School of Science and Technology, NOVA University Lisbon, 2819-516 Caparica, Portugal
| | - Paula Ferreira
- CICECO, Department of Materials and Ceramic Engineering, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Manuel Vilanova
- I3S, Institute for Research in Innovation in Health, University of Porto, 4200-135 Porto, Portugal
- IBMC, Institute for Molecular and Cell Biology, University of Porto, 4200-135 Porto, Portugal
- ICBAS, School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
| | - Manuel A. Coimbra
- REQUIMTE-LAQV, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Elisabete Coelho
- REQUIMTE-LAQV, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
19
|
Wu B. Acute moderate to severe ulcerative colitis treated by traditional Chinese medicine: A case report. World J Clin Cases 2022; 10:13356-13363. [PMID: 36683617 PMCID: PMC9851011 DOI: 10.12998/wjcc.v10.i36.13356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/15/2022] [Accepted: 12/08/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Ulcerative colitis (UC), also known as chronic nonspecific UC, is an inflammatory bowel disease characterized by diffuse colonic mucosal inflammation. The incidence and prevalence of UC have risen markedly, and the disease seriously affects the quality of life of patients and imposes a great burden on the world health care infrastructure and economy.
CASE SUMMARY Case I describes a 34-year-old female who came to see a doctor because of repeated abdominal pain, diarrhoea and purulent blood for 2 mo. This patient had UC with an initial onset, an active stage and a wide range of lesions. After the poor effect of sulfasalazine and mesalazine, the patient's condition gradually deteriorated, her abdominal pain and bloody stools continued, and anemia occurred. She began treatment with the Chinese medicine Guizhi Dahuang decoction, which was taken orally twice a day, 200 mL each time. After 6 mo of treatment, abdominal pain, diarrhoea, bloody stool and other symptoms disappeared. No abnormality was found by repeat electronic enteroscopye, and the anemia was corrected. The patient's condition did not recur after nearly 4 years of follow-up.
CONCLUSION A series of symptoms in this UC patient significantly improved with the administration of traditional Chinese medicine.
Collapse
Affiliation(s)
- Bin Wu
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310005, Zhejiang Province, China
| |
Collapse
|
20
|
Mucoadhesive carriers for oral drug delivery. J Control Release 2022; 351:504-559. [PMID: 36116580 PMCID: PMC9960552 DOI: 10.1016/j.jconrel.2022.09.024] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/10/2022] [Accepted: 09/12/2022] [Indexed: 12/24/2022]
Abstract
Among the various dosage forms, oral medicine has extensive benefits including ease of administration and patients' compliance, over injectable, suppositories, ocular and nasal. Despite of extensive demand and emerging advantages, over 50% of therapeutic molecules are not available in oral form due to their physicochemical properties. More importantly, most of the biologics, proteins, peptide, and large molecular drugs are mostly available in injectable form. Conventional oral drug delivery system has limitation such as degradation and lack of stability within stomach due to presence of highly acidic gastric fluid, hinders their therapeutic efficacy and demand more frequent and higher dosing. Hence, formulation for controlled, sustained, and targeted drug delivery, need to be designed with feasibility to target the specific region of gastrointestinal (GI) tract such as stomach, small intestine, intestine lymphatic, and colon is challenging. Among various oral delivery approaches, mucoadhesive vehicles are promising and has potential for improving oral drug retention and controlled absorption to treat local diseases within the GI tract, as well systemic diseases. This review provides the overview about the challenges and opportunities to design mucoadhesive formulation for oral delivery of therapeutics in a way to target the specific region of the GI tract. Finally, we have concluded with future perspective and potential of mucoadhesive formulations for oral local and systemic delivery.
Collapse
|
21
|
Feng X, Xie Q, Xu H, Zhang T, Li X, Tian Y, Lan H, Kong L, Zhang Z. Yeast Microcapsule Mediated Natural Products Delivery for Treating Ulcerative Colitis through Anti-Inflammatory and Regulation of Macrophage Polarization. ACS APPLIED MATERIALS & INTERFACES 2022; 14:31085-31098. [PMID: 35770618 DOI: 10.1021/acsami.2c05642] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The common and frequent disease, ulcerative colitis (UC), causes serious physical and mental distress to patients. M2 macrophages have proven to play a role in anti-inflammation, which is a new potential target for UC therapy. In this study, we designed a safe and macrophages-targeting oral drug delivery system. Natural products, berberine (BBR), and Epigallocatechin Gallate (EGCG) with anti-inflammatory activity were assembled and encapsulated into yeast microcapsule (YM), generating therapeutic system BBR/MPN@YM. BBR and EGCG exhibited synergistic effects against UC through the effect of antioxidation. Through the interaction between β-1,3-d-glucan on the surface of YM and dectin-1 receptors on macrophages, BBR/MPN@YM could be effectively transported to inflammation parts and internalized into macrophages, avoiding gastric degradation. In the in vivo UC mouse model, BBR/MPN@YM could transform M1 macrophages into anti-inflammatory M2 macrophages, thus exerting specific anti-inflammatory effects. Therefore, this BBR/MPN@YM targeted oral drug delivery system provided a new macrophages-targeting strategy for the clinical treatment of UC.
Collapse
Affiliation(s)
- Xingxing Feng
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Xie
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongbo Xu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tiantian Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaonan Li
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yinmei Tian
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hongbing Lan
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Engineering Research Center for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
22
|
Tan Y, Chen L, Li K, Lou B, Liu Y, Liu Z. Yeast as carrier for drug delivery and vaccine construction. J Control Release 2022; 346:358-379. [PMID: 35483637 DOI: 10.1016/j.jconrel.2022.04.032] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/19/2022] [Accepted: 04/19/2022] [Indexed: 12/16/2022]
Abstract
Yeast has been employed as an effective derived drug carrier as a unicellular microorganism. Many research works have been devoted to the encapsulation of nucleic acid compounds, insoluble small molecule drugs, small molecules, liposomes, polymers, and various nanoparticles in yeast for the treatment of disease. Recombinant yeast-based vaccine carriers (WYV) have played a major role in the development of vaccines. Herein, the latest reports on the application of yeast carriers and the development of related research are summarized, a conceptual description of gastrointestinal absorption of yeast carriers, as well as the various package forms of different drug molecules and nanoparticles in yeast carriers are introduced. In addition, the advantages and development of recombinant yeast vaccine carriers for the disease, veterinary and aquaculture applications are discussed. Moreover, the current challenges and future directions of yeast carriers are proposed.
Collapse
Affiliation(s)
- Yifu Tan
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, PR China
| | - Liwei Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, PR China
| | - Ke Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Beibei Lou
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha 410083, Hunan Province, PR China.
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan Province, PR China; Molecular Imaging Research Center of Central South University, Changsha 410008, Hunan, PR China.
| |
Collapse
|
23
|
Cassinotti A, Batticciotto A, Parravicini M, Lombardo M, Radice P, Cortelezzi CC, Segato S, Zanzi F, Cappelli A, Segato S. Evidence-based efficacy of methotrexate in adult Crohn's disease in different intestinal and extraintestinal indications. Therap Adv Gastroenterol 2022; 15:17562848221085889. [PMID: 35340755 PMCID: PMC8949794 DOI: 10.1177/17562848221085889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 02/18/2022] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Methotrexate (MTX) is included in the therapeutic armamentarium of Crohn's disease (CD), although its positioning is currently uncertain in an era in which many effective biological drugs are available. No systematic reviews or meta-analysis have stratified the clinical outcomes of MTX according to the specific clinical scenarios of its use. METHODS Medline, PubMed and Scopus were used to extract eligible studies, from database inception to May 2021. A total of 163 studies were included. A systematic review was performed by stratifying the outcomes of MTX according to formulation, clinical indication and criteria of efficacy. RESULTS The use of MTX is supported by randomized clinical trials only in steroid-dependent CD, with similar outcomes to thiopurines. The use of MTX in patients with steroid-refractoriness, failure of thiopurines or in combination with biologics is not supported by high levels of evidence. Combination therapy with biologics can optimize the immunogenic profile of the biological drug, but the impact on long-term clinical outcomes is described only in small series with anti-TNFα. Other off-label uses, such as fistulizing disease, mucosal healing, postoperative prevention and extraintestinal manifestations, are described in small uncontrolled series. The best performance in most indications was shown by parenteral MTX, favouring higher doses (25 mg/week) in the induction phase. DISCUSSION Evidence from high-quality studies in favour of MTX is scarce and limited to the steroid-dependent disease, in which other drugs are the leading players today. Many limitations on study design have been found, such as the prevalence of retrospective underpowered studies and the lack of stratification of outcomes according to specific types of patients and formulations of MTX. CONCLUSION MTX is a valid option as steroid-sparing agent in steroid-dependent CD. Numerous other clinical scenarios require well-designed clinical studies in terms of patient profile, drug formulation and dosage, and criteria of efficacy.
Collapse
Affiliation(s)
| | | | | | | | - Paolo Radice
- Ophtalmology Unit, ASST Sette Laghi, Varese, Italy
| | | | - Simone Segato
- Gastroenterology Unit, ASST Sette Laghi, Varese, Italy
| | | | | | - Sergio Segato
- Gastroenterology Unit, ASST Sette Laghi, Varese, Italy
| |
Collapse
|
24
|
Wei W, Zhang Y, Li R, Cao Y, Yan X, Ma Y, Zhang Y, Yang M, Zhang M. Oral Delivery of Pterostilbene by L-Arginine-Mediated "Nano-Bomb" Carrier for the Treatment of Ulcerative Colitis. Int J Nanomedicine 2022; 17:603-616. [PMID: 35177902 PMCID: PMC8843770 DOI: 10.2147/ijn.s347506] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/25/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic inflammatory bowel disease (IBD) of unknown aetiology affecting the colon and rectum. Pterostilbene (PS) has been reported as an effective antioxidant and anti-inflammatory agent in preclinical IBD models. However, the therapeutic outcomes of PS are limited by potential side effects associated with the systemic exposure and the modest bioavailability afforded by its oral administration. These issues can be improved with the use of intelligent responsive nanoparticles with the ability of lysosome escape, given their high drug delivery capacity and reducing the side effects. MATERIALS AND METHODS Herein, the hyaluronic acid (HA)-modified L-arginine CO2 nanoparticles (HA-L-Arg-CO2@NPs) loaded with PS (HA-PS@NPs) are constructed. Under lysosomal pH conditions, HA-PS@NPs liberate CO2 and generate a pH-activated nano-bomb effect to augment the cytosolic delivery of PS. RESULTS HA-L-Arg-CO2@NPs show great biocompatibility and the excellent ability to target the colon. Using lipopolysaccharide-induced inflammation in vitro, the prominent anti-inflammatory effect of HA-L-Arg-CO2@NPs and HA-PS@NPs is observed. Further, orally administered HA-L-Arg-CO2@NPs and HA-PS@NPs via the colon-targeted chitosan/alginate (CA) hydrogel downregulate pro-inflammatory cytokines and reduce intestinal permeability, yielding significant outcomes in alleviating the symptoms of UC. CONCLUSION This pH-activated "nano-bomb" carrier with therapeutic effect can be exploited as efficient oral drug carriers for UC treatment.
Collapse
Affiliation(s)
- Wei Wei
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Xi’an No.1 Hospital, Shaanxi Institute of Ophthalmology, Shaanxi Key Laboratory of Ophthalmology, Clinical Research Center for Ophthalmology Diseases of Shaanxi Province, First Affiliated Hospital of Northwestern University, Xi’an, Shaanxi, People’s Republic of China
| | - Yujie Zhang
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Runqing Li
- Department of Radiology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Yameng Cao
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Xiangji Yan
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Yana Ma
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Yuanyuan Zhang
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Mei Yang
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
- Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi’an Jiaotong University, Xi’an, Shaanxi, People’s Republic of China
| |
Collapse
|
25
|
Bastos R, Oliveira PG, Gaspar VM, Mano JF, Coimbra MA, Coelho E. Brewer's yeast polysaccharides - A review of their exquisite structural features and biomedical applications. Carbohydr Polym 2022; 277:118826. [PMID: 34893243 DOI: 10.1016/j.carbpol.2021.118826] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/27/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022]
Abstract
Recent advances on brewer's yeast cell wall polysaccharides have unraveled exquisite structural features and diverse composition with (β1→3), (β1→6), (α1→4), (β1→4)-mix-linked glucans that are recognized to interact with different cell receptors and trigger specific biological responses. Herein, a comprehensive showcase of structure-biofunctional relationships between yeast polysaccharides and their biological targets is highlighted, with a focus on polysaccharide features that govern the biomedical activity. The insolubility of β-glucans is a crucial factor for binding and activation of Dectin-1 receptor, operating as adjuvants of immune responses. Contrarily, soluble low molecular weight β-glucans have a strong inhibition of reactive oxygen species production, acting as antagonists of Dectin-1 mediated signaling. Soluble glucan-protein moieties can also act as antitumoral agents. The balance between mannoproteins-TLR2 and β-glucans-Dectin-1 receptors-activation is crucial for osteogenesis. Biomedical applications value can also be obtained from yeast microcapsules as oral delivery systems, where highly branched (β1→6)-glucans lead to higher receptor affinity.
Collapse
Affiliation(s)
- Rita Bastos
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Patrícia G Oliveira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal; CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Vítor M Gaspar
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - João F Mano
- CICECO-Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Manuel A Coimbra
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Elisabete Coelho
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| |
Collapse
|
26
|
|
27
|
Ivanova E. Yeasts in nanotechnology-enabled oral vaccine and gene delivery. Bioengineered 2021; 12:8325-8335. [PMID: 34592900 PMCID: PMC8806958 DOI: 10.1080/21655979.2021.1985816] [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: 07/12/2021] [Accepted: 09/22/2021] [Indexed: 11/30/2022] Open
Abstract
Oral vaccine and gene delivery systems must be engineered to withstand several different physiological environments, such as those present in the oral cavity, stomach, and jejunum, each of which exhibits varying pH levels and enzyme distributions. Additionally, these systems must be designed to ensure appropriate gastrointestinal absorption and tissue/cellular targeting properties. Yeasts-based delivery vehicles are excellent candidates for oral vaccine and oral gene therapies as many species possess cellular characteristics resulting in enhanced resistance to the harsh gastrointestinal (GI) environment and facilitated passage across the mucosal barrier. Yeast capsules can stimulate and modulate host immune responses, which is beneficial for vaccine efficacy. In addition, recombinant modification of yeasts to express cell penetrating proteins and injection mechanisms along with efficient cell adhering capabilities can potentially improve transfection rates of genetic material. In this literature review, we present evidence supporting the beneficial role yeast-based delivery systems can play in increasing the efficacy of oral administration of vaccines and gene therapies.
Collapse
Affiliation(s)
- Elena Ivanova
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| |
Collapse
|
28
|
Wang L, Liao Y, Yang R, Zhu Z, Zhang L, Wu Z, Sun X. An engineered probiotic secreting Sj16 ameliorates colitis via Ruminococcaceae/butyrate/retinoic acid axis. Bioeng Transl Med 2021; 6:e10219. [PMID: 34589596 PMCID: PMC8459592 DOI: 10.1002/btm2.10219] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/14/2021] [Accepted: 03/18/2021] [Indexed: 12/29/2022] Open
Abstract
Most inflammatory bowel disease (IBD) patients are unable to maintain a lifelong remission. Developing a novel therapeutic strategy is urgently needed. In this study, we adopt a new strategy to attenuate colitis using the Escherichia coli Nissle 1917 probiotic strain to express a schistosome immunoregulatory protein (Sj16) in the gastrointestinal tract. The genetically engineered Nissle 1917 (EcN-Sj16) highly expressed Sj16 in the gastrointestinal tracts of dextran sulfate sodium-induced colitis mice and significantly attenuated the clinical activity of colitis mice. Mechanistically, EcN-Sj16 increased the intestinal microbiota diversity and selectively promoted the growth of Ruminococcaceae and therefore enhanced the butyrate production. Butyrate induced the expression of retinoic acid, which further attenuated the clinical activity of colitis mice by increasing Treg cells and decreasing Th17. Strikingly, retinoic acid inhibitor inhibited the therapeutic effects of EcN-Sj16 in colitis mice. These findings suggest that EcN-Sj16 represents a novel engineered probiotic that may be used to treat IBD.
Collapse
Affiliation(s)
- Lifu Wang
- Department of Parasitology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Tropical Disease ControlMinistry of Education, Sun Yat‐sen UniversityGuangzhouChina
- Provincial Engineering Technology Research Center for Biological Vector ControlGuangzhouChina
| | - Yao Liao
- Department of Parasitology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Tropical Disease ControlMinistry of Education, Sun Yat‐sen UniversityGuangzhouChina
- Provincial Engineering Technology Research Center for Biological Vector ControlGuangzhouChina
| | - Ruibing Yang
- Department of Parasitology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Tropical Disease ControlMinistry of Education, Sun Yat‐sen UniversityGuangzhouChina
- Provincial Engineering Technology Research Center for Biological Vector ControlGuangzhouChina
| | - Zifeng Zhu
- Department of Parasitology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Tropical Disease ControlMinistry of Education, Sun Yat‐sen UniversityGuangzhouChina
- Provincial Engineering Technology Research Center for Biological Vector ControlGuangzhouChina
| | - Lichao Zhang
- Department of Parasitology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Tropical Disease ControlMinistry of Education, Sun Yat‐sen UniversityGuangzhouChina
- Provincial Engineering Technology Research Center for Biological Vector ControlGuangzhouChina
| | - Zhongdao Wu
- Department of Parasitology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Tropical Disease ControlMinistry of Education, Sun Yat‐sen UniversityGuangzhouChina
- Provincial Engineering Technology Research Center for Biological Vector ControlGuangzhouChina
| | - Xi Sun
- Department of Parasitology, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
- Key Laboratory of Tropical Disease ControlMinistry of Education, Sun Yat‐sen UniversityGuangzhouChina
- Provincial Engineering Technology Research Center for Biological Vector ControlGuangzhouChina
| |
Collapse
|
29
|
Gao T, Wu Y, Wang W, Deng C, Chen Y, Yi L, Song Y, Li W, Xu L, Xie Y, Fang L, Jin Q, Zhang L, Tang BZ, Xie M. Biomimetic Glucan Particles with Aggregation-Induced Emission Characteristics for Noninvasive Monitoring of Transplant Immune Response. ACS NANO 2021; 15:11908-11928. [PMID: 34264052 DOI: 10.1021/acsnano.1c03029] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Real-time monitoring of post-transplant immune response is critical to prolong the survival of grafts. The current gold standard for assessing the immune response to graft is biopsy. However, such a method is invasive and prone to false negative results due to limited tissue size available and the heterogeneity of the rejection site. Herein, we report biomimetic glucan particles with aggregation-induced emission (AIE) characteristics (HBTTPEP/GPs) for real-time noninvasive monitoring of post-transplant immune response. We have found that the positively charged near-infrared AIEgens can effectively aggregate in the confined space of glucan particles (GPs), thereby turning on the fluorescence emission. HBTTPEP/GPs can track macrophages for 7 days without hampering the bioactivity. Oral administration of HBTTPEP/GPs can specially target macrophages by mimicking yeast, which then migrate to the transplant rejection site. The fluorescence emitted from HBTTPEP/GPs correlated well with the infiltration of macrophages and the degree of allograft rejection. Furthermore, a single oral HBTTPEP/GPs dose can dynamically evaluate the therapeutic response to immunosuppressive therapy. Consequently, the biomimetic AIE-active glucan particles can be developed as a promising probe for immune-monitoring in solid organ transplantation.
Collapse
Affiliation(s)
- Tang Gao
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Ya Wu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Wenyuan Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Cheng Deng
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Luyang Yi
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yishu Song
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Wenqu Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Lingling Xu
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yuji Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Lingyun Fang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Ben Zhong Tang
- Department of Chemistry, Hong Kong Branch of Chinese National Engineering Research Center for Tissue Restoration and Reconstruction and Institute for Advanced Study, The Hong Kong University of Science and Technology (HKUST), Clear Water Bay, Kowloon, Hong Kong, China
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Clinical Research Center for Medical Imaging, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| |
Collapse
|
30
|
Chen Q, Luo R, Han X, Zhang J, He Y, Qi S, Pu X, Nie W, Dong L, Xu H, Liu F, Lin M, Zhong H, Fu C, Gao F. Entrapment of Macrophage-Target Nanoparticles by Yeast Microparticles for Rhein Delivery in Ulcerative Colitis Treatment. Biomacromolecules 2021; 22:2754-2767. [PMID: 34019390 DOI: 10.1021/acs.biomac.1c00425] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In this study, we developed an advanced colitis-targeted nanoparticles (NPs)-into-yeast cell wall microparticles (YPs) drug delivery system for ulcerative colitis (UC) therapy. In brief, YPs entrap hyaluronic acid (HA), and polyethylenimine (PEI) modified rhein (RH)-loaded ovalbumin NPs (HA/PEI-RH NPs) to form HA/PEI-RH NYPs. YPs can make HA/PEI-RH NPs pass through gastric environment stably and be degraded by β-glucanase to promote drug release from HA/PEI-RH NYPs in the colon. Cellular uptake evaluation confirmed that HA/PEI-RH NPs could specifically target and enhance the uptake rate via HA ligands. In biodistribution studies, HA/PEI-RH NYPs were able to efficiently accumulate in the inflammed colon in mice. In vivo experiments revealed that the HA/PEI-RH NYPs could significantly alleviate inflammation by inhibiting the TLR4/MyD88/NF-κB signaling pathway. Therefore, HA/PEI-RH NYPs have advantages of good gastric stability, β-glucanase-sensitive release ability, macrophage-targeted ability, and anti-UC effects. These advantages indicate YPs-entrapped multifunctional NPs are a promising oral drug delivery system for UC therapy.
Collapse
Affiliation(s)
- Qiyan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Ruifeng Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Xiaoqin Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Yao He
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Shanshan Qi
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Xiulan Pu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Wenbiao Nie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Lingling Dong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Haiting Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Fang Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Meisi Lin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China.,Sichuan Provincial Acupuncture School, Chengdu 611731, China
| | - Huiyun Zhong
- Sichuan Vocational College of Health and Rehabilitation, Zigong 643000, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Fei Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| |
Collapse
|
31
|
Wang L, Yu M, Yang H. Recent Progress in the Diagnosis and Precise Nanocarrier-Mediated Therapy of Inflammatory Bowel Disease. J Inflamm Res 2021; 14:1701-1716. [PMID: 33953597 PMCID: PMC8092629 DOI: 10.2147/jir.s304101] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/30/2021] [Indexed: 12/23/2022] Open
Abstract
The effective colon drug delivery remains to be an international frontier research in inflammatory bowel disease (IBD) therapy. The exploration and research of nanocarrier-based nanomedicine with great potential brings new opportunities for IBD therapy and diagnoses. Functional nanocarriers with varying morphology and characteristics can not only effectively avoid the destruction of the complex gastrointestinal (GI) tract microenvironment but also endow drugs with target therapy and improved bioavailability, thus elevating therapeutic efficacy. In this review, we illustrated several challenges in IBD therapy, then emphasis on some latest research progress of nanoparticles based therapy of oral administration, rectal administration and parenteral administration, as well as IBD diagnoses. Finally, we described the future perspective of nanocarriers in the treatment and diagnoses of IBD.
Collapse
Affiliation(s)
- Liucan Wang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Min Yu
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, People’s Republic of China
| | - Hua Yang
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
32
|
Feng GN, Huang XT, Jiang XL, Deng TW, Li QX, Li JX, Wu QN, Li SP, Sun XQ, Huang YG, Qin AP, Liang L, Fu JJ. The Antibacterial Effects of Supermolecular Nano-Carriers by Combination of Silver and Photodynamic Therapy. Front Chem 2021; 9:666408. [PMID: 33937203 PMCID: PMC8082423 DOI: 10.3389/fchem.2021.666408] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/19/2021] [Indexed: 11/16/2022] Open
Abstract
The over-use of antibiotics has promoted multidrug resistance and decreased the efficacy of antibiotic therapy. Thus, it is still in great need to develop efficient treatment strategies to combat the bacteria infection. The antimicrobial photodynamic therapy (aPDT) and silver nanoparticles have been emerged as effective antibacterial methods. However, the silver therapy may induce serious damages to human cells at high concentrations and, the bare silver nanoparticles may rapidly aggregate, which would reduce the antibacterial efficacy. The encapsulation of sliver by nano-carrier is a promising way to avoid its aggregation and facilitates the co-delivery of drugs for combination therapy, which does not require high concentration of sliver to exert antibacterial efficacy. This work constructed a self-assembled supermolecular nano-carrier consisting of the photosensitizers (PSs), the anti-inflammatory agent and silver. The synthesized supermolecular nano-carrier produced reactive oxygen species (ROS) under the exposure of 620-nm laser. It exhibited satisfying biocompatibility in L02 cells. And, this nano-carrier showed excellent antibacterial efficacy in Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus) as indicated by bacterial growth and colony formation. Its antibacterial performance is further validated by the bacteria morphology through the scanning electron microscope (SEM), showing severely damaged structures of bacteria. To summary, the supermolecular nano-carrier TCPP-MTX-Ag-NP combining the therapeutic effects of ROS and silver may serve as a novel strategy of treatment for bacterial infection.
Collapse
Affiliation(s)
- Gui-ning Feng
- The Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The First Affiliated Hospital of Guangzhou Medical University and the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Xiao-tao Huang
- The Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The First Affiliated Hospital of Guangzhou Medical University and the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Xin-lin Jiang
- The Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The First Affiliated Hospital of Guangzhou Medical University and the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Ting-wei Deng
- School of Pharmaceutical Sciences, Shenzhen University, Shenzhen, China
| | - Qiu-xia Li
- The Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The First Affiliated Hospital of Guangzhou Medical University and the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Jie-xia Li
- The Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The First Affiliated Hospital of Guangzhou Medical University and the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Qian-ni Wu
- The Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The First Affiliated Hospital of Guangzhou Medical University and the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Song-pei Li
- The Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The First Affiliated Hospital of Guangzhou Medical University and the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Xian-qiang Sun
- The Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The First Affiliated Hospital of Guangzhou Medical University and the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Yu-gang Huang
- The Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The First Affiliated Hospital of Guangzhou Medical University and the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Ai-ping Qin
- The Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The First Affiliated Hospital of Guangzhou Medical University and the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Lu Liang
- The Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The First Affiliated Hospital of Guangzhou Medical University and the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Ji-jun Fu
- The Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, The First Affiliated Hospital of Guangzhou Medical University and the Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
33
|
Xiao Q, Li X, Li Y, Wu Z, Xu C, Chen Z, He W. Biological drug and drug delivery-mediated immunotherapy. Acta Pharm Sin B 2021; 11:941-960. [PMID: 33996408 PMCID: PMC8105778 DOI: 10.1016/j.apsb.2020.12.018] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/03/2020] [Accepted: 11/15/2020] [Indexed: 12/11/2022] Open
Abstract
The initiation and development of major inflammatory diseases, i.e., cancer, vascular inflammation, and some autoimmune diseases are closely linked to the immune system. Biologics-based immunotherapy is exerting a critical role against these diseases, whereas the usage of the immunomodulators is always limited by various factors such as susceptibility to digestion by enzymes in vivo, poor penetration across biological barriers, and rapid clearance by the reticuloendothelial system. Drug delivery strategies are potent to promote their delivery. Herein, we reviewed the potential targets for immunotherapy against the major inflammatory diseases, discussed the biologics and drug delivery systems involved in the immunotherapy, particularly highlighted the approved therapy tactics, and finally offer perspectives in this field.
Collapse
Key Words
- AAs, amino acids
- ACT, adoptive T cell therapy
- AHC, Chlamydia pneumonia
- ALL, acute lymphoblastic leukemia
- AP, ascorbyl palmitate
- APCs, antigen-presenting cells
- AS, atherosclerosis
- ASIT, antigen-specific immunotherapy
- Adoptive cell transfer
- ApoA–I, apolipoprotein A–I
- ApoB LPs, apolipoprotein-B-containing lipoproteins
- Atherosclerosis
- BMPR-II, bone morphogenetic protein type II receptor
- Biologics
- Bregs, regulatory B lymphocytes
- CAR, chimeric antigen receptor
- CCR9–CCL25, CC receptor 9–CC chemokine ligand 25
- CD, Crohn's disease
- CETP, cholesterol ester transfer protein
- CTLA-4, cytotoxic T-lymphocyte-associated protein-4
- CX3CL1, CXXXC-chemokine ligand 1
- CXCL 16, CXC-chemokine ligand 16
- CXCR 2, CXC-chemokine receptor 2
- Cancer immunotherapy
- CpG ODNs, CpG oligodeoxynucleotides
- DAMPs, danger-associated molecular patterns
- DCs, dendritic cells
- DDS, drug delivery system
- DMARDs, disease-modifying antirheumatic drugs
- DMPC, 1,2-dimyristoyl-sn-glycero-3-phosphatidylcholine
- DSS, dextran sulfate sodium
- Dex, dexamethasone
- Drug delivery
- ECM, extracellular matrix
- ECs, endothelial cells
- EGFR, epidermal growth factor receptor
- EPR, enhanced permeability and retention effect
- ET-1, endothelin-1
- ETAR, endothelin-1 receptor type A
- FAO, fatty acid oxidation
- GM-CSF, granulocyte–macrophage colony-stimulating factor
- HA, hyaluronic acid
- HDL, high density lipoprotein
- HER2, human epidermal growth factor-2
- IBD, inflammatory bowel diseases
- ICOS, inducible co-stimulator
- ICP, immune checkpoint
- IFN, interferon
- IL, interleukin
- IT-hydrogel, inflammation-targeting hydrogel
- Immune targets
- Inflammatory diseases
- JAK, Janus kinase
- LAG-3, lymphocyte-activation gene 3
- LDL, low density lipoprotein
- LPS, lipopolysaccharide
- LTB4, leukotriene B4
- MCP-1, monocyte chemotactic protein-1
- MCT, monocrotaline
- MDSC, myeloid-derived suppressor cell
- MHCs, major histocompatibility complexes
- MHPC, 1-myristoyl-2-hydroxy-sn-glycero-phosphocholine
- MIF, migration inhibitory factor
- MM, multiple myeloma
- MMP, matrix metalloproteinase
- MOF, metal–organic framework
- MPO, myeloperoxidase
- MSCs, mesenchymal stem cells
- NF-κB, nuclear factor κ-B
- NK, natural killer
- NPs, nanoparticles
- NSAIDs, nonsteroidal anti-inflammatory drugs
- PAECs, pulmonary artery endothelial cells
- PAH, pulmonary arterial hypertension
- PASMCs, pulmonary arterial smooth muscle cells
- PBMCs, peripheral blood mononuclear cells
- PCSK9, proprotein convertase subtilisin kexin type 9
- PD-1, programmed death protein-1
- PD-L1, programmed cell death-ligand 1
- PLGA, poly lactic-co-glycolic acid
- Pulmonary artery hypertension
- RA, rheumatoid arthritis
- ROS, reactive oxygen species
- SHP-2, Src homology 2 domain–containing tyrosine phosphatase 2
- SLE, systemic lupus erythematosus
- SMCs, smooth muscle cells
- Src, sarcoma gene
- TCR, T cell receptor
- TGF-β, transforming growth factor β
- TILs, tumor-infiltrating lymphocytes
- TIM-3, T-cell immunoglobulin mucin 3
- TLR, Toll-like receptor
- TNF, tumor necrosis factor
- TRAF6, tumor necrosis factor receptor-associated factor 6
- Teff, effector T cell
- Th17, T helper 17
- Tph, T peripheral helper
- Tregs, regulatory T cells
- UC, ulcerative colitis
- VEC, vascular endothelial cadherin
- VEGF, vascular endothelial growth factor
- VISTA, V-domain immunoglobulin-containing suppressor of T-cell activation
- YCs, yeast-derived microcapsules
- bDMARDs, biological DMARDs
- hsCRP, high-sensitivity C-reactive protein
- mAbs, monoclonal antibodies
- mPAP, mean pulmonary artery pressure
- nCmP, nanocomposite microparticle
- rHDL, recombinant HDL
- rhTNFRFc, recombinant human TNF-α receptor II-IgG Fc fusion protein
- scFv, single-chain variable fragment
- α1D-AR, α1D-adrenergic receptor
Collapse
Affiliation(s)
- Qingqing Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yi Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenfeng Wu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong 999077, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
34
|
Wu Y, Jin Q, Chen Y, Li H, Deng C, Sun Z, Li Y, Wang B, Li H, Wu C, Zhang L, Xie M. Bioinspired β-glucan microcapsules deliver FK506 to lymph nodes for treatment of cardiac allograft acute rejection. Biomater Sci 2020; 8:5282-5292. [PMID: 32749395 DOI: 10.1039/d0bm01028f] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lymph node (LN)-targeted delivery exhibits enormous potential to improve the treatment efficacy of immunosuppressants for transplantation. However, current strategies are still limited by the inefficiency of delivery by passive targeting, the high cost of antibody-mediated active targeting, as well as poor patient compliance by parenteral delivery. Herein, bioinspired β-glucan microcapsules (GM) was used to load and transfer low dose FK506 into LNs via oral administration, which may relieve cardiac allograft acute rejection with low nephrotoxicity. The LN distribution study showed that both DiR and FK506 were delivered into the LNs effectively via GM-mediated transport after 24 h and were present in the LNs for at least 48 h. The FK506-loaded GM (GM-FK506) significantly prolonged allograft survival compared with the PBS group (mean survival time, 17.8 ± 1.9 versus 7.3 ± 1.0 days; P < 0.01), and marked decreased the acute rejection grade. Furthermore, T cell infiltration, and secretion of IL-2 and IFN-γ were dramatically reduced in the GM-FK506 group. As expected, no nephrotoxicity was observed after five consecutive administrations of GM-FK506. Our results demonstrate that GM-FK506 is a promising strategy for the treatment of cardiac allograft acute rejection, indicating that GM mediated LNs targeting may provide a potential opportunity for managing immune-related diseases.
Collapse
Affiliation(s)
- Ya Wu
- Department of Ultrasound, Union Hospital, Tongji Medical College of Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|