1
|
Genkel V, Zaripova Y, Kuznetsova A, Sluchanko A, Minasova A, Zotova M, Saenko A, Savochkina A, Dolgushina A. Neutrophils at the Crossroads of Inflammatory Bowel Disease and Atherosclerosis: A State-of-the-Art Review. Cells 2025; 14:738. [PMID: 40422241 DOI: 10.3390/cells14100738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/12/2025] [Accepted: 05/16/2025] [Indexed: 05/28/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a growing global problem, particularly in regions with low sociodemographic indices and growing populations. IBD incidence is increasing among children and adolescents, leading to a growing economic burden. The prevalence of atherosclerotic cardiovascular diseases among patients with IBD is also higher than in the general population. While mortality rates have decreased, cardiovascular disease (CVD) remains a significant contributor to mortality and disability in IBD patients. According to the current understanding, neutrophils play an important role in both the atherogenesis and pathogenesis of IBD. This review addresses the state of the art of neutrophil involvement in the development of atherosclerosis and IBD. In the present review, we summarize the currently available evidence regarding neutrophils as a possible key driver of extraintestinal manifestations of IBD and cardiovascular complications. We provide a discussion on the potential role of neutrophil-derived markers in the development of new approaches for the precise diagnosis of atherosclerosis in patients with IBD, as well as new therapeutic targets.
Collapse
Affiliation(s)
- Vadim Genkel
- Department of Internal Medicine, South-Ural State Medical University, Chelyabinsk 454141, Russia
| | - Yana Zaripova
- Department of Internal Medicine, South-Ural State Medical University, Chelyabinsk 454141, Russia
| | - Alla Kuznetsova
- Department of Hospital Therapy, South-Ural State Medical University, Chelyabinsk 454141, Russia
| | - Alena Sluchanko
- Department of Hospital Therapy, South-Ural State Medical University, Chelyabinsk 454141, Russia
| | - Anna Minasova
- Research Institution of Immunology, South-Ural State Medical University, Chelyabinsk 454141, Russia
| | - Maria Zotova
- Research Institution of Immunology, South-Ural State Medical University, Chelyabinsk 454141, Russia
| | - Anna Saenko
- Department of Hospital Therapy, South-Ural State Medical University, Chelyabinsk 454141, Russia
| | - Albina Savochkina
- Research Institution of Immunology, South-Ural State Medical University, Chelyabinsk 454141, Russia
| | - Anastasiya Dolgushina
- Department of Hospital Therapy, South-Ural State Medical University, Chelyabinsk 454141, Russia
| |
Collapse
|
2
|
Aalhate M, Mahajan S, Dhuri A, Singh PK. Biohybrid nano-platforms manifesting effective cancer therapy: Fabrication, characterization, challenges and clinical perspective. Adv Colloid Interface Sci 2025; 335:103331. [PMID: 39522420 DOI: 10.1016/j.cis.2024.103331] [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: 03/13/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Nanotechnology-based delivery systems have brought a paradigm shift in the management of cancer. However, the main obstacles to nanocarrier-based delivery are their limited circulation duration, excessive immune clearance, inefficiency in interacting effectively in a biological context and overcoming biological barriers. This demands effective engineering of nanocarriers to achieve maximum efficacy. Nanocarriers can be maneuvered with biological components to acquire biological identity for further regulating their biodistribution and cell-to-cell cross-talk. Thus, the integration of synthetic and biological components to deliver therapeutic cargo is called a biohybrid delivery system. These delivery systems possess the advantage of synthetic nanocarriers, such as high drug loading, engineerable surface, reproducibility, adequate communication and immune evasion ability of biological constituents. The biohybrid delivery vectors offer an excellent opportunity to harness the synergistic properties of the best entities of the two worlds for improved therapeutic outputs. The major spotlights of this review are different biological components, synthetic counterparts of biohybrid nanocarriers, recent advances in hybridization techniques, and the design of biohybrid delivery systems for cancer therapy. Moreover, this review provides an overview of biohybrid systems with therapeutic and diagnostic applications. In a nutshell, this article summarizes the advantages and limitations of various biohybrid nano-platforms, their clinical potential and future directions for successful translation in cancer management.
Collapse
Affiliation(s)
- Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Anish Dhuri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India.
| |
Collapse
|
3
|
Hao ZW, Zhang ZY, Wang ZP, Wang Y, Chen JY, Chen TH, Shi G, Li HK, Wang JW, Dong MC, Hong L, Li JF. Bioactive peptides and proteins for tissue repair: microenvironment modulation, rational delivery, and clinical potential. Mil Med Res 2024; 11:75. [PMID: 39639374 PMCID: PMC11619216 DOI: 10.1186/s40779-024-00576-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 10/25/2024] [Indexed: 12/07/2024] Open
Abstract
Bioactive peptides and proteins (BAPPs) are promising therapeutic agents for tissue repair with considerable advantages, including multifunctionality, specificity, biocompatibility, and biodegradability. However, the high complexity of tissue microenvironments and their inherent deficiencies such as short half-live and susceptibility to enzymatic degradation, adversely affect their therapeutic efficacy and clinical applications. Investigating the fundamental mechanisms by which BAPPs modulate the microenvironment and developing rational delivery strategies are essential for optimizing their administration in distinct tissue repairs and facilitating clinical translation. This review initially focuses on the mechanisms through which BAPPs influence the microenvironment for tissue repair via reactive oxygen species, blood and lymphatic vessels, immune cells, and repair cells. Then, a variety of delivery platforms, including scaffolds and hydrogels, electrospun fibers, surface coatings, assisted particles, nanotubes, two-dimensional nanomaterials, and nanoparticles engineered cells, are summarized to incorporate BAPPs for effective tissue repair, modification strategies aimed at enhancing loading efficiencies and release kinetics are also reviewed. Additionally, the delivery of BAPPs can be precisely regulated by endogenous stimuli (glucose, reactive oxygen species, enzymes, pH) or exogenous stimuli (ultrasound, heat, light, magnetic field, and electric field) to achieve on-demand release tailored for specific tissue repair needs. Furthermore, this review focuses on the clinical potential of BAPPs in facilitating tissue repair across various types, including bone, cartilage, intervertebral discs, muscle, tendons, periodontal tissues, skin, myocardium, nervous system (encompassing brain, spinal cord, and peripheral nerve), endometrium, as well as ear and ocular tissue. Finally, current challenges and prospects are discussed.
Collapse
Affiliation(s)
- Zhuo-Wen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhe-Yuan Zhang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ze-Pu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jia-Yao Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Tian-Hong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Guang Shi
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Han-Ke Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jun-Wu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Min-Chao Dong
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Li Hong
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Jing-Feng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
4
|
He A, Huang Y, Cao C, Li X. Advances in drug delivery systems utilizing blood cells and their membrane-derived microvesicles. Drug Deliv 2024; 31:2425156. [PMID: 39520082 PMCID: PMC11552282 DOI: 10.1080/10717544.2024.2425156] [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: 08/01/2024] [Revised: 10/11/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
The advancement of drug delivery systems (DDSs) in recent decades has demonstrated significant potential in enhancing the efficacy of pharmacological agents. Despite the approval of certain DDSs for clinical use, challenges such as rapid clearance from circulation, toxic accumulation in the body, and ineffective targeted delivery persist as obstacles to successful clinical application. Blood cell-based DDSs have emerged as a popular strategy for drug administration, offering enhanced biocompatibility, stability, and prolonged circulation. These DDSs are well-suited for systemic drug delivery and have played a crucial role in formulating optimal drug combinations for treating a variety of diseases in both preclinical studies and clinical trials. This review focuses on recent advancements and applications of DDSs utilizing blood cells and their membrane-derived microvesicles. It addresses the current therapeutic applications of blood cell-based DDSs at the organ and tissue levels, highlighting their successful deployment at the cellular level. Furthermore, it explores the mechanisms of cellular uptake of drug delivery vectors at the subcellular level. Additionally, the review discusses the opportunities and challenges associated with these DDSs.
Collapse
Affiliation(s)
- Andong He
- Center for Medical and Engineering Innovation, Central Laboratory, The First Affiliated Hospital, Ningbo University School of Medicine, Ningbo, China
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| | - Yuye Huang
- Center for Medical and Engineering Innovation, Central Laboratory, The First Affiliated Hospital, Ningbo University School of Medicine, Ningbo, China
| | - Chao Cao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Xuejin Li
- Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| |
Collapse
|
5
|
Yang W, Lin P, Gao R, Fang Z, Wang Z, Ma Z, Shi J, Yu W. Cell-derived biomimetic drug delivery system for inflammatory bowel disease therapy. Mater Today Bio 2024; 29:101332. [PMID: 39606424 PMCID: PMC11600033 DOI: 10.1016/j.mtbio.2024.101332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/24/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic recurrent disease with an increasing incidence year by year. At present, no safe and effective treatment for IBD exists. Thus, there is an urgent need to create new therapeutic options that have decreased adverse effects and positive clinical efficacy. A range of nanomaterials have fueled the advancement of nanomedicine in recent years, which is establishing more appealing and prospective treatment approaches for IBD. However, traditional synthetic nanomaterials still have some problems in the IBD drug delivery process, such as weak targeting ability of vectors, difficulty escaping immune surveillance, and poor biosecurity. Natural sources of biological nanomaterials have been identified to solve the above problems. A drug delivery system based on bionic technology is expected to achieve a new breakthrough in the targeted therapy of IBD by nanotechnology due to its organic integration of low immunogenicity and natural targeting of biological materials and the controllability and versatility of synthetic nanocarrier design. We begin this review by outlining the fundamental traits of both inflammatory and healthy intestinal microenvironments. Subsequently, we review the latest application of a cell-derived bionic drug delivery system in IBD therapy. Finally, we discuss the development prospects of this delivery system and challenges to its clinical translation. Biomimetic nanotherapy is believed to offer a new strategy for the treatment of IBD.
Collapse
Affiliation(s)
- Wenjing Yang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Peihong Lin
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Rui Gao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Zhengyu Fang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Zhouru Wang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Zhen Ma
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Jing Shi
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Wenying Yu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China
| |
Collapse
|
6
|
Ran K, Wang J, Li D, Jiang Z, Ding B, Yu F, Hu S, Wang L, Sun W, Xu H. Sustained-release of SOD from multivesicular liposomes accelerated the colonic mucosal healing of colitis mice by inhibiting oxidative stress. Colloids Surf B Biointerfaces 2024; 243:114143. [PMID: 39128435 DOI: 10.1016/j.colsurfb.2024.114143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/19/2024] [Accepted: 08/04/2024] [Indexed: 08/13/2024]
Abstract
Oxidative stress has long been known as a pathogenic factor of ulcerative colitis. Superoxide dismutase (SOD) has been demonstrated to mitigate gut mucosal injury via combating oxidative stress. Herein, we developed SOD-loaded multivesicular liposomes (S-MVLs) as sustained-release depot for ulcerative colitis treatment. S-MVLs were spherical honeycomb-like particles with average particle size of 27.3 ± 5.4 μm and encapsulating efficiency of 78.7 ± 2.6 %. Moreover, the two-phase release profiles of SOD from S-MVLs were exhibited, that was, the burst release phase within 4 h and the sustained-release phase within 96 h. After intraperitoneally injecting S-MVLs, in situ retention time of SOD at bowel cavity extended by 4-fold in comparison with SOD solution. In vitro cells experiment showed that S-MVLs had the protective effect on LPS-treated RAW 264.7 cells via scavenging ROS and inhibiting pro-inflammatory cytokines production. S-MVLs ameliorated the body weight loss, DAI score and the colon shortening of colitis mice. Meanwhile, the colonic morphology and the epithelial barrier of colitis mice were effectively recovered after S-MVLs treatment. The therapeutic mechanism might be associated with polymerizing M1 macrophages to M2 phenotypes and alleviating oxidative stress. Collectively, multivesicular liposomes might be a promising sustained-release depot of SOD for ulcerative colitis treatments.
Collapse
Affiliation(s)
- Kunjie Ran
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Cixi Biomedical Research Institute of Wenzhou Medical University, China
| | - Jie Wang
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Cixi Biomedical Research Institute of Wenzhou Medical University, China
| | - Dingwei Li
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Cixi Biomedical Research Institute of Wenzhou Medical University, China
| | - Zhijiang Jiang
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Cixi Biomedical Research Institute of Wenzhou Medical University, China
| | - Bingyu Ding
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Cixi Biomedical Research Institute of Wenzhou Medical University, China
| | - Fengnan Yu
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Cixi Biomedical Research Institute of Wenzhou Medical University, China
| | - Sunkuan Hu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province 325000, China
| | - Lifen Wang
- Research Center for Drug Safety Evaluation, Hainan Medical University, Haikou City, Hainan Province, China.
| | - Wenwen Sun
- Pathology Department, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China.
| | - Helin Xu
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; Cixi Biomedical Research Institute of Wenzhou Medical University, China.
| |
Collapse
|
7
|
Qiu S, Zhu F, Tong L. Application of targeted drug delivery by cell membrane-based biomimetic nanoparticles for inflammatory diseases and cancers. Eur J Med Res 2024; 29:523. [PMID: 39472940 PMCID: PMC11523786 DOI: 10.1186/s40001-024-02124-8] [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: 08/02/2024] [Accepted: 10/23/2024] [Indexed: 11/02/2024] Open
Abstract
Drug-carrying nanoparticles can be recognized and captured by macrophages and cleared away by the immune system, resulting in reduced drug efficacy and representing the main drawbacks. Biomimetic nanoparticles, which are coated with cell membranes from natural resources, have been applied to address this problem. This type of nanoparticle maintains some specific biological activities, allowing them to carry drugs reaching designated tissues effectively and have a longer time in circulation. This review article aims to summarize recent progress on biomimetic nanoparticles based on cell membranes. In this paper, we have introduced the classification of biomimetic nanoparticles, their preparation and characterization, and their applications in inflammatory diseases and malignant tumors. We have also analyzed the shortcomings and prospects of this technology, hoping to provide some clues for basic researchers and clinicians engaged in this field.
Collapse
Affiliation(s)
- Shijie Qiu
- Department of General Surgery, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, 163316, Heilongjiang Province, China
| | - Feifan Zhu
- Department of General Surgery, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, 163316, Heilongjiang Province, China
| | - Liquan Tong
- Department of General Surgery, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, 163316, Heilongjiang Province, China.
| |
Collapse
|
8
|
Ge D, An R, Xue L, Qiu M, Zhu Y, Wen G, Shi Y, Ren H, Li W, Wang J. Developing Cell-Membrane-Associated Liposomes for Liver Diseases. ACS NANO 2024; 18:29421-29438. [PMID: 39404084 DOI: 10.1021/acsnano.4c12122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Over the past decade, a marked escalation in the prevalence of hepatic pathologies has been observed, adversely impacting the quality of life for many. The predominant therapeutic strategy for liver diseases has been pharmacological intervention; however, its efficacy is often constrained. Currently, liposomes are tiny structures that can deliver drugs directly to targeted areas, enhancing their effectiveness. Specifically, cell membrane-associated liposomes have gained significant attention. Despite this, there is still much to learn about the binding mechanism of this type of liposome. Thus, this review comprehensively summarizes relevant information on cell membrane-associated liposomes, including their clinical applications and future development directions. First, we will briefly introduce the composition and types of cell membrane-associated liposomes. We will provide an overview of their structure and discuss the various types of liposomes associated with cell membranes. Second, we will thoroughly discuss various strategies of drug delivery using these liposomes. Lastly, we will discuss the application and clinical challenges associated with using cell membrane-associated liposomes in treating liver diseases. We will explore their potential benefits while also addressing the obstacles that need to be overcome. Furthermore, we will provide prospects for future development in this field. In summary, this review underscores the promise of cell membrane-associated liposomes in enhancing liver disease treatment and highlights the need for further research to optimize their utilization. In summary, this review underscores the promise of cell membrane-associated liposomes in enhancing liver disease treatment and highlights the need for further research to optimize their utilization.
Collapse
Affiliation(s)
- Dongxue Ge
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Ran An
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Lingling Xue
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Mengdi Qiu
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Yawen Zhu
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Gaolin Wen
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Yunpeng Shi
- Department of Hepatobiliary and Pancreatic Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Haozhen Ren
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Wei Li
- Department of Neurosurgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Jinglin Wang
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| |
Collapse
|
9
|
Weng MT, Hsiung CY, Wei SC, Chen Y. Nanotechnology for Targeted Inflammatory Bowel Disease Therapy: Challenges and Opportunities. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1999. [PMID: 39439396 DOI: 10.1002/wnan.1999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024]
Abstract
Inflammatory bowel disease (IBD) is a complex and recurring inflammatory disorder that affects the gastrointestinal tract and is influenced by genetic predisposition, immune dysregulation, the gut microbiota, and environmental factors. Advanced therapies, such as biologics and small molecules, target diverse immune pathways to manage IBD. Nanoparticle (NP)-based drugs have emerged as effective tools, offering controlled drug release and targeted delivery. This review highlights NP modifications for anti-inflammatory purposes, utilizing changes such as those in size, charge, redox reactions, and ligand-receptor interactions in drug delivery systems. By using pathological and microenvironmental cues to guide NP design, precise targeting can be achieved. In IBD, a crucial aspect of NP intervention is targeting specific types of cells, such as immune and epithelial cells, to address compromised intestinal barrier function and reduce overactive immune responses. This review also addresses current challenges and future prospects, with the goal of advancing the development of NP-mediated strategies for IBD treatment.
Collapse
Affiliation(s)
- Meng-Tzu Weng
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Hsin-Chu Branch, Hsinchu, Taiwan
| | - Chia-Yueh Hsiung
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Shu-Chen Wei
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
10
|
Han R, Ren Z, Wang Q, Zha H, Wang E, Wu M, Zheng Y, Lu J. Synthetic Biomimetic Liposomes Harness Efferocytosis Machinery for Highly Efficient Macrophages-Targeted Drug Delivery to Alleviate Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308325. [PMID: 38790144 PMCID: PMC11304272 DOI: 10.1002/advs.202308325] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 03/20/2024] [Indexed: 05/26/2024]
Abstract
Macrophages play pivotal roles in the regulation of inflammatory responses and tissue repair, making them a prime target for inflammation alleviation. However, the accurate and efficient macrophages targeting is still a challenging task. Motivated by the efficient and specific removal of apoptotic cells by macrophages efferocytosis, a novel biomimetic liposomal system called Effero-RLP (Efferocytosis-mediated Red blood cell hybrid Liposomes) is developed which incorporates the membrane of apoptotic red blood cells (RBCs) with liposomes for the purpose of highly efficient macrophages targeting. Rosiglitazone (ROSI), a PPARγ agonist known to attenuate macrophage inflammatory responses, is encapsulated into Effero-RLP as model drug to regulate macrophage functions in DSS-induced colitis mouse model. Intriguingly, the Effero-RLP exhibits selective and efficient uptake by macrophages, which is significantly inhibited by the efferocytosis blocker Annexin V. In animal models, the Effero-RLP demonstrates rapid recognition by macrophages, leading to enhanced accumulation at inflammatory sites. Furthermore, ROSI-loaded Effero-RLP effectively alleviates inflammation and protects colon tissue from injury in the colitis mouse model, which is abolished by deletion of macrophages from mice model. In conclusion, the study highlights the potential of macrophage targeting using efferocytosis biomimetic liposomes. The development of Effero-RLP presents novel and promising strategies for alleviating inflammation.
Collapse
Affiliation(s)
- Run Han
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical Sciences (ICMS)University of MacauMacau999078China
| | - Zhengyu Ren
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical Sciences (ICMS)University of MacauMacau999078China
| | - Qi Wang
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical Sciences (ICMS)University of MacauMacau999078China
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM)Nanjing University of Posts & TelecommunicationsNanjing210023China
| | - Haidong Zha
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical Sciences (ICMS)University of MacauMacau999078China
| | - Erjin Wang
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical Sciences (ICMS)University of MacauMacau999078China
| | - Mingyue Wu
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical Sciences (ICMS)University of MacauMacau999078China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical Sciences (ICMS)University of MacauMacau999078China
- Faculty of Health SciencesUniversity of MacauMacau999078China
| | - Jia‐Hong Lu
- State Key Laboratory of Quality Research in Chinese MedicineInstitute of Chinese Medical Sciences (ICMS)University of MacauMacau999078China
- Faculty of Health SciencesUniversity of MacauMacau999078China
- Guangdong‐Hong Kong‐Macau Joint Lab on Chinese Medicine and Immune Disease ResearchUniversity of MacauMacau999078China
| |
Collapse
|
11
|
Wang HC, Yang W, Xu L, Han YH, Lin Y, Lu CT, Kim K, Zhao YZ, Yu XC. BV2 Membrane-Coated PEGylated-Liposomes Delivered hFGF21 to Cortical and Hippocampal Microglia for Alzheimer's Disease Therapy. Adv Healthc Mater 2024; 13:e2400125. [PMID: 38513154 DOI: 10.1002/adhm.202400125] [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: 01/12/2024] [Revised: 03/18/2024] [Indexed: 03/23/2024]
Abstract
Microglia-mediated inflammation is involved in the pathogenesis of Alzheimer's disease (AD), whereas human fibroblast growth factor 21 (hFGF21) has demonstrated the ability to regulate microglia activation in Parkinson's disease, indicating a potential therapeutic role in AD. However, challenges such as aggregation, rapid inactivation, and the blood-brain barrier hinder its effectiveness in treating AD. This study develops targeted delivery of hFGF21 to activated microglia using BV2 cell membrane-coated PEGylated liposomes (hFGF21@BCM-LIP), preserving the bioactivity of hFGF21. In vitro, hFGF21@BCM-LIP specifically targets Aβ1-42-induced BV2 cells, with uptake hindered by anti-VCAM-1 antibody, indicating the importance of VCAM-1 and integrin α4/β1 interaction in targeted delivery to BV2 cells. In vivo, following subcutaneous injection near the lymph nodes of the neck, hFGF21@BCM-LIP diffuses into lymph nodes and distributes along the meningeal lymphatic vasculature and brain parenchyma in amyloid-beta (Aβ1-42)-induced mice. Furthermore, the administration of hFGF21@BCM-LIP to activated microglia improves cognitive deficits caused by Aβ1-42 and reduces levels of tau, p-Tau, and BACE1. It also decreases interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) release while increasing interleukin-10 (IL-10) release both in vivo and in vitro. These results indicate that hFGF21@BCM-LIP can be a promising treatment for AD, by effectively crossing the blood-brain barrier and targeting delivery to brain microglia via the neck-meningeal lymphatic vasculature-brain parenchyma pathways.
Collapse
Affiliation(s)
- Heng-Cai Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Wei Yang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
| | - Ling Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
| | - Yong-Hui Han
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang Province, 325101, China
| | - Yi Lin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
| | - Cui-Tao Lu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
| | - Kwonseop Kim
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Ying-Zheng Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, Zhejiang Province, 315302, China
| | - Xi-Chong Yu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province, 325035, China
| |
Collapse
|
12
|
Gazzi R, Gelli R, Aleandri S, Carone M, Luciani P. Bioinspired and bioderived nanomedicine for inflammatory bowel disease. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1986. [PMID: 39140489 DOI: 10.1002/wnan.1986] [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: 04/05/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 08/15/2024]
Abstract
Due to its chronic nature and complex pathophysiology, inflammatory bowel disease (IBD) poses significant challenges for treatment. The long-term therapies for patients, often diagnosed between the ages of 20 and 40, call for innovative strategies to target inflammation, minimize systemic drug exposure, and improve patients' therapeutic outcomes. Among the plethora of strategies currently pursued, bioinspired and bioderived nano-based formulations have garnered interest for their safety and versatility in the management of IBD. Bioinspired nanomedicine can host and deliver not only small drug molecules but also biotherapeutics, be made gastroresistant and mucoadhesive or mucopenetrating and, for these reasons, are largely investigated for oral administration, while surprisingly less for rectal delivery, recommended first-line treatment approach for several IBD patients. The use of bioderived nanocarriers, mostly extracellular vesicles (EVs), endowed with unique homing abilities, is still in its infancy with respect to the arsenal of nanomedicine under investigation for IBD treatment. An emerging source of EVs suited for oral administration is ingesta, that is, plants or milk, thanks to their remarkable ability to resist the harsh environment of the upper gastrointestinal tract. Inspired by the unparalleled properties of natural biomaterials, sophisticated avenues for enhancing therapeutic efficacy and advancing precision medicine approaches in IBD care are taking shape, although bottlenecks arising either from the complexity of the nanomedicine designed or from the lack of a clear regulatory pathway still hinder a smooth and efficient translation to the clinics. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Rafaela Gazzi
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Rita Gelli
- Department of Chemistry "Ugo Schiff" and CSGI, University of Florence, Florence, Italy
| | - Simone Aleandri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Marianna Carone
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Paola Luciani
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| |
Collapse
|
13
|
Huang Z, Li X, Yu D, Wang H, Chun C, Zhao Y. Efferocytosis-Inspired Biomimetic Nanoplatform for Targeted Acute Lung Injury Therapy. Adv Healthc Mater 2024; 13:e2304304. [PMID: 38306647 DOI: 10.1002/adhm.202304304] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/30/2024] [Indexed: 02/04/2024]
Abstract
Acute lung injury (ALI) is a serious inflammatory disease that causes impairment of pulmonary function. Phenotypic modulation of macrophage in the lung using fibroblast growth factor 21 (FGF21) may be a potential strategy to alleviate lung inflammation. Consequently, achieving specific delivery of FGF21 to the inflamed lung and subsequent efficient FGF21 internalization by macrophages within the lung becomes critical for effective ALI treatment. Here, an apoptotic cell membrane-coated zirconium-based metal-organic framework UiO-66 is reported for precise pulmonary delivery of FGF21 (ACM@U-FGF21) whose design is inspired by the process of efferocytosis. ACM@U-FGF21 with apoptotic signals is recognized and internalized by phagocytes in the blood and macrophages in the lung, and then the intracellular ACM@U-FGF21 can inhibit the excessive secretion of pro-inflammatory cytokines by these cells to relieve the inflammation. Utilizing the homologous targeting properties inherited from the source cells and the spontaneous recruitment of immune cells to inflammatory sites, ACM@U-FGF21 can accumulate preferentially in the lung after injection. The results prove that ACM@U-FGF21 effectively reduces inflammatory damage to the lung by modulating lung macrophage polarization and suppressing the excessive secretion of pro-inflammatory cytokines by activated immune cells. This study demonstrates the usefulness of efferocytosis-inspired ACM@U-FGF21 in the treatment of ALI.
Collapse
Affiliation(s)
- Zhiwei Huang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Xinze Li
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China
| | - Dedong Yu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hengcai Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Changju Chun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yingzheng Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo, 315300, China
| |
Collapse
|
14
|
Yuan S, Hu Q. Convergence of nanomedicine and neutrophils for drug delivery. Bioact Mater 2024; 35:150-166. [PMID: 38318228 PMCID: PMC10839777 DOI: 10.1016/j.bioactmat.2024.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/20/2024] [Accepted: 01/21/2024] [Indexed: 02/07/2024] Open
Abstract
Neutrophils have recently emerged as promising carriers for drug delivery due to their unique properties including rapid response toward inflammation, chemotaxis, and transmigration. When integrated with nanotechnology that has enormous advantages in improving treatment efficacy and reducing side effects, neutrophil-based nano-drug delivery systems have expanded the repertoire of nanoparticles employed in precise therapeutic interventions by either coating nanoparticles with their membranes, loading nanoparticles inside living cells, or engineering chimeric antigen receptor (CAR)-neutrophils. These neutrophil-inspired therapies have shown superior biocompatibility, targeting ability, and therapeutic robustness. In this review, we summarized the benefits of combining neutrophils and nanotechnologies, the design principles and underlying mechanisms, and various applications in disease treatments. The challenges and prospects for neutrophil-based drug delivery systems were also discussed.
Collapse
Affiliation(s)
- Sichen Yuan
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, 53705, United States
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, United States
| |
Collapse
|
15
|
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
|
16
|
Peng X, Chen J, Gan Y, Yang L, Luo Y, Bu C, Huang Y, Chen X, Tan J, Yang YY, Yuan P, Ding X. Biofunctional lipid nanoparticles for precision treatment and prophylaxis of bacterial infections. SCIENCE ADVANCES 2024; 10:eadk9754. [PMID: 38578994 PMCID: PMC10997193 DOI: 10.1126/sciadv.adk9754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 03/04/2024] [Indexed: 04/07/2024]
Abstract
The lack of bacterial-targeting function in antibiotics and their prophylactic usage have caused overuse of antibiotics, which lead to antibiotic resistance and inevitable long-term toxicity. To overcome these issues, we develop neutrophil-bacterial hybrid cell membrane vesicle (HMV)-coated biofunctional lipid nanoparticles (LNP@HMVs), which are designed to transport antibiotics specifically to bacterial cells at the infection site for the effective treatment and prophylaxis of bacterial infection. The dual targeting ability of HMVs to inflammatory vascular endothelial cells and homologous Gram-negative bacterial cells results in targeted accumulation of LNP@HMVs in the site of infections. LNP@HMVs loaded with the antibiotic norfloxacin not only exhibit enhanced activity against planktonic bacteria and bacterial biofilms in vitro but also achieve potent therapeutic efficacy in treating both systemic infection and lung infection. Furthermore, LNP@HMVs trigger the activation of specific humoral and cellular immunity to prevent bacterial infection. Together, LNP@HMVs provide a promising strategy to effectively treat and prevent bacterial infection.
Collapse
Affiliation(s)
- Xinran Peng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Jiaoyu Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Yingying Gan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Li Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Yuanjing Luo
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Changxin Bu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Yi Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Xinhai Chen
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen 518132, PR China
| | - Jeremy Tan
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Yi Yan Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
| | - Xin Ding
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, PR China
- State Key Laboratory of Anti-Infective Drug Discovery and Development; School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
17
|
Gao Q, Yan Y, Zhang J, Li X, Wang J, Feng Y, Li P, Wang H, Zhang Y, He L, Shan Z, Li B. Autologous cryo-shocked neutrophils enable targeted therapy of sepsis via broad-spectrum neutralization of pro-inflammatory cytokines and endotoxins. Front Chem 2024; 12:1359946. [PMID: 38449477 PMCID: PMC10914999 DOI: 10.3389/fchem.2024.1359946] [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: 12/22/2023] [Accepted: 01/25/2024] [Indexed: 03/08/2024] Open
Abstract
Background: Sepsis is a life-threatening disease characterized by multiple organ failure due to excessive activation of the inflammatory response and cytokine storm. Despite recent advances in the clinical use of anti-cytokine biologics, sepsis treatment efficacy and improvements in mortality remain unsatisfactory, largely due to the mechanistic complexity of immune regulation and cytokine interactions. Methods: In this study, a broad-spectrum anti-inflammatory and endotoxin neutralization strategy was developed based on autologous "cryo-shocked" neutrophils (CS-Neus) for the management of sepsis. Neutrophils were frozen to death using a novel liquid nitrogen "cryo-shock" strategy. The CS-Neus retained the source cell membrane structure and functions related to inflammatory site targeting, broad-spectrum inflammatory cytokines, and endotoxin (LPS) neutralizing properties. This strategy aimed to disable harmful pro-inflammatory functions of neutrophils, such as cytokine secretion. Autologous cell-based therapy strategies were employed to avoid immune rejection and enhance treatment safety. Results: In both LPS-induced sepsis mouse models and clinical patient-derived blood samples, CS-Neus treatment significantly ameliorated cytokine storms by removing inflammatory cytokines and endotoxin. The therapy showed notable anti-inflammatory therapeutic effects and improved the survival rate of mice. Discussion: The results of this study demonstrate the potential of autologous "cryo-shocked" neutrophils as a promising therapeutic approach for managing sepsis. By targeting inflammatory organs and exhibiting anti-inflammatory activity, CS-Neus offer a novel strategy to combat the complexities of sepsis treatment. Further research and clinical trials are needed to validate the efficacy and safety of this approach in broader populations and settings.
Collapse
Affiliation(s)
- Qiuxia Gao
- School of Inspection, Ningxia Medical University, Yinchuan, Ningxia, China
- Institute of Translational Medicine, The First People’s Hospital of Foshan, Foshan, Guangdong, China
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yan Yan
- Department of Critical Care Medicine, Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jie Zhang
- Engineering Research Center of Tibetan Medicine Detection Technology, Ministry of Education, Xizang Minzu University, Xianyang, Shaanxi, China
| | - Xiaoxue Li
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Jiamei Wang
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yi Feng
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Peiran Li
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Huanhuan Wang
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yunlong Zhang
- Department of Critical Care Medicine, Department of Emergency, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Lingjie He
- Molecular Diagnosis and Treatment Center for Infectious Diseases, Dermatology Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Zhiyan Shan
- Department of Histology and Embryology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Bin Li
- School of Inspection, Ningxia Medical University, Yinchuan, Ningxia, China
- Institute of Translational Medicine, The First People’s Hospital of Foshan, Foshan, Guangdong, China
| |
Collapse
|
18
|
Shabana S, Hamouda HI, Hamadou AH, Ahmed B, Chi Z, Liu C. Marine phospholipid nanoliposomes: A promising therapeutic approach for inflammatory bowel disease: Preparation, safety, and efficacy evaluation. Colloids Surf B Biointerfaces 2024; 234:113702. [PMID: 38113752 DOI: 10.1016/j.colsurfb.2023.113702] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
Promising findings have been emerged from studies utilizing n3 polyunsaturated fatty acids (PUFA) supplementation in animal models of inflammatory bowel disease (IBD). Introduction of marine phospholipids which combine n3 PUFA with phosphatidylcholine in a nanoliposome formulation offers enhanced pharmacological efficacy due to physical stability, improved bioavailability, and specific targeting to inflamed colitis tissues. In the present study, a marine phospholipid-based nanoliposome formulation was developed and optimized, resulting in nanovesicles of approximately 107.7 ± 1.3 nm in size, 0.18 ± 0.01 PDI, and - 32.03 ± 3.16 mV ZP. The nanoliposomes exhibited spherical vesicles with stable properties upon incubation at SGF as shown by the TEM, DLS, and turbidity measurements over 3 h. MPL nanoliposomes were cytocompatible until the concentration of 500 µg/mL as per MTT assay and taken by macrophages through macropinocytosis and caveolae pathways, and demonstrated significant inhibitory activity against reactive oxygen species (ROS) in LPS-stimulated macrophages. They were also shown to be blood-compatible and safe for administration in healthy mice. In a colitis mouse model, the nanoliposomes displayed preferential distribution in the inflamed gut, delaying the onset of colitis when administered prophylactically. These findings highlight the potential of marine phospholipid nanoliposomes as a promising therapeutic approach for managing inflammatory bowel disease.
Collapse
Affiliation(s)
- Samah Shabana
- Department of Biochemistry and Molecular Biology, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China; Egyptian Ministry of Health and Population, Cairo 11516, Egypt
| | - Hamed I Hamouda
- Dalian Engineering Research Center for Carbohydrate Agricultural Preparations, Liaoning Provincial Key Laboratory of Carbohydrates, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, CAS, Dalian 116023, PR China; Processes Development Department, Egyptian Petroleum Research Institute (EPRI), Nasr City 11727, Cairo, Egypt
| | | | - Busati Ahmed
- Department of Biochemistry and Molecular Biology, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Zhe Chi
- Department of Biochemistry and Molecular Biology, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Chenguang Liu
- Department of Biochemistry and Molecular Biology, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China.
| |
Collapse
|
19
|
Mohammad-Rafiei F, Khojini JY, Ghazvinian F, Alimardan S, Norioun H, Tahershamsi Z, Tajbakhsh A, Gheibihayat SM. Cell membrane biomimetic nanoparticles in drug delivery. Biotechnol Appl Biochem 2023; 70:1843-1859. [PMID: 37387120 DOI: 10.1002/bab.2487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 06/05/2023] [Indexed: 07/01/2023]
Abstract
Despite the efficiency of nanoparticle (NP) therapy, in vivo investigations have shown that it does not perform as well as in vitro. In this case, NP confronts many defensive hurdles once they enter the body. The delivery of NP to sick tissue is inhibited by these immune-mediated clearance mechanisms. Hence, using a cell membrane to hide NP for active distribution offers up a new path for focused treatment. These NPs are better able to reach the disease's target location, leading to enhanced therapeutic efficacy. In this emerging class of drug delivery vehicles, the inherent relation between the NPs and the biological components obtained from the human body was utilized, which mimic the properties and activities of native cells. This new technology has shown the viability of using biomimicry to evade immune system-provided biological barriers, with an emphasis on restricting clearance from the body before reaching its intended target. Furthermore, by providing signaling cues and transplanted biological components that favorably change the intrinsic immune response at the disease site, the NPs would be capable interacting with immune cells regarding the biomimetic method. Thus, we aimed to provide a current landscape and future trends of biomimetic NPs in drug delivery.
Collapse
Affiliation(s)
- Fatemeh Mohammad-Rafiei
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Javad Yaghmoorian Khojini
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Ghazvinian
- Department of Life science and biotechnology, Faculty of Natural Sciences, University of Shahid Beheshti, Tehran, Iran
| | - Sajad Alimardan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Norioun
- Medical Genetics Department, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Munich, Germany
| |
Collapse
|
20
|
Liu L, Zhang Y, Li X, Deng J. Microenvironment of pancreatic inflammation: calling for nanotechnology for diagnosis and treatment. J Nanobiotechnology 2023; 21:443. [PMID: 37996911 PMCID: PMC10666376 DOI: 10.1186/s12951-023-02200-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Acute pancreatitis (AP) is a common and life-threatening digestive disorder. However, its diagnosis and treatment are still impeded by our limited understanding of its etiology, pathogenesis, and clinical manifestations, as well as by the available detection methods. Fortunately, the progress of microenvironment-targeted nanoplatforms has shown their remarkable potential to change the status quo. The pancreatic inflammatory microenvironment is typically characterized by low pH, abundant reactive oxygen species (ROS) and enzymes, overproduction of inflammatory cells, and hypoxia, which exacerbate the pathological development of AP but also provide potential targeting sites for nanoagents to achieve early diagnosis and treatment. This review elaborates the various potential targets of the inflammatory microenvironment of AP and summarizes in detail the prospects for the development and application of functional nanomaterials for specific targets. Additionally, it presents the challenges and future trends to develop multifunctional targeted nanomaterials for the early diagnosis and effective treatment of AP, providing a valuable reference for future research.
Collapse
Affiliation(s)
- Lu Liu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Street, Nanchong, 637001, China
| | - Yiqing Zhang
- Institute of Burn Research Southwest Hospital State Key Lab of Trauma Burn and Combined Injury Chongqing Key Laboratory for Disease Proteomics Army Medical University, Chongqing, 400038, China
- Research Center for Tissue Repair and Regeneration Affiliated to the Medical Innovation Research Division and the 4th Medical Center of Chinese PLA General Hospita, PLA Medical College, 28 Fu Xing Road, Beijing, 100853, China
| | - Xinghui Li
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Street, Nanchong, 637001, China.
| | - Jun Deng
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, 1 South Maoyuan Street, Nanchong, 637001, China.
- Institute of Burn Research Southwest Hospital State Key Lab of Trauma Burn and Combined Injury Chongqing Key Laboratory for Disease Proteomics Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
21
|
Liu WS, Wu LL, Chen CM, Zheng H, Gao J, Lu ZM, Li M. Lipid-hybrid cell-derived biomimetic functional materials: A state-of-the-art multifunctional weapon against tumors. Mater Today Bio 2023; 22:100751. [PMID: 37636983 PMCID: PMC10448342 DOI: 10.1016/j.mtbio.2023.100751] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/29/2023] Open
Abstract
Tumors are among the leading causes of death worldwide. Cell-derived biomimetic functional materials have shown great promise in the treatment of tumors. These materials are derived from cell membranes, extracellular vesicles and bacterial outer membrane vesicles and may evade immune recognition, improve drug targeting and activate antitumor immunity. However, their use is limited owing to their low drug-loading capacity and complex preparation methods. Liposomes are artificial bionic membranes that have high drug-loading capacity and can be prepared and modified easily. Although they can overcome the disadvantages of cell-derived biomimetic functional materials, they lack natural active targeting ability. Lipids can be hybridized with cell membranes, extracellular vesicles or bacterial outer membrane vesicles to form lipid-hybrid cell-derived biomimetic functional materials. These materials negate the disadvantages of both liposomes and cell-derived components and represent a promising delivery platform in the treatment of tumors. This review focuses on the design strategies, applications and mechanisms of action of lipid-hybrid cell-derived biomimetic functional materials and summarizes the prospects of their further development and the challenges associated with it.
Collapse
Affiliation(s)
- Wen-Shang Liu
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, 200011, China
| | - Li-Li Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Cui-Min Chen
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Hao Zheng
- Department of Gastrointestinal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Zheng-Mao Lu
- Department of Gastrointestinal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Meng Li
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, 200011, China
| |
Collapse
|
22
|
Lei P, Yu H, Ma J, Du J, Fang Y, Yang Q, Zhang K, Luo L, Jin L, Wu W, Sun D. Cell membrane nanomaterials composed of phospholipids and glycoproteins for drug delivery in inflammatory bowel disease: A review. Int J Biol Macromol 2023; 249:126000. [PMID: 37532186 DOI: 10.1016/j.ijbiomac.2023.126000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/04/2023]
Abstract
Inflammatory bowel disease (IBD) is a serious chronic intestinal disorder with an increasing global incidence. However, current treatment strategies, such as anti-inflammatory drugs and probiotics, have limitations in terms of safety, stability, and effectiveness. The emergence of targeted nanoparticles has revolutionized IBD treatment by enhancing the biological properties of drugs and promoting efficiency and safety. Unlike synthetic nanoparticles, cell membrane nanomaterials (CMNs) consist primarily of biological macromolecules, including phospholipids, proteins, and sugars. CMNs include red blood cell membranes, macrophage membranes, and leukocyte membranes, which possess abundant glycoprotein receptors and ligands on their surfaces, allowing for the formation of cell-to-cell connections with other biological macromolecules. Consequently, they exhibit superior cell affinity, evade immune responses, and target inflammation effectively, making them ideal material for targeted delivery of IBD therapies. This review explores various CMNs delivery systems for IBD treatment. However, due to the complexity and harsh nature of the intestinal microenvironment, the lack of flexibility or loss of selectivity poses challenges in designing single CMNs delivery strategies. Therefore, we propose a hierarchically programmed delivery modality that combines CMNs with pH, charge, ROS and ligand-modified responsive nanoparticles. This approach significantly improves delivery efficiency and points the way for future research in this area.
Collapse
Affiliation(s)
- Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Haiyang Yu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiao Du
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Li Luo
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan, Guangdong 523059, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
23
|
Yang J, Li D, Zhang M, Lin G, Hu S, Xu H. From the updated landscape of the emerging biologics for IBDs treatment to the new delivery systems. J Control Release 2023; 361:568-591. [PMID: 37572962 DOI: 10.1016/j.jconrel.2023.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/06/2023] [Accepted: 08/06/2023] [Indexed: 08/14/2023]
Abstract
Inflammatory bowel diseases (IBDs) treatments have shifted from small-molecular therapeutics to the oncoming biologics. The first-line biologics against the moderate-to-severe IBDs are mainly involved in antibodies against integrins, cytokines and cell adhesion molecules. Besides, other biologics including growth factors, antioxidative enzyme, anti-inflammatory peptides, nucleic acids, stem cells and probiotics have also been explored at preclinical or clinical studies. Biologics with variety of origins have their unique potentials in attenuating immune inflammation or gut mucosa healing. Great advances in use of biologics for IBDs treatments have been archived in recent years. But delivering issues for biologic have also been confronted due to their liable nature. In this review, we will focus on biologics for IBDs treatments in the recent publications; summarize the current landscapes of biologics and their promise to control disease progress. Alternatively, the confronted challenges for delivering biologics will also be analyzed. To combat these drawbacks, some new delivering strategies are provided: firstly, designing the functional materials with high affinity toward biologics; secondly, the delivering vehicle systems to encapsulate the liable biologics; thirdly, the topical adhering delivery systems as enema. To our knowledge, this review is the first study to summarize the updated usage of the oncoming biologics for IBDs, their confronted challenges in term of delivery and the potential combating strategies.
Collapse
Affiliation(s)
- Jiaojiao Yang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Dingwei Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Mengjiao Zhang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Gaolong Lin
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Sunkuan Hu
- Department of Gastroenterology, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province 325000, China
| | - Helin Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China.
| |
Collapse
|
24
|
Lin G, Yang J, Liu J, Shangguan J, Pan H, Zhang Y, Ran K, Li D, Yu F, Xu H. In situ polyphenol-adhesive hydrogel enhanced the noncarcinogenic repairing of KGF on the gut epithelial barrier on TNBS-induced colitis rats. Int J Biol Macromol 2023; 231:123323. [PMID: 36669631 DOI: 10.1016/j.ijbiomac.2023.123323] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/09/2023] [Accepted: 01/14/2023] [Indexed: 01/19/2023]
Abstract
Ulcerative colitis (UC) is a chronic recurrent disease affecting the gastrointestinal tract especially colorectum. Keratinocyte growth factor (KGF) plays the vital roles in maintaining the colonic mucosal barrier. The poor stability and off-target of KGF were two hindering factors for its clinical application. Herein, in situ hydrogel (PE) with mucoadhesive ability was constructed by using temperature-sensitive poloxamer and EGCG as hydrogel-forming material and adhesive enhancer, respectively. Incorporation of EGCG led to the slight decrease of the gelled temperature and shortened the gelled time of PE hydrogel. When the concentration of EGCG is 0.1 %, PE hydrogel exhibits the suitable viscosity of 280 ± 20 Pa·s and the strong adhesive force of 725 ± 25 mN. KGF was soluble in cold PE solution to obtain KGF-loaded PE hydrogel (KGF@PE). PE hydrogel could improve the stability of KGF in vitro. KGF@PE not only could recover greatly the body weight of TNBS-induced rats but also repair their colonic morphology and goblet cell function. Moreover, the potential of repairing the epithelial barrier was indicated by upregulating tight junction proteins. Importantly, the safety of KGF@PE hydrogel for colitis was also confirmed on AOM/DSS-induced mice models. Conclusively, KGF@PE may be a promising therapeutic platform without obvious side effect for ulcerative colitis.
Collapse
Affiliation(s)
- Gaolong Lin
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Jiaojiao Yang
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Jiayi Liu
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Jianxun Shangguan
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Hanxiao Pan
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Yingying Zhang
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Kunjie Ran
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Dingwei Li
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Fengnan Yu
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Helin Xu
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China.
| |
Collapse
|
25
|
Prospective use of amniotic mesenchymal stem cell metabolite products for tissue regeneration. J Biol Eng 2023; 17:11. [PMID: 36759827 PMCID: PMC9912508 DOI: 10.1186/s13036-023-00331-1] [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: 09/09/2022] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Chronic disease can cause tissue and organ damage constituting the largest obstacle to therapy which, in turn, reduces patients' quality-adjusted life-year. Degenerative diseases such as osteoporosis, Alzheimer's disease, Parkinson's disease, and infectious conditions such as hepatitis, cause physical injury to organs. Moreover, damage resulting from chronic conditions such as diabetes can also culminate in the loss of organ function. In these cases, organ transplantation constitutes the therapy of choice, despite the associated problems of immunological rejection, potential disease transmission, and high morbidity rates. Tissue regeneration has the potential to heal or replace tissues and organs damaged by age, disease, or trauma, as well as to treat disabilities. Stem cell use represents an unprecedented strategy for these therapies. However, product availability and mass production remain challenges. A novel therapeutic alternative involving amniotic mesenchymal stem cell metabolite products (AMSC-MP) has been developed using metabolites from stem cells which contain cytokines and growth factors. Its potential role in regenerative therapy has recently been explored, enabling broad pharmacological applications including various gastrointestinal, lung, bladder and renal conditions, as well as the treatment of bone wounds, regeneration and skin aging due to its low immunogenicity and anti-inflammatory effects. The various kinds of growth factors present in AMSC-MP, namely bFGF, VEGF, TGF-β, EGF and KGF, have their respective functions and activities. Each growth factor is formed by different proteins resulting in molecules with various physicochemical properties and levels of stability. This knowledge will assist in the manufacture and application of AMSC-MP as a therapeutic agent.
Collapse
|
26
|
Mohajeri S, Moayedi S, Mohajeri S, Yadegar A, Haririan I. Targeting pathophysiological changes using biomaterials-based drug delivery systems: A key to managing inflammatory bowel disease. Front Pharmacol 2022; 13:1045575. [PMID: 36438794 PMCID: PMC9685402 DOI: 10.3389/fphar.2022.1045575] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/27/2022] [Indexed: 08/04/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a gastrointestinal disorder, affecting about several million people worldwide. Current treatments fail to adequately control some clinical symptoms in IBD patients, which can adversely impact the patient's quality of life. Hence, the development of new treatments for IBD is needed. Due to their unique properties such as biocompatibility and sustained release of a drug, biomaterials-based drug delivery systems can be regarded as promising candidates for IBD treatment. It is noteworthy that considering the pathophysiological changes occurred in the gastrointestinal tract of IBD patients, especially changes in pH, surface charge, the concentration of reactive oxygen species, and the expression of some biomolecules at the inflamed colon, can help in the rational design of biomaterials-based drug delivery systems for efficient management of IBD. Here, we discuss about targeting these pathophysiological changes using biomaterials-based drug delivery systems, which can provide important clues to establish a strategic roadmap for future studies.
Collapse
Affiliation(s)
- Sahar Mohajeri
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Moayedi
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Shabnam Mohajeri
- Faculty of Food Science and Technology, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Niu W, Chen Y, Wang L, Li J, Cui Z, Lv J, Yang F, Huo J, Zhang Z, Ju J. The combination of sodium alginate and chlorogenic acid enhances the therapeutic effect on ulcerative colitis by the regulation of inflammation and the intestinal flora. Food Funct 2022; 13:10710-10723. [PMID: 36173280 DOI: 10.1039/d2fo01619b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Chlorogenic acid (CA) and sodium alginate (SA) each have good therapeutic effects on ulcerative colitis (UC) owing to their antioxidant and anti-inflammatory activity. This study aimed to investigate the effects of CA alone and in combination with SA on inflammatory cells and UC mice. In the Lipopolysaccharide (LPS)-induced RAW 264.7 inflammatory cell model, Nitric oxide (NO) and interleukin-6 (IL-6) levels were significantly lower after treatment with CA plus SA than with CA alone. In the DSS-induced UC mouse model, compared with CA alone, CA plus SA showed a better ability to alleviate weight loss, reduce the disease activity index (DAI), improve the colonic mucosa, reduce the expression of inflammatory factors in the serum and myeloperoxidase (MPO) in colonic tissue, increase superoxide dismutase (SOD) levels, protect the intestinal mucosa and regulate the abundance of Actinobacteriota, Lactobacillus, Bifidobacterium, Bacteroides, Subdoligranulum and Streptococcus. Thus, CA plus SA can improve the therapeutic efficacy of CA in UC by regulating inflammatory factors, oxidative stress, and the intestinal flora and by protecting ulcerative wounds. These findings broaden our understanding of the role of the combination of SA and CA in enhancing the effects of CA on UC and provide strategies for prevention and treatment.
Collapse
Affiliation(s)
- Wei Niu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Yuxuan Chen
- School of Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ligui Wang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Jia Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Zhao Cui
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Jiajie Lv
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Fuyan Yang
- Anhui University of Chinese Medicine, Hefei, China
| | - Jiege Huo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Zhenhai Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| | - Jianming Ju
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China. .,Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
28
|
Hu Y, Zhang S, Wen Z, Fu H, Hu J, Ye X, Kang L, Li X, Yang X. Oral delivery of curcumin via multi-bioresponsive polyvinyl alcohol and guar gum based double-membrane microgels for ulcerative colitis therapy. Int J Biol Macromol 2022; 221:806-820. [PMID: 36099999 DOI: 10.1016/j.ijbiomac.2022.09.050] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 08/21/2022] [Accepted: 09/06/2022] [Indexed: 11/05/2022]
Abstract
Anti-inflammatory drugs for ulcerative colitis (UC) treatment should specifically penetrate and accumulate in the colon tissue. Herein, a multi-bioresponsive anti-inflammatory drug (curcumin, CUR)-loaded heterogeneous double-membrane microgels (CUR@microgels) for oral administration was fabricated in this study, in which the inner core was derived from polyvinyl alcohol (PVA) and guar gum (GG) and the outer gel was decoration with alginate and chitosan by polyelectrolyte interactions. The structure and morphology of microgels were characterized. In vitro, the formulation exhibited good bio-responses at different pH conditions and sustained-release properties in simulated colon fluid with a drug-release rate of 84.6 % over 34 h. With the assistance of the outlayer gels, the microgels effectively delayed the premature drug release of CUR in the upper gastrointestinal tract. In vivo studies revealed that CUR@microgels specifically accumulated in the colon tissue for 24 h, which suggest that the interlayer gels were apt to reach colon lesion. As expected, the oral administration of microgels remarkably alleviated the symptoms of UC and protected the colon tissue in DSS-induced UC mice. The above results indicated that these facilely fabricated microgels which exhibited excellent biocompatibility and multi-bioresponsive drug release, had an apparent effect on the treatment of UC, which represents a promising drug delivery strategy for CUR in a clinical application.
Collapse
Affiliation(s)
- Yan Hu
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China.
| | - Shangwen Zhang
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Zhijie Wen
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Hudie Fu
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Jie Hu
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Xuexin Ye
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Li Kang
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Xiaojun Li
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China
| | - Xinzhou Yang
- School of Pharmaceutical Science, South-Central MinZu University, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central MinZu University, Wuhan 430074, China; Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, South-Central MinZu University, Wuhan 430074, China.
| |
Collapse
|
29
|
Su JY, Li WH, Li YM. New opportunities for immunomodulation of the tumour microenvironment using chemical tools. Chem Soc Rev 2022; 51:7944-7970. [PMID: 35996977 DOI: 10.1039/d2cs00486k] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Immunotherapy is recognised as an attractive method for the treatment of cancer, and numerous treatment strategies have emerged over recent years. Investigations of the tumour microenvironment (TME) have led to the identification of many potential therapeutic targets and methods. However, many recently applied immunotherapies are based on previously identified strategies, such as boosting the immune response by combining commonly used stimulators, and the release of drugs through changes in pH. Although methodological improvements such as structural optimisation and combining strategies can be undertaken, applying those novel targets and methods in immunotherapy remains an important goal. In this review, we summarise the latest research on the TME, and discuss how small molecules, immune cells, and their interactions with tumour cells can be regulated in the TME. Additionally, the techniques currently employed for delivery of these agents to the TME are also mentioned. Strategies to modulate cell phenotypes and interactions between immune cells and tumours are mainly discussed. We consider both modulatory and targeting methods aiming to bridge the gap between the TME and chemical modulation thereof.
Collapse
Affiliation(s)
- Jing-Yun Su
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, 100084 Beijing, China.
| | - Wen-Hao Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, 100084 Beijing, China.
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, 100084 Beijing, China. .,Center for Synthetic and Systems Biology, Tsinghua University, 100084 Beijing, China.,Beijing Institute for Brain Disorders, 100069 Beijing, China
| |
Collapse
|
30
|
Rong R, Raza F, Liu Y, Yuan WE, Su J, Qiu M. Blood cell-based drug delivery systems: a biomimetic platform for antibacterial therapy. Eur J Pharm Biopharm 2022; 177:273-288. [PMID: 35868489 DOI: 10.1016/j.ejpb.2022.07.009] [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: 03/24/2022] [Revised: 06/28/2022] [Accepted: 07/18/2022] [Indexed: 11/18/2022]
Abstract
With the rapid increase in multidrug-resistance against antibiotics, higher doses of antibiotics or more effective antibiotics are needed to treat diseases, which ultimately leads to a decrease in the body's immunity and seriously threatens human health worldwide. The efficiency of antibiotics has been a large challenge for years. To overcome this problem, many carriers are utilized for anti-bacteria, attempting to optimize the delivery of such drugs and transport them safely and directly to the site of disease. Blood cell-based drug delivery systems present several advantages as compared to polymeric delivery system. These blood cells including red blood cells (RBCs), leukocytes, platelets. The blood cells and their membranes can both be used as drug carriers to deliver antibacterial drugs. In addition, blood cells can overcome many physiological/pathological obstacles faced by nanoparticles in vivo and effectively deliver drugs to the site of the disease. In this paper, we review studies on blood cell-based delivery systems used in antibacterial therapy, and analyze different roles in antibacterial therapy, which provide basis for further study in this field.
Collapse
Affiliation(s)
- Ruonan Rong
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China
| | - Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China
| | - Yuhao Liu
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China
| | - Wei-En Yuan
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China
| | - Jing Su
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China.
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, 800, Dongchuan Road, 200240 Shanghai, China.
| |
Collapse
|
31
|
Huang Z, Wang H, Long J, Lu Z, Chun C, Li X. Neutrophil Membrane-Coated Therapeutic Liposomes for Targeted Treatment in Acute Lung Injury. Int J Pharm 2022; 624:121971. [PMID: 35787461 PMCID: PMC9365401 DOI: 10.1016/j.ijpharm.2022.121971] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/22/2022] [Accepted: 06/29/2022] [Indexed: 10/26/2022]
Abstract
Acute lung injury (ALI) is one of the most common comorbidities associated with sepsis and can lead to acute respiratory distress syndrome. Intense inflammatory response due to excessive activation and uncontrolled infiltration of neutrophils are the central processes in the development of sepsis-induced ALI. In this study, a biomimetic nanoplatform that is a neutrophil membrane-coated liposome-loaded acidic fibroblast growth factor (aFGF@NMLs), which can selectively target the inflamed lung and effectively alleviate sepsis-induced ALI via inflammation suppression, was constructed. In vitro findings revealed that aFGF@NMLs has pro-inflammatory cytokine binding capabilities and can promote cellular uptake, substantially attenuate inflammatory responses, and enhance cellular antioxidant capacity. The in vivo results show that aFGF@NMLs can specifically accumulate in injured lungs in ALI mice after intravenous injection, thereby reducing the secretion of pro-inflammatory cytokines, inhibiting pulmonary cell apoptosis, and promoting lung function recovery. In conclusion, aFGF@NMLs demonstrated anti-inflammatory effects, mitigated the progression of ALI, and contributed to the disease prognosis. This research offers an innovative strategy and concept for the clinical treatment of diseases related to pulmonary inflammation.
Collapse
Affiliation(s)
- Zhiwei Huang
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea; School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Hengcai Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Juan Long
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325035, China
| | - Zhongqiu Lu
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325035, China
| | - Changju Chun
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Xinze Li
- Department of Emergency, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Wenzhou Key Laboratory of emergency and disaster medicine, Wenzhou 325035, China.
| |
Collapse
|
32
|
Yasmin F, Najeeb H, Shaikh S, Hasanain M, Naeem U, Moeed A, Koritala T, Hasan S, Surani S. Novel drug delivery systems for inflammatory bowel disease. World J Gastroenterol 2022; 28:1922-1933. [PMID: 35664964 PMCID: PMC9150062 DOI: 10.3748/wjg.v28.i18.1922] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/22/2022] [Accepted: 03/26/2022] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic illness characterized by relapsing inflammation of the intestines. The disorder is stratified according to the severity and is marked by its two main phenotypical representations: Ulcerative colitis and Crohn's disease. Pathogenesis of the disease is ambiguous and is expected to have interactivity between genetic disposition, environmental factors such as bacterial agents, and dysregulated immune response. Treatment for IBD aims to reduce symptom extent and severity and halt disease progression. The mainstay drugs have been 5-aminosalicylates (5-ASAs), corticosteroids, and immunosuppressive agents. Parenteral, oral and rectal routes are the conventional methods of drug delivery, and among all, oral administration is most widely adopted. However, problems of systematic drug reactions and low specificity in delivering drugs to the inflamed sites have emerged with these regular routes of delivery. Novel drug delivery systems have been introduced to overcome several therapeutic obstacles and for localized drug delivery to target tissues. Enteric-coated microneedle pills, various nano-drug delivery techniques, prodrug systems, lipid-based vesicular systems, hybrid drug delivery systems, and biologic drug delivery systems constitute some of these novel methods. Microneedles are painless, they dislodge their content at the affected site, and their release can be prolonged. Recombinant bacteria such as genetically engineered Lactococcus Lactis and eukaryotic cells, including GM immune cells and red blood cells as nanoparticle carriers, can be plausible delivery methods when evaluating biologic systems. Nano-particle drug delivery systems consisting of various techniques are also employed as nanoparticles can penetrate through inflamed regions and adhere to the thick mucus of the diseased site. Prodrug systems such as 5-ASAs formulations or their derivatives are effective in reducing colonic damage. Liposomes can be modified with both hydrophilic and lipophilic particles and act as lipid-based vesicular systems, while hybrid drug delivery systems containing an internal nanoparticle section for loading drugs are potential routes too. Leukosomes are also considered as possible carrier systems, and results from mouse models have revealed that they control anti- and pro-inflammatory molecules.
Collapse
Affiliation(s)
- Farah Yasmin
- Department of Medicine, Dow University of Health Science, Karachi 74200, Pakistan
| | - Hala Najeeb
- Department of Medicine, Dow University of Health Science, Karachi 74200, Pakistan
| | - Shehryar Shaikh
- Department of Medicine, Dow OJha University Hospital, Karachi 74200, Pakistan
| | - Muhammad Hasanain
- Department of Medicine, Dow University of Health Science, Karachi 74200, Pakistan
| | - Unaiza Naeem
- Department of Medicine, Dow University of Health Science, Karachi 74200, Pakistan
| | - Abdul Moeed
- Department of Medicine, Dow University of Health Science, Karachi 74200, Pakistan
| | - Thoyaja Koritala
- Department of Medicine, Mayo Clinic Health System, Mankato, MN 56001, United States
| | - Syedadeel Hasan
- Department of Medicine, University of Louisville, Louisville, KY 40292, United States
| | - Salim Surani
- Department of Medicine, Texas A&M University, College Station, TX 77843, United States
- Department of Anesthesiology, Mayo Clinic, Rochester, MN 55901, United States
| |
Collapse
|
33
|
Zhao Y, Xue P, Lin G, Tong M, Yang J, Zhang Y, Ran K, Zhuge D, Yao Q, Xu H. A KPV-binding double-network hydrogel restores gut mucosal barrier in an inflamed colon. Acta Biomater 2022; 143:233-252. [PMID: 35245681 DOI: 10.1016/j.actbio.2022.02.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/29/2022] [Accepted: 02/25/2022] [Indexed: 02/08/2023]
Abstract
Ulcerative colitis (UC) usually occurs in the superficial mucosa of the colorectum. Here, a double-network hydrogel (PMSP) was constructed from maleimided γ-polyglutamic acid and thiolated γ-polyglutamic acid through crosslinking of thiol-maleimide and self-oxidized thiols. PMSP with a negative charge specifically adhered to the inflamed mucosa with positively charged proteins rather than to the healthy mucosa. PMSP exhibited good mechanical strength with storage modulus (G') of 17.6 Pa and a linear viscoelastic region (LVR) of 107.2% strain. Moreover, PMSP showed a stronger bio-adhesive force toward the inflamed tissue-mimicking substrate than toward its healthy counterpart. In vivo imaging confirmed that PMSP specifically adhered to the inflamed colonic mucosa of rats with TNBS-induced UC. KPV (Lys-Pro-Val) as a model drug was easily captured by PMSP through electrostatic interactions, thus retaining its bioactivity for a longer time under high temperature conditions. Moreover, the alleviating effect of KPV on rats with TNBS-induced colitis was significantly improved by PMSP after intracolonic administration. The epithelial barrier of the colon also effectively recovered following PMSP-KPV treatment. PMSP-KPV also modulated the gut flora, markedly augmenting the abundance of beneficial microorganisms in gut homeostasis. The mechanism by which PMSP-KPV induces a therapeutic effect may be associated with the inhibition of oxidative stress. Conclusively, the PMSP hydrogel seems to be a promising rectal delivery system for the therapy of UC. STATEMENT OF SIGNIFICANCE: Ulcerative colitis (UC) is a chronic and relapsing disease of the gastrointestinal tract. A key therapeutic approach to treat UC is to repair the mucosal barriers. Here, a double-network hydrogel (PMSP) was constructed from maleimided and thiolated γ-polyglutamic acid through crosslinking of thiol-maleimide and self-oxidized thiols. The negatively charged PMSP specifically adhered to the inflamed colon rather than its healthy counterpart and was retained for a longer time. KPV as a model drug was easily captured by PMSP, which provided better stability to KPV when exposed to high temperature of 50 °C. The epithelial mucosal barrier of the colon was effectively recovered by the rectal administration of PMSP-KPV to rats with TNBS-induced UC. Moreover, PMSP-KPV modulated the gut flora of colitic rats, markedly augmenting the abundance of beneficial microorganisms. Conclusively, PMSP seems to be a promising rectal delivery system for UC therapy.
Collapse
|
34
|
Tang L, He S, Yin Y, Li J, Xiao Q, Wang R, Gao L, Wang W. Combining nanotechnology with the multifunctional roles of neutrophils against cancer and inflammatory disease. NANOSCALE 2022; 14:1621-1645. [PMID: 35079756 DOI: 10.1039/d1nr07725b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Neutrophils, the most abundant leukocytes in humans, play a crucial role in acute inflammation during infection and tumorigenesis. Neutrophils are the major types of cells recruited to the inflammation sites induced by pathogens, exhibiting great homing ability towards inflammatory disorders and tumor sites. Therefore, a neutrophil-based drug delivery system (NDDS) has become a promising platform for anti-cancer and anti-inflammatory treatment. Recent decades have witnessed the huge progress of applying nanomaterials in drug delivery. Nanomaterials are regarded as innovative components to enrich the field of neutrophil-based therapies due to their unique physiochemical characteristics. In this review, the latest advancement of combining diverse nanomaterials with an NDDS for cancer and inflammatory disease treatment will be summarized. It is discussed how nanomaterials empower the therapeutic area of an NDDS and how an NDDS circumvents the limitations of nanomaterials. Moreover, based on the finding that neutrophils are closely involved in the progression of cancer and inflammatory diseases, emerging therapeutic strategies that target neutrophils will be outlined. Finally, as neutrophils were demonstrated to play a central role in the immunopathology of COVID-19, which causes necroinflammation that is responsible for the cytokine storm and sepsis during coronavirus infections, novel therapeutic approaches that anchor neutrophils against the pathological consequences related to COVID-19 will be highlighted as well.
Collapse
Affiliation(s)
- Lu Tang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Shun He
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Yue Yin
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Jing Li
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Qiaqia Xiao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Ruotong Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Lijun Gao
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| | - Wei Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
- NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, Jiangsu, P.R. China.
| |
Collapse
|
35
|
Hartwig O, Shetab Boushehri MA, Shalaby KS, Loretz B, Lamprecht A, Lehr CM. Drug delivery to the inflamed intestinal mucosa - targeting technologies and human cell culture models for better therapies of IBD. Adv Drug Deliv Rev 2021; 175:113828. [PMID: 34157320 DOI: 10.1016/j.addr.2021.113828] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/19/2022]
Abstract
Current treatment strategies for inflammatory bowel disease (IBD) seek to alleviate the undesirable symptoms of the disorder. Despite the higher specificity of newer generation therapeutics, e.g. monoclonal antibodies, adverse effects still arise from their interference with non-specific systemic immune cascades. To circumvent such undesirable effects, both conventional and newer therapeutic options can benefit from various targeting strategies. Of course, both the development and the assessment of the efficiency of such targeted delivery systems necessitate the use of suitable in vivo and in vitro models representing relevant pathophysiological manifestations of the disorder. Accordingly, the current review seeks to provide a comprehensive discussion of the available preclinical models with emphasis on human in vitro models of IBD, along with their potentials and limitations. This is followed by an elaboration on the advancements in the field of biology- and nanotechnology-based targeted drug delivery systems and the potential rooms for improvement to facilitate their clinical translation.
Collapse
Affiliation(s)
- Olga Hartwig
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, D-66123 Saarbrücken, Germany
| | | | - Karim S Shalaby
- Department of Pharmaceutics, University of Bonn, D-53121 Bonn, Germany; Department of Pharmaceutics and Industrial Pharmacy, Ain Shams University, Cairo, Egypt
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany
| | - Alf Lamprecht
- Department of Pharmaceutics, University of Bonn, D-53121 Bonn, Germany.
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS) - Helmholtz Centre for Infection Research (HZI), D-66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, D-66123 Saarbrücken, Germany.
| |
Collapse
|
36
|
Cell membrane-biomimetic coating via click-mediated liposome fusion for mitigating the foreign-body reaction. Biomaterials 2021; 271:120768. [PMID: 33812321 DOI: 10.1016/j.biomaterials.2021.120768] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/02/2021] [Accepted: 03/15/2021] [Indexed: 12/19/2022]
Abstract
The foreign-body reaction (FBR) caused by the implantation of synthetic polymer scaffolds seriously affects tissue-biomaterial integration and tissue repair. To address this issue, we developed a cell membrane-biomimetic coating formed by "click"-mediated liposome immobilization and fusion on the surface of electrospun fibers to mitigate the FBR. Utilization of electrospun polystyrene microfibrous scaffold as a model matrix, we deposited azide-incorporated silk fibroin on the surface of the fibers by the layer-by-layer assembly, finally, covalently modified with clickable liposomes via copper-free SPAAC click reaction. Compared with physical adsorption, liposomes click covalently binding can quickly fuse to form lipid film and maintain fluidity, which also improved liposome stability in vitro and in vivo. Molecular dynamics simulation proved that "click" improves the binding rate and strength of liposome to silk substrate. Importantly, histological observation and in vivo fluorescent probes imaging showed that liposome-functionalized electrospun fibers had negligible characteristics of the FBR and were accompanied by many infiltrated host cells and new blood vessels. We believe that the promotion of macrophage polarization toward a pro-regenerative phenotype plays an important role in vascularization. This bioinspired strategy paves the way for utilizing cell membrane biomimetic coating to resist the FBR and promote tissue-scaffold integration.
Collapse
|
37
|
Oroojalian F, Beygi M, Baradaran B, Mokhtarzadeh A, Shahbazi MA. Immune Cell Membrane-Coated Biomimetic Nanoparticles for Targeted Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006484. [PMID: 33577127 DOI: 10.1002/smll.202006484] [Citation(s) in RCA: 264] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/20/2020] [Indexed: 06/12/2023]
Abstract
Nanotechnology has provided great opportunities for managing neoplastic conditions at various levels, from preventive and diagnostic to therapeutic fields. However, when it comes to clinical application, nanoparticles (NPs) have some limitations in terms of biological stability, poor targeting, and rapid clearance from the body. Therefore, biomimetic approaches, utilizing immune cell membranes, are proposed to solve these issues. For example, macrophage or neutrophil cell membrane coated NPs are developed with the ability to interact with tumor tissue to suppress cancer progression and metastasis. The functionality of these particles largely depends on the surface proteins of the immune cells and their preserved function during membrane extraction and coating process on the NPs. Proteins on the outer surface of immune cells can render a wide range of activities to the NPs, including prolonged blood circulation, remarkable competency in recognizing antigens for enhanced targeting, better cellular interactions, gradual drug release, and reduced toxicity in vivo. In this review, nano-based systems coated with immune cells-derived membranous layers, their detailed production process, and the applicability of these biomimetic systems in cancer treatment are discussed. In addition, future perspectives and challenges for their clinical translation are also presented.
Collapse
Affiliation(s)
- Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran
| | - Mohammad Beygi
- Department of Agricultural Engineering, Isfahan University of Technology (IUT), Isfahan, 84156-83111, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 51666-14731, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 51666-14731, Iran
| | - Mohammad-Ali Shahbazi
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Zanjan Pharmaceutical Nanotechnology Research Center (ZPNRC), Zanjan University of Medical Sciences, Zanjan, 45139-56184, Iran
| |
Collapse
|
38
|
Zhang S, Kang L, Hu S, Hu J, Fu Y, Hu Y, Yang X. Carboxymethyl chitosan microspheres loaded hyaluronic acid/gelatin hydrogels for controlled drug delivery and the treatment of inflammatory bowel disease. Int J Biol Macromol 2020; 167:1598-1612. [PMID: 33220374 DOI: 10.1016/j.ijbiomac.2020.11.117] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/31/2020] [Accepted: 11/16/2020] [Indexed: 12/12/2022]
Abstract
A major drawback of oral treatment of inflammatory bowel disease (IBD) is the non-specific distribution of drugs during long-term treatment. Despite its effectiveness as an anti-inflammatory drug, curcumin (CUR) is limited by its low bioavailability in IBD treatment. Herein, a pH-sensitive composite hyaluronic acid/gelatin (HA/GE) hydrogel drug delivery system containing carboxymethyl chitosan (CC) microspheres loaded with CUR was fabricated for IBD treatment. The composition and structure of the composite system were optimized and the physicochemical properties were characterized using infrared spectroscopy, X-ray diffraction, swelling, and release behavior studies. In vitro, the formulation exhibited good sustained release property and the drug release rate was 65% for 50 h. In vivo pharmacokinetic experiments indicated that high level of CUR was maintained in the colon tissue for more than 24 h; it also played an anti-inflammatory role by evaluating the histopathological changes through hematoxylin and eosin (H&E), myeloperoxidase (MPO), and immunofluorescent staining. Additionally, the formulation substantially inhibited the level of the main pro-inflammatory cytokines of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) secreted by macrophages, compared to the control group. The pharmacodynamic experiment showed that the formulation group of CUR@gels had the best therapeutic effect on colitis in mice. The composite gel delivery system has potential for the effective delivery of CUR in the treatment of colitis. This study also provides a reference for the design and preparation of a new oral drug delivery system with controlled release behavior.
Collapse
Affiliation(s)
- Shangwen Zhang
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central University for Nationalities, Wuhan 430074, China
| | - Li Kang
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central University for Nationalities, Wuhan 430074, China
| | - Sheng Hu
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central University for Nationalities, Wuhan 430074, China
| | - Jie Hu
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central University for Nationalities, Wuhan 430074, China
| | - Yanping Fu
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central University for Nationalities, Wuhan 430074, China
| | - Yan Hu
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central University for Nationalities, Wuhan 430074, China.
| | - Xinzhou Yang
- School of Pharmaceutical Science, South-Central University for Nationalities, Wuhan 430074, China; National Demonstration Center for Experimental Ethnopharmacology Education, South-Central University for Nationalities, Wuhan 430074, China.
| |
Collapse
|
39
|
Naeem M, Lee J, Oshi MA, Cao J, Hlaing SP, Im E, Jung Y, Yoo JW. Colitis-targeted hybrid nanoparticles-in-microparticles system for the treatment of ulcerative colitis. Acta Biomater 2020; 116:368-382. [PMID: 32937207 DOI: 10.1016/j.actbio.2020.09.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 12/26/2022]
Abstract
Nanoparticle (NP)-based drug delivery systems accumulate in the disrupted epithelium of inflamed colon tissue in ulcerative colitis. However, premature early drug release and uptake or degradation of NPs during their passage through the harsh gastric or intestinal environment compromise their therapeutic outcomes. This study aimed to develop an advanced colitis-targeted hybrid nanoparticles-in-microparticles (NPsinMPs) drug delivery system to overcome the aforementioned challenges. First, sustained drug releasing poly(lactic-co-glycolic acid) NPs were generated and further encapsulated in pH-sensitive Eudragit FS30D MPs to ensure complete drug protection in a gastric-like pH and for selective delivery of NPs to the colon. SEM and confocal microscopy for the NPsinMPs revealed successful NP encapsulation. NPsinMPs prevented drug release in an acidic gastric-like and intestinal-like pH and presented a sustained release thereafter at an ileal and colonic pH, indicating the degradation of the outer pH-sensitive MPs and release of NPs. Furthermore, in vivo imaging of gastrointestinal tract of a colitis mouse orally administered with fluorescent NPsinMPs revealed higher fluorescence intensities selectively in the colon, demonstrating the release of loaded NPs and their concomitant accumulation at the site of colon inflammation. NPsinMPs markedly mitigated experimental colitis in mice indicated by improved histopathological analysis, decreased myeloperoxidase activity, neutrophils and macrophage infiltration, and expression of proinflammatory cytokines in colonic tissues compared with NP-treated mice. The present results show the successful formulation of an NPsinMP-based drug delivery system and provide a platform to improve NP-based colon-targeted drug delivery through improved protection of encapsulated NPs and their payload in the early small intestine.
Collapse
Affiliation(s)
- Muhammad Naeem
- College of Pharmacy, Pusan National University, Busan609-735, South Korea; Department of Biological Sciences, National University of Medical Sciences, Rawalpindi, Punjab, Pakistan
| | - Juho Lee
- College of Pharmacy, Pusan National University, Busan609-735, South Korea
| | - Murtada A Oshi
- College of Pharmacy, Pusan National University, Busan609-735, South Korea
| | - Jiafu Cao
- College of Pharmacy, Pusan National University, Busan609-735, South Korea
| | - Shwe Phyu Hlaing
- College of Pharmacy, Pusan National University, Busan609-735, South Korea
| | - Eunok Im
- College of Pharmacy, Pusan National University, Busan609-735, South Korea
| | - Yunjin Jung
- College of Pharmacy, Pusan National University, Busan609-735, South Korea
| | - Jin-Wook Yoo
- College of Pharmacy, Pusan National University, Busan609-735, South Korea.
| |
Collapse
|
40
|
Xue P, Wang L, Xu J, Liu J, Pan X, Zhao Y, Xu H. Temperature-sensitive hydrogel for rectal perfusion improved the therapeutic effect of Kangfuxin liquid on DSS-induced ulcerative colitis mice: The inflammation alleviation and the colonic mucosal barriers repair. Int J Pharm 2020; 589:119846. [PMID: 32891717 DOI: 10.1016/j.ijpharm.2020.119846] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/19/2020] [Accepted: 08/31/2020] [Indexed: 01/05/2023]
Abstract
Kangfuxin liquid (KFX) is a Chinese medicine extracted from Periplaneta americana dried worms, which presented the bioactive functions of anti-inflammation and promoting the gastrointestinal mucosal barriers repair. But the low availability of KFX exposed to the distal colon affects its therapeutic effect on ulcerative colitis. Herein, an in situ hydrogel containing KFX was designed by using temperature-sensitive poloxamer 407 (P-407) as material for rectal administration. Three KFX-P formulations with different P407 concentrations (17%, 20% and 25%) were designed and screened by detecting the gelation time, gelation temperature and mechanical strength of hydrogel. P407 in these formulations was able to be completely dissolved in KFX at 4 ℃ and then was in situ gelled at 37 ℃ to form a semisolid hydrogel. Moreover, the gelation time, the gelation temperature and the mechanical strength of KFX-P hydrogel are highly dependent on P407 concentration. With P407 concentration increasing, both the gelation time and gelation temperature of KFX-P accordingly decreased and the gelation temperature range becomes narrowed; while the mechanical strength increased. KFX-P-20% displayed the moderate gelation temperature (28-30 ℃), the short gelation time (26 s) and the moderate mechanical strength (G' = 4.2 × 103 Pa), which was chosen for animal study. Thereafter, ulcerative colitis mice model (UC) was established by dextran sulfate sodium (DSS) and the therapeutic effect of KFX-P on UC was evaluated by inflammation symptoms relief, colon length, colonic MPO level and colonography. After rectal administration of KFX or KFX-P, the symptoms including diarrhea and hematochezia (DAI scores), weight loss and spleen swelling were significantly hindered. Meanwhile, the colonic MPO level in these groups was significantly decreased in comparison with PBS treatment. But the therapeutic effect of KFX-P was better than KFX. Besides, the morphology and mucosal barrier of colon were evaluated by HE staining, ZO-1 and claudin-5 staining. The mucosa epithelium layer, crypt, muscle layer mucosa and submucosa were also well repaired after KFX-P treatment. The strong fluorescence of ZO-1 and claudin-5 were uniformly distributed along the whole epithelial mucosa after KFX-P treatment, indicating the effective repairing of the colonic mucosal barrier. Collectively, the temperature-sensitive KFX-P for rectal delivery could effectively promote the repair of the colon mucosal barrier and inhibit the colonic inflammation in DSS-induced mice, which may be a potential strategy for UC treatment.
Collapse
Affiliation(s)
- Pengpeng Xue
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Lifen Wang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Jiawei Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Jiayi Liu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Xiaohong Pan
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Yingzheng Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Helin Xu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China.
| |
Collapse
|
41
|
Tang Q, Zhang W, Zhang C, Guan Y, Ding J, Yuan C, Tan C, Gao X, Tan S. Oxymatrine loaded nitric oxide-releasing liposomes for the treatment of ulcerative colitis. Int J Pharm 2020; 586:119617. [DOI: 10.1016/j.ijpharm.2020.119617] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/18/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023]
|
42
|
Liu L, Wei P, Yuan W, Liu Z, Xue F, Zhang X, Yi T. Detecting Basal Myeloperoxidase Activity in Living Systems with a Near-Infrared Emissive "Turn-On" Probe. Anal Chem 2020; 92:10971-10978. [PMID: 32674562 DOI: 10.1021/acs.analchem.9b04601] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Detecting myeloperoxidase (MPO) activity in living organisms is important because MPO contributes to the pathogenesis of many diseases such as rheumatoid arthritis and other inflammatory diseases, artherosclerosis, neurodegenerative disease, and some cancers. However, rapid and effective methods for the detection of basal MPO activity in living systems have not yet been reported. Herein, we report a near-infrared (NIR) emissive "turn-on" probe FD-301 that can specifically bind to MPO and accurately measure MPO activity in living cells and in vivo via a rapid response to initial hypochlorous acid (HOCl), produced by MPO. Notably, FD-301 could detect the basal level of MPO activity in human promyelocytic leukemia cells (HL-60) and could discriminate between MPO high-expression and low-expression cells. Furthermore, FD-301 was successfully applied to in vivo imaging of MPO in MPO-dependent diseases, such as arthritis and inflammatory bowel disease.
Collapse
Affiliation(s)
- Lingyan Liu
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200433, China
| | - Peng Wei
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Wei Yuan
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200433, China
| | - Zhongkuan Liu
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200433, China
| | - Fengfeng Xue
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200433, China
| | - Xinyu Zhang
- School of Public Health, Hongqiao International Institute of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Tao Yi
- Department of Chemistry, Fudan University, 2005 Songhu Road, Shanghai 200433, China.,College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| |
Collapse
|
43
|
Cheng S, Shen H, Zhao S, Zhang Y, Xu H, Wang L, Di B, Xu L, Hu C. Orally administered mesoporous silica capped with the cucurbit[8]uril complex to combat colitis and improve intestinal homeostasis by targeting the gut microbiota. NANOSCALE 2020; 12:15348-15363. [PMID: 32648873 DOI: 10.1039/d0nr03037f] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
RATIONALE Inflammatory bowel diseases (IBDs) are still awaiting innovative treatments that can maximize the efficiency of site-specific drug release in the colon while enhancing intestinal homeostasis. METHODS Herein, we present multilayer-coated mesoporous silica (MSs) which release payload drugs specifically in the colon tract in the presence of azoreductase produced by the gut microbiota, and simultaneously rejuvenate the tryptophan metabolism of the microbiome to induce activation of the aryl hydrocarbon receptor (AHR) for increased anti-inflammatory effects. The MSs were prepared by using cucurbit[8]uril (CB[8]) as a supramolecular "handcuff" to assemble chitosan/hyaluronic acid multilayers on the periphery of a mesoporous silica core. RESULTS Strikingly, although MSs remained fairly stable in both acidic and neutral pH, they exhibited excellent responsiveness towards dithionite, an azo-reducing agent employed as a substitute to mimic the specific azoreductase environment in vitro. In comparison with the drug in its free form, hydrocortisone-loaded MSs showed optimized accumulation of therapeutics in the colonic mucosa with minimized premature release in the upper gastrointestinal tract in in vivo imaging and biodistribution studies. The enhanced therapeutic effects of MSs were confirmed in dextran sodium sulfate-induced colitis in mice with promoted colonic epithelial barrier integrity, elevated level of AHR agonists and modulated production of inflammatory cytokines. Furthermore, 16S rRNA analysis showed that the disrupted gut homeostasis of colitic mice was partly corrected by MSs. CONCLUSION This novel drug delivery system using self-assembly of tryptophan-functionalized chitosan, which was precomplexed with CB[8], and azobenzene-functionalized hyaluronic acid on the surface of mesoporous silica nanoparticles provides a synergistic gut microbiota-targeting approach for IBD therapy.
Collapse
Affiliation(s)
- Shujie Cheng
- Department of Food Quality and Safety, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China and National R&D Center for Chinese Herbal Medicine Processing, China Pharmaceutical University, Nanjing 210009, PR China
| | - Haowen Shen
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, PR China. and Jiangsu Institute of Medical Device Testing, Nanjing 210019, PR China
| | - Sibo Zhao
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Yuanxin Zhang
- Department of Food Quality and Safety, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China and National R&D Center for Chinese Herbal Medicine Processing, China Pharmaceutical University, Nanjing 210009, PR China
| | - Hui Xu
- Department of Food Quality and Safety, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China and National R&D Center for Chinese Herbal Medicine Processing, China Pharmaceutical University, Nanjing 210009, PR China
| | - Lancheng Wang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, PR China.
| | - Bin Di
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, PR China.
| | - Lili Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, China Pharmaceutical University, Ministry of Education, Nanjing 210009, PR China.
| | - Chi Hu
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 211198, PR China.
| |
Collapse
|
44
|
Wang H, Liu Y, He R, Xu D, Zang J, Weeranoppanant N, Dong H, Li Y. Cell membrane biomimetic nanoparticles for inflammation and cancer targeting in drug delivery. Biomater Sci 2020; 8:552-568. [PMID: 31769765 DOI: 10.1039/c9bm01392j] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nanoparticle capture and elimination by the immune system are great obstacles for drug delivery. Camouflaging nanoparticles with cell membrane represents a promising strategy to communicate and negotiate with the immune system. As a novel class of nanotherapeutics, such biomimetic nanoparticles inherit specific biological functionalities of the source cells (e.g., erythrocytes, immune cells, cancer cells and platelets) in order to evade immune elimination, prolong circulation time, and even target a disease region by virtue of the homing tendency of the cell membrane protein. In this review, we begin with an overview of different cell membranes that can be utilized to create a biointerface on nanoparticles. Subsequently, we elaborate on the state-of-the-art of cell membrane biomimetic nanoparticles for drug delivery. In particular, a summary of data on circulation capacity and targeting efficiency by camouflaged nanoparticles is presented. In addition to cancer therapy, inflammation treatment, as an emerging application of biomimetic nanoparticles, is specifically included. The challenges and outlook of this technology are discussed.
Collapse
Affiliation(s)
- Huaiji Wang
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai 200092, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Zhang J, Zhao Y, Hou T, Zeng H, Kalambhe D, Wang B, Shen X, Huang Y. Macrophage-based nanotherapeutic strategies in ulcerative colitis. J Control Release 2020; 320:363-380. [DOI: 10.1016/j.jconrel.2020.01.047] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/11/2020] [Accepted: 01/26/2020] [Indexed: 12/21/2022]
|
46
|
Advanced liposome-loaded scaffolds for therapeutic and tissue engineering applications. Biomaterials 2020; 232:119706. [DOI: 10.1016/j.biomaterials.2019.119706] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 11/30/2019] [Accepted: 12/18/2019] [Indexed: 01/02/2023]
|
47
|
Yan H, Shao D, Lao Y, Li M, Hu H, Leong KW. Engineering Cell Membrane-Based Nanotherapeutics to Target Inflammation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900605. [PMID: 31406672 PMCID: PMC6685500 DOI: 10.1002/advs.201900605] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/28/2019] [Indexed: 05/10/2023]
Abstract
Inflammation is ubiquitous in the body, triggering desirable immune response to defend against dangerous signals or instigating undesirable damage to cells and tissues to cause disease. Nanomedicine holds exciting potential in modulating inflammation. In particular, cell membranes derived from cells involved in the inflammatory process may be used to coat nanotherapeutics for effective targeted delivery to inflammatory tissues. Herein, the recent progress of rationally engineering cell membrane-based nanotherapeutics for inflammation therapy is highlighted, and the challenges and opportunities presented in realizing the full potential of cell-membrane coating in targeting and manipulating the inflammatory microenvironment are discussed.
Collapse
Affiliation(s)
- Huize Yan
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Dan Shao
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Yeh‐Hsing Lao
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Mingqiang Li
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
- Guangdong Provincial Key Laboratory of Liver DiseaseThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouGuangdong510630China
| | - Hanze Hu
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
| | - Kam W. Leong
- Department of Biomedical EngineeringColumbia UniversityNew YorkNY10027USA
- Institutes of Life SciencesSchool of Biomedical Science and Engineering and National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou International CampusGuangzhouGuangdong510006China
- Department of System BiologyColumbia University Medical CenterNew YorkNY10032USA
| |
Collapse
|