1
|
Wu W, Huo F, Yin C. Classification of self-assembled fluorescent probes and their application in cancer diagnosis. Chem Commun (Camb) 2024. [PMID: 39659280 DOI: 10.1039/d4cc05494f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
The high sensitivity, high selectivity, real-time monitoring capability, non-destructiveness, and versatility of small molecule fluorescent probes make them indispensable and powerful tools in bioscience research. Self-assembling fluorescent probes are a novel type of material that spontaneously assemble fluorescent dyes with specific molecules into nanoscale structures. Compared with ordinary small molecule fluorescent probes, self-assembled fluorescent probes have higher stability, selectivity, sensitivity, and temporal stability in detection. In recent years, the incidence and mortality of cancer have increased year by year, which has brought great challenges to the safety of human life, and traditional diagnostic methods such as nuclear magnetic resonance, ultrasound diagnosis, and X-ray tomography are time-consuming and have low resolution. The boundary between normal tissue and cancer tissue cannot be accurately distinguished during surgical resection, resulting in the possibility of recurrence after surgery. Fluorescent probes can quickly and efficiently diagnose and label cancerous tumor cells, which is of great significance for cancer discovery and treatment. In this paper, we review the classification of self-assembled fluorescent probes (molecular self-assembled fluorescent probes, nanomaterial self-assembled fluorescent probes and biological macromolecule self-assembled fluorescent probes) that are used in identifying and imaging cancerous tumor tissues. Furthermore, we discuss the current problems faced by self-assembled fluorescent probes through the specific identification and monitoring of enzymes with abnormal contents, active substances and low pH in the tumor microenvironment, hoping to give readers more inspiration.
Collapse
Affiliation(s)
- Wenjiao Wu
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan 030006, P. R. China.
| | - Fangjun Huo
- Research Institute of Applied Chemistry, Shanxi University, Taiyuan, 030006, China.
| | - Caixia Yin
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University, Taiyuan 030006, P. R. China.
| |
Collapse
|
2
|
Li C, Wang S, Liao C, Li Y, Zhou Y, Wu H, Xiong W. An In Situ Sustained-Release Chitosan Hydrogel to Attenuate Renal Fibrosis by Retaining Klotho Expression. Biomater Res 2024; 28:0099. [PMID: 39450151 PMCID: PMC11499586 DOI: 10.34133/bmr.0099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/18/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Klotho (KLO) is an anti-fibrotic protein expressed in the kidneys and has been decreasing in the development of renal fibrosis (RF). However, restoring the decline in KLO levels remains a great challenge during RF treatment. Herein, an injectable KLO-loaded chitosan (CS) hydrogel (KLO-Gel) is designed to achieve localized and prolonged release of KLO in the RF treatment. KLO-Gel was prepared by cross-linking CS with β-glycerophosphate (β-GP), followed by rapid (within 3 min) thermosensitive gelation at 37 °C. Furthermore, KLO-Gel exhibited a slow and sustained release (over 14 d) of KLO both in PBS and in the kidneys of mice with unilateral ureter obstruction (UUO). A single local injection of KLO-Gel into the renal capsule of UUO mice was more effective at reducing RF (i.e., maintaining renal function and tissue structure, alleviating extracellular matrix accumulation, and inhibiting the TGF-β1/Smad2/3 signaling pathway) over a 14-d period than daily intraperitoneal injections of free KLO or captopril. Crucially, CS was found to induce endogenous KLO secretion, highlighting the added value of using CS in RF treatment. Overall, this study demonstrated that KLO-Gel enhanced the anti-fibrotic efficacy of KLO while minimizing its off-target toxicity, and its clinical potential awaits further validation.
Collapse
Affiliation(s)
- Chenyang Li
- School of Pharmacy,
Shenzhen UniversityMedical School, Shenzhen University, Shenzhen 518055, China
| | - Shuai Wang
- School of Pharmacy,
Shenzhen UniversityMedical School, Shenzhen University, Shenzhen 518055, China
| | - Chenghui Liao
- School of Pharmacy,
Shenzhen UniversityMedical School, Shenzhen University, Shenzhen 518055, China
| | - Ying Li
- School of Pharmacy,
Shenzhen UniversityMedical School, Shenzhen University, Shenzhen 518055, China
| | - Yunfeng Zhou
- School of Basic Medical Sciences,
Shenzhen UniversityMedical School, Shenzhen University, Shenzhen 518055, China
| | - Haiqiang Wu
- School of Pharmacy,
Shenzhen UniversityMedical School, Shenzhen University, Shenzhen 518055, China
| | - Wei Xiong
- School of Pharmacy,
Shenzhen UniversityMedical School, Shenzhen University, Shenzhen 518055, China
| |
Collapse
|
3
|
Ye Y, Li M, Chen W, Wang H, He X, Liu N, Guo Z, Zheng C. Natural polysaccharides as promising reno-protective agents for the treatment of various kidney injury. Pharmacol Res 2024; 207:107301. [PMID: 39009291 DOI: 10.1016/j.phrs.2024.107301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/13/2024] [Accepted: 07/07/2024] [Indexed: 07/17/2024]
Abstract
Renal injury, a prevalent clinical outcome with multifactorial etiology, imposes a substantial burden on society. Currently, there remains a lack of effective management and treatments. Extensive research has emphasized the diverse biological effects of natural polysaccharides, which exhibit promising potential for mitigating renal damage. This review commences with the pathogenesis of four common renal diseases and the shared mechanisms underlying renal injury. The renoprotective roles of polysaccharides in vivo and in vitro are summarized in the following five aspects: anti-oxidative stress effects, anti-apoptotic effects, anti-inflammatory effects, anti-fibrotic effects, and gut modulatory effects. Furthermore, we explore the structure-activity relationship and bioavailability of polysaccharides in relation to renal injury, as well as investigate their utility as biomaterials for alleviating renal injury. The clinical experiments of polysaccharides applied to patients with chronic kidney disease are also reviewed. Broadly, this review provides a comprehensive perspective on the research direction of natural polysaccharides in the context of renal injury, with the primary aim to serve as a reference for the clinical development of polysaccharides as pharmaceuticals and prebiotics for the treatment of kidney diseases.
Collapse
Affiliation(s)
- Yufei Ye
- Department of Chinese Medicine Authentication, Faculty of Pharmacy, Second Military Medical University/Naval Medical University, 325 Guohe Road, Shanghai 200433, China; Department of Nephrology, Changhai Hospital, Second Military Medical University/Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Maoting Li
- Department of Chinese Medicine Authentication, Faculty of Pharmacy, Second Military Medical University/Naval Medical University, 325 Guohe Road, Shanghai 200433, China; Department of Nephrology, Naval Medical Center of PLA, Second Military Medical University/Naval Medical University, 338 West Huaihai Road, Shanghai 200052, China
| | - Wei Chen
- Department of Nephrology, Changhai Hospital, Second Military Medical University/Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Hongrui Wang
- Department of Chinese Medicine Authentication, Faculty of Pharmacy, Second Military Medical University/Naval Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Xuhui He
- Department of Chinese Medicine Authentication, Faculty of Pharmacy, Second Military Medical University/Naval Medical University, 325 Guohe Road, Shanghai 200433, China
| | - Nanmei Liu
- Department of Nephrology, Naval Medical Center of PLA, Second Military Medical University/Naval Medical University, 338 West Huaihai Road, Shanghai 200052, China.
| | - Zhiyong Guo
- Department of Nephrology, Changhai Hospital, Second Military Medical University/Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| | - Chengjian Zheng
- Department of Chinese Medicine Authentication, Faculty of Pharmacy, Second Military Medical University/Naval Medical University, 325 Guohe Road, Shanghai 200433, China.
| |
Collapse
|
4
|
Madbouly N, Ooda A, Nabil A, Nasser A, Ahmed E, Ali F, Mohamed F, Faried H, Badran M, Ahmed M, Ibrahim M, Rasmy M, Saleeb M, Riad V, Ibrahim Y, Farid A. The renoprotective activity of amikacin-gamma-amino butyric acid-chitosan nanoparticles: a comparative study. Inflammopharmacology 2024; 32:2629-2645. [PMID: 38662181 PMCID: PMC11300498 DOI: 10.1007/s10787-024-01464-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 03/17/2024] [Indexed: 04/26/2024]
Abstract
The development of nanoparticles (NPs) with active components with upgraded stability, and prolonged release helps in enhanced tissue regeneration. In addition, NPs are feasible strategy to boost antibiotic effectiveness and reduce drug side effects. Our study focuses on the use of amikacin (AMK) and gamma amino butyric acid (GABA) unloaded combinations or loaded on chitosan nanoparticles (CSNPs) for kidney protection. The AMK-GABA-CSNPs were prepared with the ionic gelation method, the morphology was studied using transmission electron microscopy (TEM), zetasizer and the Fourier transform-infrared spectroscopy (FT-IR) spectrum of the synthesized NPs was observed. The average size of AMK-GABA-CSNPs was 77.5 ± 16.5 nm. Zeta potential was + 38.94 ± 2.65 mV. AMK-GABA-CSNPs revealed significant in vitro antioxidant, anti-coagulation, non-hemolytic properties and good cell compatibility. To compare the effects of the unloaded AMK-GABA combination and AMK-GABA-CSNPs on the renal tissue, 42 healthy Sprague-Dawley rats were divided into seven groups. G1: normal control (NC), normal saline; G2: low-dose nephrotoxic group (LDN), AMK (20 mg/kg/day; i.p.); G3: unloaded AMK (20 mg/kg/day; i.p.) and GABA (50 mg/kg/day; i.p.); G4: AMK-GABA-CSNPs (20 mg/kg/day; i.p.); G5: high-dose nephrotoxic group (HDN), AMK (30 mg/kg/day; i.p.); G6: unloaded AMK (30 mg/kg/day; i.p.) and GABA (50 mg/kg/day; i.p.) and G7: AMK-GABA-CSNPs (30 mg/kg/day; i.p.). The results showed that AMK-GABA-CSNPs formulation is superior to unloaded AMK-GABA combination as it ameliorated kidney functions, oxidative stress and displayed a significant homeostatic role via suppression of inflammatory cytokines of Th1, Th2 and Th17 types. Hence, AMK-GABA-CSNPs could afford a potential nano-based therapeutic formula for the management of AMK-nephrotoxicity.
Collapse
Affiliation(s)
- Neveen Madbouly
- Zoology Department, Faculty of Science, Cairo University, Giza, Egypt.
| | - Adham Ooda
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Ahmed Nabil
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Areej Nasser
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Esraa Ahmed
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Fatma Ali
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Fatma Mohamed
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Habiba Faried
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Mai Badran
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Mariam Ahmed
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Mariam Ibrahim
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Mariam Rasmy
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Martina Saleeb
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Vereena Riad
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Yousr Ibrahim
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, Giza, Egypt
| | - Alyaa Farid
- Biotechnology Department, Faculty of Science, Cairo University, Giza, Egypt
| |
Collapse
|
5
|
Shang S, Li X, Wang H, Zhou Y, Pang K, Li P, Liu X, Zhang M, Li W, Li Q, Chen X. Targeted therapy of kidney disease with nanoparticle drug delivery materials. Bioact Mater 2024; 37:206-221. [PMID: 38560369 PMCID: PMC10979125 DOI: 10.1016/j.bioactmat.2024.03.014] [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: 01/11/2024] [Revised: 03/09/2024] [Accepted: 03/10/2024] [Indexed: 04/04/2024] Open
Abstract
With the development of nanomedicine, nanomaterials have been widely used, offering specific drug delivery to target sites, minimal side effects, and significant therapeutic effects. The kidneys have filtration and reabsorption functions, with various potential target cell types and a complex structural environment, making the strategies for kidney function protection and recovery after injury complex. This also lays the foundation for the application of nanomedicine in kidney diseases. Currently, evidence in preclinical and clinical settings supports the feasibility of targeted therapy for kidney diseases using drug delivery based on nanomaterials. The prerequisite for nanomedicine in treating kidney diseases is the use of carriers with good biocompatibility, including nanoparticles, hydrogels, liposomes, micelles, dendrimer polymers, adenoviruses, lysozymes, and elastin-like polypeptides. These carriers have precise renal uptake, longer half-life, and targeted organ distribution, protecting and improving the efficacy of the drugs they carry. Additionally, attention should also be paid to the toxicity and solubility of the carriers. While the carriers mentioned above have been used in preclinical studies for targeted therapy of kidney diseases both in vivo and in vitro, extensive clinical trials are still needed to ensure the short-term and long-term effects of nano drugs in the human body. This review will discuss the advantages and limitations of nanoscale drug carrier materials in treating kidney diseases, provide a more comprehensive catalog of nanocarrier materials, and offer prospects for their drug-loading efficacy and clinical applications.
Collapse
Affiliation(s)
- Shunlai Shang
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Xiangmeng Li
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
- Key Laboratory of Bone Metabolism and Physiology in Chronic Kidney Disease of Hebei Province, China
- Peking Union Medical College, Beijing, China
| | - Haoran Wang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Yena Zhou
- School of Medicine, Nankai University, Tianjin, China
| | - Keying Pang
- College of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
| | - Ping Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Xiaomin Liu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Min Zhang
- Department of Nephrology, Affiliated Beijing Chaoyang Hospital of Capital Medical University, Beijing, China
| | - Wenge Li
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Qinggang Li
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing, China
| |
Collapse
|
6
|
Hou Y, Zhu L, Ye X, Ke Q, Zhang Q, Xie X, Piao JG, Wei Y. Integrated oral microgel system ameliorates renal fibrosis by hitchhiking co-delivery and targeted gut flora modulation. J Nanobiotechnology 2024; 22:305. [PMID: 38822364 PMCID: PMC11143587 DOI: 10.1186/s12951-024-02586-2] [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: 01/10/2024] [Accepted: 05/26/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Renal fibrosis is a progressive process associated with chronic kidney disease (CKD), contributing to impaired kidney function. Active constituents in traditional Chinese herbs, such as emodin (EMO) and asiatic acid (AA), exhibit potent anti-fibrotic properties. However, the oral administration of EMO and AA results in low bioavailability and limited kidney accumulation. Additionally, while oral probiotics have been accepted for CKD treatment through gut microbiota modulation, a significant challenge lies in ensuring their viability upon administration. Therefore, our study aims to address both renal fibrosis and gut microbiota imbalance through innovative co-delivery strategies. RESULTS In this study, we developed yeast cell wall particles (YCWPs) encapsulating EMO and AA self-assembled nanoparticles (NPYs) and embedded them, along with Lactobacillus casei Zhang, in chitosan/sodium alginate (CS/SA) microgels. The developed microgels showed significant controlled release properties for the loaded NPYs and prolonged the retention time of Lactobacillus casei Zhang (L. casei Zhang) in the intestine. Furthermore, in vivo biodistribution showed that the microgel-carried NPYs significantly accumulated in the obstructed kidneys of rats, thereby substantially increasing the accumulation of EMO and AA in the impaired kidneys. More importantly, through hitchhiking delivery based on yeast cell wall and positive modulation of gut microbiota, our microgels with this synergistic strategy of therapeutic and modulatory interactions could regulate the TGF-β/Smad signaling pathway and thus effectively ameliorate renal fibrosis in unilateral ureteral obstruction (UUO) rats. CONCLUSION In conclusion, our work provides a new strategy for the treatment of renal fibrosis based on hitchhiking co-delivery of nanodrugs and probiotics to achieve synergistic effects of disease treatment and targeted gut flora modulation.
Collapse
Affiliation(s)
- Yu Hou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Lin Zhu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xiaofeng Ye
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qiaoying Ke
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Qibin Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Xiaowei Xie
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China
| | - Ji-Gang Piao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| | - Yinghui Wei
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311402, China.
| |
Collapse
|
7
|
Cheng HT, Ngoc Ta YN, Hsia T, Chen Y. A quantitative review of nanotechnology-based therapeutics for kidney diseases. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1953. [PMID: 38500369 DOI: 10.1002/wnan.1953] [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: 01/12/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/20/2024]
Abstract
Kidney-specific nanocarriers offer a targeted approach to enhance therapeutic efficacy and reduce off-target effects in renal treatments. The nanocarriers can achieve organ or cell specificity via passive targeting and active targeting mechanisms. Passive targeting capitalizes on the unique physiological traits of the kidney, with factors like particle size, charge, shape, and material properties enhancing organ specificity. Active targeting, on the other hand, achieves renal specificity through ligand-receptor interactions, modifying nanocarriers with molecules, peptides, or antibodies for receptor-mediated delivery. Nanotechnology-enabled therapy targets diseased kidney tissue by modulating podocytes and immune cells to reduce inflammation and enhance tissue repair, or by inhibiting myofibroblast differentiation to mitigate renal fibrosis. This review summarizes the current reports of the drug delivery systems that have been tested in vivo, identifies the nanocarriers that may preferentially accumulate in the kidney, and quantitatively compares the efficacy of various cargo-carrier combinations to outline optimal strategies and future research directions. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Hui-Teng Cheng
- Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Zhu Bei City, Taiwan
| | - Yen-Nhi Ngoc Ta
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
- International Intercollegiate Ph.D. Program, National Tsing Hua University, Hsinchu, Taiwan
| | - Tiffaney Hsia
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
8
|
Saha P, Ahmad F. Neuroprotective, Anti-Inflammatory and Antifibrillogenic Offerings by Emodin against Alzheimer's Dementia: A Systematic Review. ACS OMEGA 2024; 9:7296-7309. [PMID: 38405501 PMCID: PMC10882671 DOI: 10.1021/acsomega.3c07178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/07/2023] [Accepted: 12/11/2023] [Indexed: 02/27/2024]
Abstract
Background: Alzheimer's disease (AD) is among the major causes of dementia in the elderly and exerts tremendous clinical, psychological and socio-economic constraints. Currently, there are no effective disease-modifying/retarding anti-AD agents. Emodin is a bioactive phytochemical with potent multimodal anti-inflammatory, antioxidant, and antifibrillogenic properties. In particular, emodin may result in significant repression of the pathogenic mechanisms underlying AD. The purpose of this review is to accumulate and summarize all the primary research data evaluating the therapeutic actions of emodin in AD pathogenesis. Methodology: The search, selection, and retrieval of pertinent primary research articles were systematically performed using a methodically designed approach. A variety of keyword combinations were employed on online scholarly web-databases. Strict preset inclusion and exclusion criteria were used to select the retrieved studies. Data from the individual studies were summarized and compiled into different sections, based upon their findings. Results: Cellular and animal research indicates that emodin exerts robust multimodal neuroprotection in AD. While emodin effectively prevents tau and amyloid-beta (Aβ) oligomerization, it also mitigates their neurotoxicity by attenuating neuroinflammatory, oxidative, and bioenergetic defects. Evidences for emodin-mediated enhancements in memory, learning, and cognition were also found in the literature. Conclusion: Emodin is a potential anti-AD dietary supplement; however, further studies are warrantied to thoroughly understand its target players and mechanisms. Moreover, human clinical data on emodin-mediated amelioration of AD phenotype is largely lacking, and must be addressed in the future. Lastly, the safety of exogenously supplemented emodin must be thoroughly evaluated.
Collapse
Affiliation(s)
- Priyanka Saha
- Department of Biotechnology, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology, Vellore 632014, India
| | - Faraz Ahmad
- Department of Biotechnology, School of Bio Sciences and Technology (SBST), Vellore Institute of Technology, Vellore 632014, India
| |
Collapse
|
9
|
Fang H, Xu S, Wang Y, Yang H, Su D. Endogenous stimuli-responsive drug delivery nanoplatforms for kidney disease therapy. Colloids Surf B Biointerfaces 2023; 232:113598. [PMID: 37866237 DOI: 10.1016/j.colsurfb.2023.113598] [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: 10/10/2023] [Accepted: 10/14/2023] [Indexed: 10/24/2023]
Abstract
Kidney disease is one of the most life-threatening health problems, affecting millions of people in the world. Commonly used steroids and immunosuppressants often fall exceptionally short of outcomes with inescapable systemic toxicity. With the booming research in nanobiotechnology, stimuli-responsive nanoplatform has come an appealing therapeutic strategy for kidney disease. Endogenous stimuli-responsive materials have shown profuse promise owing to their enhanced spatiotemporal control and precise to the location of the lesion. This review focuses on recent advances stimuli-responsive drug delivery nano-architectonics for kidney disease. First, a brief introduction of pathogenesis of kidney disease and pathological microenvironment were provided. Then, various endogenous stimulus involved in drug delivery nanoplatforms including pH, ROS, enzymes, and glucose were categorized based on the pathological mechanisms of kidney disease. Next, we separately summarized literature examples of endogenous stimuli-responsive nanomaterials, and outlined the design strategies and response mechanisms. Finally, the paper was concluded by discussing remaining challenges and future perspectives of endogenous stimuli-responsive drug delivery nanoplatform for expediting the speed of development and clinical applications.
Collapse
Affiliation(s)
- Hufeng Fang
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China.
| | - Shan Xu
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China
| | - Yu Wang
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China
| | - Hao Yang
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China
| | - Dan Su
- Department of Pharmacy, the Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou 213100, China.
| |
Collapse
|
10
|
Gao H, Sun L, Li J, Zhou Q, Xu H, Ma X, Li R, Yu B, Tian J. Illumination of Hydroxyl Radical in Kidney Injury and High-Throughput Screening of Natural Protectants Using a Fluorescent/Photoacoustic Probe. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303926. [PMID: 37870188 PMCID: PMC10667829 DOI: 10.1002/advs.202303926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/04/2023] [Indexed: 10/24/2023]
Abstract
The hydroxyl radical (•OH) is shown to play a crucial role in the occurrence and progression of acute kidney injury (AKI). Therefore, the development of a robust •OH probe holds great promise for the early diagnosis of AKI, high-throughput screening (HTS) of natural protectants, and elucidating the molecular mechanism of intervention in AKI. Herein, the design and synthesis of an activatable fluorescent/photoacoustic (PA) probe (CDIA) for sensitive and selective imaging of •OH in AKI is reported. CDIA has near-infrared fluorescence/PA channels and fast activation kinetics, enabling the detection of the onset of •OH in an AKI model. The positive detection time of 12 h using this probe is superior to the 48-hour detection time for typical clinical assays, such as blood urea nitrogen and serum creatinine detection. Furthermore, a method is established using CDIA for HTS of natural •OH inhibitors from herbal medicines. Puerarin is screened out by activating the Sirt1/Nrf2/Keap1 signaling pathway to protect renal cells in AKI. Overall, this work provides a versatile and dual-mode tool for illuminating the •OH-related pathological process in AKI and screening additional compounds to prevent and treat AKI.
Collapse
Affiliation(s)
- Han Gao
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of TCM Evaluation and Translational ResearchCellular and Molecular Biology CenterSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing211198P. R. China
| | - Lei Sun
- Jiangsu Co‐innovation Center of Efficient Processing and Utilization of Forest Resources, Key Laboratory of Forestry Genetics & Biotechnology of Ministry of Education, Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agroforest BiomassCollege of Chemical EngineeringNanjing Forestry UniversityNanjing210037P. R. China
| | - Jiwei Li
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of TCM Evaluation and Translational ResearchCellular and Molecular Biology CenterSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing211198P. R. China
| | - Qilin Zhou
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of TCM Evaluation and Translational ResearchCellular and Molecular Biology CenterSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing211198P. R. China
| | - Haijun Xu
- Jiangsu Co‐innovation Center of Efficient Processing and Utilization of Forest Resources, Key Laboratory of Forestry Genetics & Biotechnology of Ministry of Education, Jiangsu Provincial Key Lab for the Chemistry and Utilization of Agroforest BiomassCollege of Chemical EngineeringNanjing Forestry UniversityNanjing210037P. R. China
- School of Chemistry and Chemical EngineeringHenan Normal UniversityXinxiang453002P. R. China
| | - Xiao‐Nan Ma
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of TCM Evaluation and Translational ResearchCellular and Molecular Biology CenterSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing211198P. R. China
| | - Renshi Li
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of TCM Evaluation and Translational ResearchCellular and Molecular Biology CenterSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing211198P. R. China
| | - Bo‐Yang Yu
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of TCM Evaluation and Translational ResearchCellular and Molecular Biology CenterSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing211198P. R. China
| | - Jiangwei Tian
- State Key Laboratory of Natural MedicinesJiangsu Key Laboratory of TCM Evaluation and Translational ResearchCellular and Molecular Biology CenterSchool of Traditional Chinese PharmacyChina Pharmaceutical UniversityNanjing211198P. R. China
| |
Collapse
|
11
|
Fu L, Zhang Y, Farokhzad RA, Mendes BB, Conde J, Shi J. 'Passive' nanoparticles for organ-selective systemic delivery: design, mechanism and perspective. Chem Soc Rev 2023; 52:7579-7601. [PMID: 37817741 PMCID: PMC10623545 DOI: 10.1039/d2cs00998f] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2023]
Abstract
Nanotechnology has shown tremendous success in the drug delivery field for more effective and safer therapy, and has recently enabled the clinical approval of RNA medicine, a new class of therapeutics. Various nanoparticle strategies have been developed to improve the systemic delivery of therapeutics, among which surface modification of targeting ligands on nanoparticles has been widely explored for 'active' delivery to a specific organ or diseased tissue. Meanwhile, compelling evidence has recently been reported that organ-selective targeting may also be achievable by systemic administration of nanoparticles without surface ligand modification. In this Review, we highlight this unique set of 'passive' nanoparticles and their compositions and mechanisms for organ-selective delivery. In particular, the lipid-based, polymer-based, and biomimetic nanoparticles with tropism to different specific organs after intravenous administration are summarized. The underlying mechanisms (e.g., protein corona and size effect) of these nanosystems for organ selectivity are also extensively discussed. We further provide perspectives on the opportunities and challenges in this exciting area of organ-selective systemic nanoparticle delivery.
Collapse
Affiliation(s)
- Liyi Fu
- Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, 230031, China
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Yang Zhang
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Ryan A Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Bárbara B Mendes
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - João Conde
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas, NMS|FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|
12
|
Li H, Dai W, Xiao L, Sun L, He L. Biopolymer-Based Nanosystems: Potential Novel Carriers for Kidney Drug Delivery. Pharmaceutics 2023; 15:2150. [PMID: 37631364 PMCID: PMC10459991 DOI: 10.3390/pharmaceutics15082150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Kidney disease has become a serious public health problem throughout the world, and its treatment and management constitute a huge global economic burden. Currently, the main clinical treatments are not sufficient to cure kidney diseases. During its development, nanotechnology has shown unprecedented potential for application to kidney diseases. However, nanotechnology has disadvantages such as high cost and poor bioavailability. In contrast, biopolymers are not only widely available but also highly bioavailable. Therefore, biopolymer-based nanosystems offer new promising solutions for the treatment of kidney diseases. This paper reviews the biopolymer-based nanosystems that have been used for renal diseases and describes strategies for the specific, targeted delivery of drugs to the kidney as well as the physicochemical properties of the nanoparticles that affect the targeting success.
Collapse
Affiliation(s)
| | | | | | | | - Liyu He
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha 410011, China; (H.L.)
| |
Collapse
|
13
|
Zhang N, Ru B, Hu J, Xu L, Wan Q, Liu W, Cai W, Zhu T, Ji Z, Guo R, Zhang L, Li S, Tong X. Recent advances of CREKA peptide-based nanoplatforms in biomedical applications. J Nanobiotechnology 2023; 21:77. [PMID: 36869341 PMCID: PMC9985238 DOI: 10.1186/s12951-023-01827-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 02/22/2023] [Indexed: 03/05/2023] Open
Abstract
Nanomedicine technology is a rapidly developing field of research and application that uses nanoparticles as a platform to facilitate the diagnosis and treatment of diseases. Nanoparticles loaded with drugs and imaging contrast agents have already been used in clinically, but they are essentially passive delivery carriers. To make nanoparticles smarter, an important function is the ability to actively locate target tissues. It enables nanoparticles to accumulate in target tissues at higher concentrations, thereby improving therapeutic efficacy and reducing side effects. Among the different ligands, the CREKA peptide (Cys-Arg-Glu-Lys-Ala) is a desirable targeting ligand and has a good targeting ability for overexpressed fibrin in different models, such as cancers, myocardial ischemia-reperfusion, and atherosclerosis. In this review, the characteristic of the CREKA peptide and the latest reports regarding the application of CREKA-based nanoplatforms in different biological tissues are described. In addition, the existing problems and future application perspectives of CREKA-based nanoplatforms are also addressed.
Collapse
Affiliation(s)
- Nannan Zhang
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Bin Ru
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Jiaqi Hu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Langhai Xu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Quan Wan
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Wenlong Liu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - WenJun Cai
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Tingli Zhu
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Zhongwei Ji
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Ran Guo
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Lin Zhang
- College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| | - Shun Li
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
- Center for Rehabilitation Medicine, Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| | - Xiangmin Tong
- Laboratory Medicine Center, Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
14
|
A comprehensive review of emodin in fibrosis treatment. Fitoterapia 2023; 165:105358. [PMID: 36436587 DOI: 10.1016/j.fitote.2022.105358] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/27/2022]
Abstract
Emodin is the main pharmacodynamic components of rhubarb, with significant pharmacological effects and clinical efficacy.Emodin has a variety of therapy effects, such as anti-cancer, anti-fibrosis effects, and is widely used to treat encephalitis, diabetic cataract and organ fibrosis. Several studies have shown that emodin has a good treatment effect on organ fibrosis, but the mechanism is complex. Moreover, the evidence of some studies is conflicting and confusing. This paper reviewed the mechanism, pharmacokinetics and toxicology of emodin in fibrosis treatment, and briefly discussed relevant cutting-edge new formulations to improve the efficacy, the result can provide some reference for future study.
Collapse
|
15
|
Advanced Drug Delivery Systems for Renal Disorders. Gels 2023; 9:gels9020115. [PMID: 36826285 PMCID: PMC9956928 DOI: 10.3390/gels9020115] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 02/04/2023] Open
Abstract
Kidney disease management and treatment are currently causing a substantial global burden. The kidneys are the most important organs in the human urinary system, selectively filtering blood and metabolic waste into urine via the renal glomerulus. Based on charge and/or molecule size, the glomerular filtration apparatus acts as a barrier to therapeutic substances. Therefore, drug distribution to the kidneys is challenging, resulting in therapy failure in a variety of renal illnesses. Hence, different approaches to improve drug delivery across the glomerulus filtration barrier are being investigated. Nanotechnology in medicine has the potential to have a significant impact on human health, from illness prevention to diagnosis and treatment. Nanomaterials with various physicochemical properties, including size, charge, surface and shape, with unique biological attributes, such as low cytotoxicity, high cellular internalization and controllable biodistribution and pharmacokinetics, have demonstrated promising potential in renal therapy. Different types of nanosystems have been employed to deliver drugs to the kidneys. This review highlights the features of the nanomaterials, including the nanoparticles and corresponding hydrogels, in overcoming various barriers of drug delivery to the kidneys. The most common delivery sites and strategies of kidney-targeted drug delivery systems are also discussed.
Collapse
|
16
|
Gueta O, Amiram M. Expanding the chemical repertoire of protein-based polymers for drug-delivery applications. Adv Drug Deliv Rev 2022; 190:114460. [PMID: 36030987 DOI: 10.1016/j.addr.2022.114460] [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: 05/06/2022] [Accepted: 07/12/2022] [Indexed: 01/24/2023]
Abstract
Expanding the chemical repertoire of natural and artificial protein-based polymers (PBPs) can enable the production of sequence-defined, yet chemically diverse, biopolymers with customized or new properties that cannot be accessed in PBPs composed of only natural amino acids. Various approaches can enable the expansion of the chemical repertoire of PBPs, including chemical and enzymatic treatments or the incorporation of unnatural amino acids. These techniques are employed to install a wide variety of chemical groups-such as bio-orthogonally reactive, cross-linkable, post-translation modifications, and environmentally responsive groups-which, in turn, can facilitate the design of customized PBP-based drug-delivery systems with modified, fine-tuned, or entirely new properties and functions. Here, we detail the existing and emerging technologies for expanding the chemical repertoire of PBPs and review several chemical groups that either demonstrate or are anticipated to show potential in the design of PBP-based drug delivery systems. Finally, we provide our perspective on the remaining challenges and future directions in this field.
Collapse
Affiliation(s)
- Osher Gueta
- The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel
| | - Miriam Amiram
- The Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, P.O. Box 653, Beer-Sheva 8410501, Israel.
| |
Collapse
|
17
|
Li Z, Xu X, Wang Y, Kong L, Han C. Carrier-free nanoplatforms from natural plants for enhanced bioactivity. J Adv Res 2022:S2090-1232(22)00215-6. [PMID: 36208834 PMCID: PMC10403678 DOI: 10.1016/j.jare.2022.09.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/15/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Natural plants as well as traditional Chinese medicine have made outstanding contributions to the health and reproduction of human beings and remain the basis and major resource for drug innovation. Carrier-free nanoplatforms completely self-assembled by pure molecules or therapeutic components have attracted increasing attention due to their advantages of improved pharmacodynamics/pharmacokinetics, reduced toxicity, and high drug loading. In recent years, carrier-free nanoplatforms produced by self-assembly from natural plants have contributed to progress in a variety of therapeutic modalities. Notably, these nanoplatforms based on the interactions of components from different natural plants improve efficiency and depress toxicity. AIM OF REVIEW In this review, different types of self-assembled nanoplatforms are first summarized, mainly including nanoassemblies of pure small molecules isolated from different plants, extracellular vesicles separated from fresh plants, charcoal nanocomponents obtained from charred plants, and nanoaggregates from plants formulae decoctions. Key Scientific Concepts of Review: We mainly focus on composition, self-assembly mechanisms, biological activity and modes of action. Finally, a future perspective of existing challenges with respect to the clinical application of plant-based carrier-free nanoplatforms is discussed, which may be instructive to further develop effective carrier-free nanoplatforms from natural plants in the future.
Collapse
Affiliation(s)
- Zhongrui Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China; Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, 101 longmian Avenue, Nanjing 211166, PR China
| | - Xiao Xu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Yun Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China.
| | - Chao Han
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Bioactive Natural Product Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China.
| |
Collapse
|
18
|
Chade AR, Bidwell GL. Novel Drug Delivery Technologies and Targets for Renal Disease. Hypertension 2022; 79:1937-1948. [PMID: 35652363 PMCID: PMC9378601 DOI: 10.1161/hypertensionaha.122.17944] [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] [Indexed: 11/16/2022]
Abstract
The burden of acute and chronic kidney diseases to the health care system is exacerbated by the high mortality that this disease carries paired with the still limited availability of comprehensive therapies. A reason partially resides in the complexity of the kidney, with multiple potential target cell types and a complex structural environment that complicate strategies to protect and recover renal function after injury. Management of both acute and chronic renal disease, irrespective of the cause, are mainly focused on supportive treatments and renal replacement strategies when needed. Emerging preclinical evidence supports the feasibility of drug delivery technology for the kidney, and recent studies have contributed to building a robust catalog of peptides, proteins, nanoparticles, liposomes, extracellular vesicles, and other carriers that may be fused to therapeutic peptides, proteins, nucleic acids, or small molecule drugs. These fusions can display a precise renal uptake, an enhanced circulating time, and a directed intraorgan biodistribution while protecting their cargo to improve therapeutic efficacy. However, several hurdles that slow the transition towards clinical applications are still in the way, such as solubility, toxicity, and sub-optimal renal targeting. This review will discuss the feasibility and current limitations of drug delivery technologies for the treatment of renal disease, offering an update on their potential and the future directions of these promising strategies.
Collapse
Affiliation(s)
- Alejandro R. Chade
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS
| | - Gene L. Bidwell
- Department of Neurology, University of Mississippi Medical Center, Jackson, MS
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS
- Department of Pharmacology, University of Mississippi Medical Center, Jackson, MS
| |
Collapse
|
19
|
Sun H, Shi K, Zuo B, Zhang X, Liu Y, Sun D, Wang F. Kidney-Targeted Drug Delivery System Based on Metformin-Grafted Chitosan for Renal Fibrosis Therapy. Mol Pharm 2022; 19:3075-3084. [PMID: 35938707 DOI: 10.1021/acs.molpharmaceut.1c00827] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Our previous study demonstrated that metformin plays an anti-fibrotic role in addition to its hypoglycemic effect. Worryingly, it often requires more than 5 times the hypoglycemic dose to achieve a satisfactory anti-fibrotic effect, which greatly increases the risk of systemic acidosis caused by metformin overdose. Low-molecular-weight chitosan (LMWC) has natural kidney-targeting properties and good biocompatibility and degradability. Thus, we synthesized a novel carrier metformin-grafted chitosan (CS-MET) based on an imine reaction between oxidized chitosan and metformin. Then, GFP was recruited to form GFP-loaded CS-MET nanoparticles (CS-MET/GFP NPs) with controllable particle size. We hypothesized that CS-MET/GFP NPs would enrich in the kidney and be absorbed by HK-2 cells via megalin-mediated endocytosis by intravenous injection, which may avoid systemic acidosis caused by metformin overdose. Subsequently, the nanoparticle ruptures and releases metformin to exert its anti-apoptotic, anti-inflammatory, and anti-fibrotic effects. Our results showed that CS-MET/GFP NPs have great transfection efficiency and could enter HK-2 cells mainly through megalin-mediated endocytosis. Compared to the free metformin, CS-MET/GFP NPs showed similar anti-apoptotic ability but better therapeutic effects on cellular inflammation and fibrosis in vitro. On the other hand, CS-MET/GFP NPs showed great kidney-targeting ability and superior anti-apoptotic, anti-inflammatory, and anti-fibrotic effects in vivo.
Collapse
Affiliation(s)
- Haihan Sun
- Department of Clinical Pharmacy, The First Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, People's Republic of China
| | - Kun Shi
- Department of Orthopedics, Xuzhou Central Hospital, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu Province 221006, People's Republic of China
| | - Bangjie Zuo
- Department of Nephrology, Affiliated Yancheng Hospital, School of Medicine, Southeast University, Yancheng, Jiangsu Province 224006, People's Republic of China
| | - Xin Zhang
- Department of Nephrology, The First Clinical School of Xuzhou Medical University, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221006, People's Republic of China
| | - Yue Liu
- School of Medical Imaging, Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, People's Republic of China
| | - Dong Sun
- Department of Nephrology, The First Clinical School of Xuzhou Medical University, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221006, People's Republic of China
| | - Fengzhen Wang
- Department of Clinical Pharmacy, The First Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu Province 221004, People's Republic of China.,Department of Clinical Pharmacy, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province 221006, People's Republic of China
| |
Collapse
|
20
|
Tao X, Shi H, Cao A, Cai L. Understanding of physicochemical properties and antioxidant activity of ovalbumin-sodium alginate composite nanoparticle-encapsulated kaempferol/tannin acid. RSC Adv 2022; 12:18115-18126. [PMID: 35874031 PMCID: PMC9245490 DOI: 10.1039/d2ra02708a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/09/2022] [Indexed: 11/21/2022] Open
Abstract
In this research, ovalbumin (OVA) and sodium alginate (SA) were used as the materials to prepare an OVA–SA composite carrier, which protected and encapsulated the hydrophobic kaempferol (KAE) and the hydrophilic tannic acid (TA) (OVA–SA, OVA–TA–SA, OVA–KAE–SA, and OVA–TA–KAE–SA). Results showed that the observation of small diffraction peaks in carriers proved the successful encapsulation of KAE/TA. The protein conformation of the composite nanoparticles changed. OVA–TA–SA composite nanoparticles had the highest α-helix content and the fewest random coils, so the protein structure of it had the strongest stability. OVA–TA–KAE–SA composite nanoparticles had the strongest system stability and thermal stability, which might be due to the synergistic effect of the two polyphenols, suggesting the encapsulation of KAE/TA increased the system stability and the thermal stability of OVA–SA composite nanoparticles. Additionally, the composite nanoparticles were endowed with antioxidant ability and antibacterial ability (against Staphylococcus aureus and Escherichia coli) in the order OVA–TA–SA > OVA–TA–KAE–SA > OVA–KAE–SA based on the difference in antibacterial diameter (D, mm) and square (S, mm2), indicating that polyphenols enhanced the antibacterial and antioxidant ability of OVA–SA composite nanoparticles, and the enhancement effect of TA was stronger than that of KAE. These results provide a theoretical basis for the application of OVA–SA composite nanoparticles in the delivery of bioactive compounds. Ovalbumin (OVA) and sodium alginate (SA) were used as materials to prepare an OVA–SA composite carrier, which encapsulated the hydrophobic kaempferol (KAE) and the hydrophilic tannic acid (TA) (OVA–SA, OVA–TA–SA, OVA–KAE–SA, and OVA–TA–KAE–SA).![]()
Collapse
Affiliation(s)
- Xiaoya Tao
- Ningbo Research Institute, College of Biosystems Engineering and Food Science, Zhejiang University Ningbo 315100 China +86 571 88982726 +86 571 88982726.,Institute for Innovative Development of Food Industry, Institute for Advanced Study, Shenzhen University Shenzhen 518060 China
| | - Hang Shi
- College of Food Science and Engineering, Bohai University Jinzhou 121013 China
| | - Ailing Cao
- Hangzhou Customs District Hangzhou 310007 China
| | - Luyun Cai
- Ningbo Research Institute, College of Biosystems Engineering and Food Science, Zhejiang University Ningbo 315100 China +86 571 88982726 +86 571 88982726
| |
Collapse
|
21
|
Tao X, Shi H, Cao A, Cai L. Influence of polyphenol-metal ion-coated ovalbumin/sodium alginate composite nanoparticles on the encapsulation of kaempferol/tannin acid. Int J Biol Macromol 2022; 209:1288-1297. [PMID: 35460758 DOI: 10.1016/j.ijbiomac.2022.04.108] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 04/10/2022] [Accepted: 04/15/2022] [Indexed: 11/20/2022]
Abstract
In this research, ovalbumin (OVA) and sodium alginate (SA) were used as the materials to prepare OVA-SA composite carriers, which protected and encapsulated the hydrophobic kaempferol (KAE) and the hydrophilic tannic acid (TA). To achieve the purpose of targeted delivery, the TA-Fe3+ coating film was prepared. Results showed that the observation of small diffraction peaks in carriers proved the formation of TA/Fe3+ coating film on the surface of four composite nanoparticles (pOVA, pOVA-SA, pOVA-KAE-SA, and pOVA-KAE-TA-SA). The protein structure of the composite nanoparticles coated with TA/Fe3+ changed, and the order of the changes was pOVA-KAE > pOVA > pOVA-KAE-SA > pOVA-KAE-TA-SA > pOVA-SA. This phenomenon is due to the fact that the chromophore -C=O and the auxo-chromophore -OH are in the opposite position in the benzene ring of TA, and the two substituents have opposite effects and synergize, resulting in the different degrees of redshift of the composite nanoparticle λmax. Additionally, pOVA-SA had the highest α-helix content and the lowest random coils, conferring the protein structure the strongest stability. The coating of TA/Fe3+ increased the system stability and the thermal stability of the composite nanoparticles. Additionally, the carriers were endowed with antioxidant activity, and their antibacterial ability against Staphylococcus aureus and Escherichia coli was pOVA-KAE-TA-SA > pOVA-KAE-SA > pOVA-KAE > pOVA-SA > pOVA based on the difference in antibacterial diameter (D, mm) and square (S, mm2). pOVA-KAE-TA-SA had the strongest antioxidant activity and antibacterial ability, which improved the bioavailability of TA/KAE. These results provide a theoretical basis for the application of OVA-SA composite nanoparticles in the delivery of bioactive compounds.
Collapse
Affiliation(s)
- Xiaoya Tao
- Ningbo Research Institute, Zhejiang University, Ningbo 315100, China; College of Biosystems Engineering and Food Science, National & Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China
| | - Hang Shi
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Ailing Cao
- Hangzhou Customs District, Hangzhou 310007, China.
| | - Luyun Cai
- Ningbo Research Institute, Zhejiang University, Ningbo 315100, China; College of Biosystems Engineering and Food Science, National & Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
22
|
Cheng HT, Huang HC, Lee TY, Liao YH, Sheng YH, Jin PR, Huang KW, Chen LH, Chen YT, Liu ZY, Lin TC, Wang HC, Chao CH, Juang IP, Su CT, Huang KH, Lin SL, Wang J, Sung YC, Chen Y. Delivery of sorafenib by myofibroblast-targeted nanoparticles for the treatment of renal fibrosis. J Control Release 2022; 346:169-179. [PMID: 35429575 DOI: 10.1016/j.jconrel.2022.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 02/22/2022] [Accepted: 04/04/2022] [Indexed: 10/18/2022]
Abstract
Fibrosis is an excessive accumulation of the extracellular matrix within solid organs in response to injury and a common pathway that leads functional failure. No clinically approved agent is available to reverse or even prevent this process. Herein, we report a nanotechnology-based approach that utilizes a drug carrier to deliver a therapeutic cargo specifically to fibrotic kidneys, thereby improving the antifibrotic effect of the drug and reducing systemic toxicity. We first adopted in vitro-in vivo combinatorial phage display technology to identify peptide ligands that target myofibroblasts in mouse unilateral ureteral obstruction (UUO)-induced fibrotic kidneys. We then engineered lipid-coated poly(lactic-co-glycolic acid) nanoparticles (NPs) with fibrotic kidney-homing peptides on the surface and sorafenib, a potent antineoplastic multikinase inhibitor, encapsulated in the core. Sorafenib loaded in the myofibroblast-targeted NPs significantly reduced the infiltration of α-smooth muscle actin-expressing myofibroblasts and deposition of collagen I in UUO-treated kidneys and enhanced renal plasma flow measured by Technetium-99m mercaptoacetyltriglycine scintigraphy. This study demonstrates the therapeutic potential of the newly identified peptide fragments as anchors to target myofibroblasts and represents a strategic advance for selective delivery of sorafenib to treat renal fibrosis. SIGNIFICANCE STATEMENT: Renal fibrosis is a pathological feature accounting for the majority of issues in chronic kidney disease (CKD), which may progress to end-stage renal disease (ESRD). This manuscript describes a myofibroblast-targeting drug delivery system modified with phage-displayed fibrotic kidney-homing peptides. By loading the myofibroblast-targeting nanoparticles (NPs) with sorafenib, a multikinase inhibitor, the NPs could suppress collagen synthesis in cultured human myofibroblasts. When given intravenously to mice with UUO-induced renal fibrosis, sorafenib loaded in myofibroblast-targeting NPs significantly ameliorated renal fibrosis. This approach provides an efficient therapeutic option to renal fibrosis. The myofibroblast-targeting peptide ligands and nanoscale drug carriers may be translated into clinical application in the future.
Collapse
Affiliation(s)
- Hui-Teng Cheng
- Department of Internal Medicine, National Taiwan University Hospital Hsinchu Biomedical Park Branch, Zhu Bei City 302, Taiwan; Department of Internal Medicine, National Taiwan University Hospital Hsinchu Branch, Hsinchu City 30059, Taiwan
| | - Hsi-Chien Huang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan; Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Tsung-Ying Lee
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yu-Hui Liao
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yi-Hua Sheng
- Department of Internal Medicine, National Taiwan University Hospital Hsinchu Biomedical Park Branch, Zhu Bei City 302, Taiwan; Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Pei-Ru Jin
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Kuan-Wei Huang
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ling-Hsuan Chen
- Department of Internal Medicine, National Taiwan University Hospital Hsinchu Biomedical Park Branch, Zhu Bei City 302, Taiwan; Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yi-Ting Chen
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Zi-Yan Liu
- Institute of Electrical and Control Engineering, National Yang Ming Chiao Tung University, Taiwan
| | - Tzu-Chieh Lin
- Institute of Electrical and Control Engineering, National Yang Ming Chiao Tung University, Taiwan
| | - Hsueh-Cheng Wang
- Institute of Electrical and Control Engineering, National Yang Ming Chiao Tung University, Taiwan
| | - Cheng-Han Chao
- Department of Internal Medicine, National Taiwan University Hospital Hsinchu Branch, Hsinchu City 30059, Taiwan
| | - I Pu Juang
- Department of Pathology, National Taiwan University Hospital Hsinchu Branch, Hsinchu City 30059, Taiwan
| | - Chi-Ting Su
- Department of Nephrology, Internal Medicine, National Taiwan University Hospital Yun-Lin Branch, Douliu City, Taiwan; Department of Human Genetics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kuo-How Huang
- Department of Urology, College of Medicine, National Taiwan University, and National Taiwan University Hospital, Taipei 100, Taiwan
| | - Shuei-Liong Lin
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; Renal Division, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Jane Wang
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yun-Chieh Sung
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Yunching Chen
- Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
23
|
Zhu S, Yu C, Zhao M, Liu N, Chen Z, Liu J, Li G, Deng Y, Sai X, Huang H, Guo H, Chen C, Wang X, Zheng Y, Sun T, Chen J, Zhuang J, Zhu P. Histatin-1 loaded multifunctional, adhesive and conductive biomolecular hydrogel to treat diabetic wound. Int J Biol Macromol 2022; 209:1020-1031. [PMID: 35390401 DOI: 10.1016/j.ijbiomac.2022.03.214] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 03/09/2022] [Accepted: 03/31/2022] [Indexed: 11/05/2022]
Abstract
Micro-/macroangiopathy, neuropathy and prolonged inflammation are common in diabetic wound, however, traditional wound dressing cannot treat these problems in the same time. Herein, we developed a multifunctional hydrogel with promoted angiogenesis, cell proliferation and anti-inflammation ability to treat diabetic wound. The hydrogel was composed of natural polymers, including gelatin and chitosan, which have excellent biocompatibility. Histatin-1 (His-1) was added into the hydrogel to improve the cell adhesion, proliferation and angiogenesis. Besides, polypyrrole based conductive nanoparticles (G-Ppy) were introduced in the hydrogel to enhance the electrical signal conduction between skin and promote the mechanical strength of the hydrogel. The polypyrrole nanoparticles were growth in the chain of methacryloyl grafted gelatin (Gel-MA), leading to a better biocompatibility and water dispersibility. In vivo wound healing experiment proved that the hydrogel accelerated the wound healing rate, down regulation the expression of pro-inflammation factor TNF-α and upregulation the expression of CD31 and α-SMA, indicating the prospects in the application of diabetic wound healing.
Collapse
Affiliation(s)
- Shuoji Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Changjiang Yu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Mingyi Zhao
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Nanbo Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - ZeRui Chen
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Jian Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Ge Li
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Yuzhi Deng
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Xiyalatu Sai
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Huanlei Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Huiming Guo
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Chen Chen
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Xiaowu Wang
- Department of Cardiovascular Surgery, ZhuJiang Hospital of Southern Medical University, Guangzhou 510280, China
| | - Yuanyuan Zheng
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Tucheng Sun
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China
| | - Jimei Chen
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China.
| | - Jian Zhuang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China.
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510100, China.
| |
Collapse
|
24
|
Bio-orthogonally crosslinked catechol–chitosan hydrogel for effective hemostasis and wound healing. Carbohydr Polym 2022; 281:119039. [DOI: 10.1016/j.carbpol.2021.119039] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 12/01/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022]
|
25
|
Advancements in nanomedicines for the detection and treatment of diabetic kidney disease. BIOMATERIALS AND BIOSYSTEMS 2022; 6:100047. [PMID: 36824160 PMCID: PMC9934479 DOI: 10.1016/j.bbiosy.2022.100047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/22/2022] [Accepted: 03/27/2022] [Indexed: 12/18/2022] Open
Abstract
In the diabetic kidneys, morbidities such as accelerated ageing, hypertension and hyperglycaemia create a pro-inflammatory microenvironment characterised by extensive fibrogenesis. Radiological techniques are not yet optimised generating inconsistent and non-reproducible data. The gold standard procedure to assess renal fibrosis is kidney biopsy, followed by histopathological assessment. However, this method is risky, invasive, subjective and examines less than 0.01% of kidney tissue resulting in diagnostic errors. As such, less than 10% of patients undergo kidney biopsy, limiting the accuracy of the current diabetic kidney disease (DKD) staging method. Standard treatments suppress the renin-angiotensin system to control hypertension and use of pharmaceuticals aimed at controlling diabetes have shown promise but can cause hypoglycaemia, diuresis and malnutrition as a result of low caloric intake. New approaches to both diagnosis and treatment are required. Nanoparticles (NPs) are an attractive candidate for managing DKD due to their ability to act as theranostic tools that can carry drugs and enhance image contrast. NP-based point-of-care systems can provide physiological information previously considered unattainable and provide control over the rate and location of drug release. Here we discuss the use of nanotechnology in renal disease, its application to both the treatment and diagnosis of DKD. Finally, we propose a new method of NP-based DKD classification that overcomes the current systems limitations.
Collapse
|
26
|
Wang M, Zeng F, Ning F, Wang Y, Zhou S, He J, Li C, Wang C, Sun X, Zhang D, Xiao J, Hu P, Reilly S, Xin H, Xu X, Zhang X. Ceria nanoparticles ameliorate renal fibrosis by modulating the balance between oxidative phosphorylation and aerobic glycolysis. J Nanobiotechnology 2022; 20:3. [PMID: 34983531 PMCID: PMC8725394 DOI: 10.1186/s12951-021-01122-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 11/02/2021] [Indexed: 12/19/2022] Open
Abstract
Background and aims Renal fibrosis is the common outcome in all progressive forms of chronic kidney disease. Unfortunately, the pathogenesis of renal fibrosis remains largely unexplored, among which metabolic reprogramming plays an extremely crucial role in the evolution of renal fibrosis. Ceria nanoparticles (CeNP-PEG) with strong ROS scavenging and anti-inflammatory activities have been applied for mitochondrial oxidative stress and inflammatory diseases. The present study aims to determine whether CeNP-PEG has therapeutic value for renal fibrosis. Methods The unilateral ureteral obstructive fibrosis model was used to assess the therapeutic effects in vivo. Transforming growth factor beta1-induced epithelial-to-mesenchymal transition in HK-2 cells was used as the in vitro cell model. The seahorse bioscience X96 extracellular flux analyzer was used to measure the oxygen consumption rate and extracellular acidification rate. Results In the present study, CeNP-PEG treatment significantly ameliorated renal fibrosis by increased E-cadherin protein expression, and decreased α-SMA, Vimentin and Fibronectin expression both in vitro and in vivo. Additionally, CeNP-PEG significantly reduced the ROS formation and improved the levels of mitochondrial ATP. The seahorse analyzer assay demonstrated that the extracellular acidification rate markedly decreased, whereas the oxygen consumption rate markedly increased, in the presence of CeNP-PEG. Furthermore, the mitochondrial membrane potential markedly enhanced, hexokinase 1 and hexokinase 2 expression significantly decreased after treatment with CeNP-PEG. Conclusions CeNP-PEG can block the dysregulated metabolic status and exert protective function on renal fibrosis. This may provide another therapeutic option for renal fibrosis. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01122-w.
Collapse
Affiliation(s)
- Mengling Wang
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, 201203, China
| | - Feng Zeng
- Artemisinin Research Center, Institute of Science and Technology, The First Clinical Medical School, Lingnan Medical Research Center, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Fengling Ning
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, 201203, China
| | - Yinhang Wang
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, 201203, China
| | - Shilin Zhou
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, 201203, China
| | - Jiaqi He
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, 201203, China
| | - Cong Li
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, 201203, China
| | - Cong Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, Shanghai, China, Academy for Engineering and Technology, Fudan University, 20 Handan Road, Yangpu District, Shanghai, 200433, China
| | - Xiaolin Sun
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, 201203, China
| | - Dongliang Zhang
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, 201203, China
| | - Jisheng Xiao
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Ping Hu
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, 201203, China
| | - Svetlana Reilly
- Division of Cardiovascular Medicine, Department of Medicine, University of Oxford, John Radcliffe Hospital, RadcliffeOxford, UK
| | - Hong Xin
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, 201203, China.
| | - Xudong Xu
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, 201203, China.
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Minhang Hospital, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
27
|
Cui X, Ng KR, Chai KF, Chen WN. Clinically relevant materials & applications inspired by food technologies. EBioMedicine 2022; 75:103792. [PMID: 34974308 PMCID: PMC8728048 DOI: 10.1016/j.ebiom.2021.103792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 12/14/2022] Open
Abstract
Food science and technology have a fundamental and considerable overlap with medicine, and many clinically important applications were borne out of translational food science research. Globally, the food industry - through various food processing technologies - generates huge quantities of agro-waste and food processing byproducts that retain a significant biochemical potential for upcycling into important medical applications. This review explores some distinct clinical applications that are fabricable from food-based biopolymers and substances, often originating from food manufacturing side streams. These include antibacterial wound dressings and tissue scaffolding from the biopolymers cellulose and chitosan and antimicrobial food phytochemicals for combating antibiotic-resistant nosocomial infections. Furthermore, fermentation is discussed as the epitome of a translational food technology that unlocks further therapeutic value from recalcitrant food-based substrates and enables sustainable large-scale production of high-value pharmaceuticals, including novel fermented food-derived bioactive peptides (BPs).
Collapse
Affiliation(s)
- Xi Cui
- Interdisciplinary Graduate School, Nanyang Technological University, 50 Nanyang Avenue, 639798, Singapore; Advanced Environmental Biotechnology Centre, Nanyang Environment & Water Research Institute, Nanyang Technological University, 1 CleanTech Loop, CleanTech One, No. 06-08, 637141, Singapore; Food Science and Technology Programme, Nanyang Technological University, 62 Nanyang Drive, 637459, Singapore
| | - Kuan Rei Ng
- Food Science and Technology Programme, Nanyang Technological University, 62 Nanyang Drive, 637459, Singapore
| | - Kong Fei Chai
- Food Science and Technology Programme, Nanyang Technological University, 62 Nanyang Drive, 637459, Singapore
| | - Wei Ning Chen
- Food Science and Technology Programme, Nanyang Technological University, 62 Nanyang Drive, 637459, Singapore; School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, 637459, Singapore.
| |
Collapse
|
28
|
Zhang J, Hu K, Di L, Wang P, Liu Z, Zhang J, Yue P, Song W, Zhang J, Chen T, Wang Z, Zhang Y, Wang X, Zhan C, Cheng YC, Li X, Li Q, Fan JY, Shen Y, Han JY, Qiao H. Traditional herbal medicine and nanomedicine: Converging disciplines to improve therapeutic efficacy and human health. Adv Drug Deliv Rev 2021; 178:113964. [PMID: 34499982 DOI: 10.1016/j.addr.2021.113964] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/28/2021] [Accepted: 09/01/2021] [Indexed: 02/08/2023]
Abstract
Traditional herbal medicine (THM), an ancient science, is a gift from nature. For thousands of years, it has helped humans fight diseases and protect life, health, and reproduction. Nanomedicine, a newer discipline has evolved from exploitation of the unique nanoscale morphology and is widely used in diagnosis, imaging, drug delivery, and other biomedical fields. Although THM and nanomedicine differ greatly in time span and discipline dimensions, they are closely related and are even evolving toward integration and convergence. This review begins with the history and latest research progress of THM and nanomedicine, expounding their respective developmental trajectory. It then discusses the overlapping connectivity and relevance of the two fields, including nanoaggregates generated in herbal medicine decoctions, the application of nanotechnology in the delivery and treatment of natural active ingredients, and the influence of physiological regulatory capability of THM on the in vivo fate of nanoparticles. Finally, future development trends, challenges, and research directions are discussed.
Collapse
|
29
|
Wang X, Deng B, Yu M, Zeng T, Chen Y, Hu J, Wu Q, Li A. Constructing a passive targeting and long retention therapeutic nanoplatform based on water-soluble, non-toxic and highly-stable core-shell poly(amino acid) nanocomplexes. Biomater Sci 2021; 9:7065-7075. [PMID: 34590101 DOI: 10.1039/d1bm01246k] [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
Drug delivery nanoplatforms have been applied in bioimaging, medical diagnosis, drug delivery and medical therapy. However, insolubility, toxicity, instability, nonspecific targeting and short retention of many hydrophobic drugs limit their extensive applications. Herein, we have constructed a passive targeting and long retention therapeutic nanoplatform of core-shell gefitinib/poly (ethylene glycol)-polytyrosine nanocomplexes (Gef-PY NCs). The Gef-PY NCs have good water-solubility, non-toxicity (correspond to 1/10 dosage of effective gefitinib (hydrochloride) (Gef·HCl) (normal drug administration and slow-release) and high stability (120 days, 80% drug retention at 4 or 25 °C). The core-shell Gef-PY NCs present unexpected kidney targeting and drug slow-release capacity (ca. 72 h). The good water-solubility, non-toxicity and high stability of Gef-PY NCs effectively solve the bottleneck question that Gef-based therapy could be used only in intraperitoneal injection due to its insolubility and severe toxicity. Such excellent properties (e.g., water-solubility, non-toxicity, high stability, kidney targeting and long retention) of Gef-PY NCs create their prominent anti-fibrosis capabilities, such as decreasing approximately 40% tubulointerstitial fibrosis area and 68% expression of collagen I within 7 days. This therapeutic efficacy is well-matched with that of 10 times the dosage of toxic Gef·HCl. It is very hopeful that Gef-PY NCs could realize clinical applications and such a strategy offers an effective route to design high-efficiency treatments for kidney- and tumor-related diseases.
Collapse
Affiliation(s)
- Xin Wang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Bingqing Deng
- Nanobiological Medicine Center, Key Lab of Fuel Cell Technology of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Meng Yu
- Nanobiological Medicine Center, Key Lab of Fuel Cell Technology of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Tao Zeng
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yuyu Chen
- Nanobiological Medicine Center, Key Lab of Fuel Cell Technology of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Jianqiang Hu
- Nanobiological Medicine Center, Key Lab of Fuel Cell Technology of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Qianqing Wu
- Nanobiological Medicine Center, Key Lab of Fuel Cell Technology of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou, 510640, China.
| | - Aiqing Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Renal Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
30
|
Zhang Z, Xie L, Ju Y, Dai Y. Recent Advances in Metal-Phenolic Networks for Cancer Theranostics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2100314. [PMID: 34018690 DOI: 10.1002/smll.202100314] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/15/2021] [Indexed: 06/12/2023]
Abstract
Nanomedicine integrates different functional materials to realize the customization of carriers, aiming at increasing the cancer therapeutic efficacy and reducing the off-target toxicity. However, efforts on developing new drug carriers that combine precise diagnosis and accurate treatment have met challenges of uneasy synthesis, poor stability, difficult metabolism, and high cytotoxicity. Metal-phenolic networks (MPNs), making use of the coordination between phenolic ligands and metal ions, have emerged as promising candidates for nanomedicine, most notably through the service as multifunctional theranostic nanoplatforms. MPNs present unique properties, such as rapid preparation, negligible cytotoxicity, and pH responsiveness. Additionally, MPNs can be further modified and functionalized to meet specific application requirements. Here, the classification of polyphenols is first summarized, followed by the introduction of the properties and preparation strategies of MPNs. Then, their recent advances in biomedical sciences including bioimaging and anti-tumor therapies are highlighted. Finally, the main limitations, challenges, and outlooks regarding MPNs are raised and discussed.
Collapse
Affiliation(s)
- Zhan Zhang
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, SAR, 999078, China
| | - Lisi Xie
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, SAR, 999078, China
| | - Yi Ju
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, 3083, Australia
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau, SAR, 999078, China
| |
Collapse
|
31
|
Xu H, Wu T, Huang L. Therapeutic and delivery strategies of phytoconstituents for renal fibrosis. Adv Drug Deliv Rev 2021; 177:113911. [PMID: 34358538 DOI: 10.1016/j.addr.2021.113911] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 07/07/2021] [Accepted: 07/29/2021] [Indexed: 12/11/2022]
Abstract
Chronic kidney disease (CKD) is one of the most common diseases endangering human health and life. By 2030, 14 per 100,000 people may die from CKD. Renal fibrosis (RF) is an important intermediate link and the final pathological change during CKD progression to the terminal stage. Therefore, identifying safe and effective treatment methods for RF has become an important goal. In 2018, the World Health Organization introduced traditional Chinese medicine into its effective global medical program. Various phytoconstituents that affect the RF process have been extracted from different plants. Here, we review the potential therapeutic capabilities of active phytoconstituents in RF treatment and discuss how phytoconstituents can be structurally modified or combined with other ingredients to enhance efficiency and reduce toxicity. We also summarize phytoconstituent delivery strategies to overcome renal barriers and improve bioavailability and targeting.
Collapse
Affiliation(s)
- Huan Xu
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, People's Republic of China.
| | - Tianyi Wu
- Department of Pharmacy, School of Chemistry and Chemical Engineering, Liaoning Normal University, Dalian 116029, People's Republic of China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States
| |
Collapse
|
32
|
Liu D, Du Y, Jin FY, Xu XL, Du YZ. Renal Cell-Targeted Drug Delivery Strategy for Acute Kidney Injury and Chronic Kidney Disease: A Mini-Review. Mol Pharm 2021; 18:3206-3222. [PMID: 34337953 DOI: 10.1021/acs.molpharmaceut.1c00511] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Kidney diseases, including acute kidney injury (AKI) and chronic kidney disease (CKD), have become a global public health concern associated with high morbidity, mortality, and healthcare costs. However, at present, very few effective and specific drug therapies are available, owing to the poor therapeutic efficacy and systemic side effects. Kidney-targeted drug delivery, as a potential strategy for solving these problems, has received great attention in the fields of AKI and CKD in recent years. Here, we review the literature on renal targeted, more specifically, renal cell-targeted formulations of AKI and CKD that offered biodistribution data. First, we provide a broad overview of the unique structural characteristics and injured cells of acute and chronic injured kidneys. We then separately summarize literature examples of renal targeted formulations according to the difference of target cells and elaborate on the appropriate formulation design criteria for AKI and CKD. Finally, we propose a hypothetic strategy to improve the renal accumulation of glomerular cell-targeted formulation by escaping the uptake of the reticuloendothelial system and provide some perspectives for future studies.
Collapse
Affiliation(s)
- Di Liu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Yan Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Fei-Yang Jin
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Xiao-Ling Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| | - Yong-Zhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yu-Hang-Tang Road, Hangzhou 310058, China
| |
Collapse
|
33
|
Zhao Y, Pu M, Wang Y, Yu L, Song X, He Z. Application of nanotechnology in acute kidney injury: From diagnosis to therapeutic implications. J Control Release 2021; 336:233-251. [PMID: 34171444 DOI: 10.1016/j.jconrel.2021.06.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 12/29/2022]
Abstract
Acute kidney injury (AKI), a major health issue concerning ~50% of patients treated in intensive care units, generally leads to severe renal damage associated with high mortality rate. The application of nanotechnology for the management of AKI has profound potential of further development, providing innovative strategies for predicting the early onset and progression of renal disease and improving the treatment efficacy of the life-threating AKI. This review has comprehensively summarized the nanomedicines in the application of AKI diagnosis and emphatically discussed the unique potential of various nanotechnology-based drug delivery systems (e.g., polymeric nanoparticles, organic nanoparticles, inorganic nanoparticles, lipid-based nanoparticles, hydrogels etc.) in the treatment of AKI, allowing for improved therapeutic index by enhancing both efficacy and safety concurrently. These approaches may mechanically mitigate oxidative stress, inflammation, and mitochondrial and other organellar damage, etc. In addition, the combination of nanotechnology with stem cells-based therapy or gene therapy has been explored for reducing renal tissues damage and promoting kidney repair or recovery from AKI. The review provides insights into the synthesis, advantages, and limitations of innovative nanomedicine application in the early detection and effective treatment of AKI.
Collapse
Affiliation(s)
- Yi Zhao
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao 266100, China; College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266100, China
| | - Mingju Pu
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao 266100, China; College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266100, China
| | - Yanan Wang
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao 266100, China; College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266100, China
| | - Liangmin Yu
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao 266100, China; College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266100, China.
| | - Xinyu Song
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao 266100, China; College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266100, China
| | - Zhiyu He
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao 266100, China; College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266100, China.
| |
Collapse
|
34
|
Fleischmann D, Goepferich A. General sites of nanoparticle biodistribution as a novel opportunity for nanomedicine. Eur J Pharm Biopharm 2021; 166:44-60. [PMID: 34087354 DOI: 10.1016/j.ejpb.2021.05.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/22/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
The development of nanomedical devices has led to a considerable number of clinically applied nanotherapeutics. Yet, the overall poor translation of nanoparticular concepts into marketable systems has not met the initial expectations and led to increasing criticism in recent years. Most novel nano approaches thereby use highly refined formulations including a plethora of active targeting sequences, but ultimately fail to reach their target due to a generally high off-target deposition in organs such as the liver or kidney. In this context, we argue that initial nanoparticle (NP) development should not entirely become set on conventional formulation aspects. In contrast, we propose a change of focus towards a prior analysis of general sites of NP in vivo deposition and an assessment of how accumulation in these organs or tissues can be harnessed to develop therapies for site-related pathologies. We therefore give a comprehensive overview of existing nanotherapeutic targeting strategies for specific cell types within three of the usual suspects, i.e. the liver, kidney and the vascular system. We discuss the physiological surroundings and relevant pathologies of described tissues as well as the implications for NP-mediated drug delivery. Additionally, successful cell-selective NP concepts using active targeting strategies are assessed. By bringing together both (patho)physiological aspects and concepts for cell-selective NP formulations, we hope to show a novel opportunity for the development of more promising nanotherapeutic devices.
Collapse
Affiliation(s)
- Daniel Fleischmann
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany.
| |
Collapse
|
35
|
Huang X, Ma Y, Li Y, Han F, Lin W. Targeted Drug Delivery Systems for Kidney Diseases. Front Bioeng Biotechnol 2021; 9:683247. [PMID: 34124026 PMCID: PMC8193852 DOI: 10.3389/fbioe.2021.683247] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Kidney diseases have gradually become a global health burden. Along with the development of nanotechnology, many hybrids or nanomaterials have been utilized to promote treatment efficiency with negligible side effects. These therapeutic agents have been successfully applied in many fields. In particular, some efforts have also been made to ameliorate the treatment of kidney diseases through targeted delivery nanomaterials. Though most of the delivery systems have not yet been transmitted into clinical use or even still at an early stage, they have shown great potential in carrying immunosuppressants like tacrolimus and triptolide, antioxidants, or siRNAs. Excitingly, some of them have achieved significant treatment effectiveness and reduced systemic side effect in kidney disease animal models. Here, we have reviewed the recent advances and presented nanotherapeutic devices designed for kidney targeted delivery.
Collapse
Affiliation(s)
- Xiaohan Huang
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, Zhejiang University School of Medicine, The First Affiliated Hospital, Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Yanhong Ma
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, Zhejiang University School of Medicine, The First Affiliated Hospital, Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Yangyang Li
- Key Laboratory of Women's Reproductive Health Research of Zhejiang Province, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Han
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, Zhejiang University School of Medicine, The First Affiliated Hospital, Institute of Nephrology, Zhejiang University, Hangzhou, China
| | - Weiqiang Lin
- Department of Nephrology, The Fourth Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
36
|
Gao L, Zhang L, Zhu X, Chen J, Zhao M, Li S, Yu C, Hu L, Qiao H, Guo Z. Hyaluronic acid functionalized gold nanorods combined with copper-based therapeutic agents for chemo-photothermal cancer therapy. J Mater Chem B 2021; 8:4841-4845. [PMID: 32108202 DOI: 10.1039/d0tb00097c] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
We herein report a hybrid nanocomposite (AuNRs-CTN@THA) which is based on hyaluronic acid-coated gold nanorods with loading of a copper complex through strong bonds. AuNRs-CTN@THA exhibits durable photothermal conversion capacity for pH-dominant and pH/temperature dual sensitive drug release, accomplishing synergetic antitumor efficacy and deep tumor penetration.
Collapse
Affiliation(s)
- Lina Gao
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China.
| | - Lei Zhang
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China.
| | - Xuyu Zhu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China.
| | - Jing Chen
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China
| | - Meng Zhao
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China.
| | - Simin Li
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China.
| | - Chengli Yu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China.
| | - Lihong Hu
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China.
| | - Hongzhi Qiao
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China. and Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing University of Chinese Medicine, Nanjing 210023, P. R. China and State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
37
|
Li J, Du N, Tan Y, Hsu HY, Tan C, Jiang Y. Conjugated Polymer Nanoparticles Based on Copper Coordination for Real-Time Monitoring of pH-Responsive Drug Delivery. ACS APPLIED BIO MATERIALS 2021; 4:2583-2590. [PMID: 35014375 DOI: 10.1021/acsabm.0c01564] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Metal coordination-driven composite systems have excellent stability and pH-responsive ability, making them suitable for specific drug delivery in physiological conditions. In this study, an anionic conjugated polymer PPEIDA with a poly(p-phenylene ethynylene) backbone and iminodiacetic acid (IDA) side chains is used as a drug carrier to construct a class of pH-responsive nanoparticles, PPEIDA-Cu-DOX conjugated polymer nanoparticles (CPNs), by taking advantage of the metal coordination interaction of Cu2+ with PPEIDA and the drug doxorubicin (DOX). PPEIDA-Cu-DOX CPNs have high drug loading and encapsulation efficiency (EE), calculated to be 54.30 ± 1.10 and 95.80 ± 0.84%, respectively. Due to the good spectral overlap, Förster resonance energy transfer (FRET) takes place between PPEIDA and the drug DOX, which enables the observation of the loading and the release of DOX from CPNs in an acidic environment by monitoring fluorescence emission changes. Therefore, PPEIDA-Cu-DOX CPNs can also be used in real-time cell imaging to monitor drug release in addition to delivering DOX targeting tumor cells. Compared with free DOX, PPEIDA-Cu-DOX CPNs show a similar inhibition to tumor cells and lower toxicity to normal cells. Our results demonstrate the feasibility and potential of constructing pH-responsive CPNs via metal-ligand coordination interactions for cancer treatment.
Collapse
Affiliation(s)
- Jiatong Li
- State Key Laboratory of Chemical Oncogenomics, The Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China.,Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Nan Du
- State Key Laboratory of Chemical Oncogenomics, The Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Ying Tan
- State Key Laboratory of Chemical Oncogenomics, The Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Hsien-Yi Hsu
- School of Energy and Environment & Department of Materials Science and Engineering, City University of Hong Kong, Kowloon Tong, Hong Kong, P. R. China.,Shenzhen Research Institute of City, University of Hong Kong, Shenzhen 518057, P. R. China
| | - Chunyan Tan
- State Key Laboratory of Chemical Oncogenomics, The Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China
| | - Yuyang Jiang
- State Key Laboratory of Chemical Oncogenomics, The Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, P. R. China.,School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
38
|
Samyn P. A platform for functionalization of cellulose, chitin/chitosan, alginate with polydopamine: A review on fundamentals and technical applications. Int J Biol Macromol 2021; 178:71-93. [PMID: 33609581 DOI: 10.1016/j.ijbiomac.2021.02.091] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 02/09/2021] [Accepted: 02/12/2021] [Indexed: 12/19/2022]
Abstract
Nature provides concepts and materials with interesting functionalities to be implemented in innovative and sustainable materials. In this review, it is illustrated how the combination of biological macromolecules, i.e. polydopamine and polysaccharides (cellulose, chitin/chitosan, alginate), enables to create functional materials with controlled properties. The mussel-adhesive properties rely on the secretion of proteins having 3,4-dihydroxyphenylalanine amino acid with catechol groups. Fundamental understanding on the biological functionality and interaction mechanisms of dopamine in the mussel foot plaque is presented in parallel with the development of synthetic analogues through extraction or chemical polymer synthesis. Subsequently, modification of cellulose, chitin/chitosan or alginate and their nanoscale structures with polydopamine is discussed for various technical applications, including bio- and nanocomposites, films, filtration or medical membranes, adhesives, aerogels, or hydrogels. The presence of polydopamine stretches far beyond surface adhesive properties, as it can be used as an intermediate to provide additional performance of hydrophobicity, self-healing, antimicrobial, photocatalytic, sensoric, adsorption, biocompatibility, conductivity, coloring or mechanical properties. The dopamine-based 'green' chemistry can be extended towards generalized catechol chemistry for modification of polysaccharides with tannic acid, caffeic acid or laccase-mediated catechol functionalization. Therefore, the modification of polysaccharides with polydopamine or catechol analogues provides a general platform for sustainable material functionalization.
Collapse
Affiliation(s)
- Pieter Samyn
- Hasselt University, Institute for Materials Research, Applied and Analytical Chemistry, Agoralaan Gebouw D, B-3590 Diepenbeek, Belgium.
| |
Collapse
|
39
|
Bai L, Wang M, Zhang L, Zhao M, Ren M, Zheng L, Lei M, Shen H. Poly(Amino Acid) Coordination Nanoparticle as a Potent Sonosensitizer for Cancer Therapy. ACS APPLIED BIO MATERIALS 2021. [DOI: 10.1021/acsabm.0c01383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Lintao Bai
- State Key Laboratory of Organic−Inorganic Composites, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Mingkun Wang
- State Key Laboratory of Organic−Inorganic Composites, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Lin Zhang
- State Key Laboratory of Chemical Resource Engineering, Institute of Computational Chemistry, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Meijun Zhao
- State Key Laboratory of Organic−Inorganic Composites, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Mei Ren
- State Key Laboratory of Organic−Inorganic Composites, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Lirong Zheng
- Beijing Synchrotron Radiation Facility, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Ming Lei
- State Key Laboratory of Chemical Resource Engineering, Institute of Computational Chemistry, College of Chemistry, Beijing University of Chemical Technology, Beijing 100029, P.R. China
| | - Heyun Shen
- State Key Laboratory of Organic−Inorganic Composites, Beijing Key Laboratory of Bioprocess, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| |
Collapse
|
40
|
Montroni D, Palanca M, Morellato K, Fermani S, Cristofolini L, Falini G. Hierarchical chitinous matrices byssus-inspired with mechanical properties tunable by Fe(III) and oxidation. Carbohydr Polym 2021; 251:116984. [DOI: 10.1016/j.carbpol.2020.116984] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/05/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
|
41
|
Engineering of stepwise-targeting chitosan oligosaccharide conjugate for the treatment of acute kidney injury. Carbohydr Polym 2020; 256:117556. [PMID: 33483059 DOI: 10.1016/j.carbpol.2020.117556] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/10/2020] [Accepted: 12/19/2020] [Indexed: 01/20/2023]
Abstract
Acute kidney injury (AKI) is a common and serious clinical syndrome of acute renal dysfunction in a short period. One of therapeutic interventions for AKI is to reduce ROS massively generated in the mitochondria and then ameliorate cell damage and apoptosis induced by oxidative stress. In this study, stepwise-targeting chitosan oligosaccharide, triphenyl phosphine-low molecular weight chitosan-curcumin (TPP-LMWC-CUR, TLC), was constructed for sepsis-induced AKI via removing excessive ROS in renal tubular epithelial cells. Benefiting from good water solubility and low molecular weight, TLC was rapidly and preferentially distributed in the renal tissues and then specifically internalized by tubular epithelium cells via interaction between Megalin receptor and LMWC. The intracellular TLC could further delivery CUR to mitochondria due to high buffering capacity of LMWC and delocalized positive charges of TPP. Both in vitro and in vivo pharmacodynamic results demonstrated the enhanced therapeutic effect of TLC in the treatment of AKI.
Collapse
|
42
|
Chitosan coordination driven self-assembly for effective delivery of curcumin. Int J Biol Macromol 2020; 165:2267-2274. [PMID: 33098899 DOI: 10.1016/j.ijbiomac.2020.10.097] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/03/2020] [Accepted: 10/13/2020] [Indexed: 02/07/2023]
Abstract
Self-assembly of metal-ligand coordination is of immense scientific interest in supramolecular construction of functional materials duo to their desirable functional properties. Herein, we investigated a designable coordination driven self-assembly to simultaneously enhance the water solubility and biological stability of curcumin (Cur). On the basis of amino group in chitosan (CS), it was chosen as the high-affinity anchors for coordination nanocomplexes, in which Cur were incorporated into the amino group by coordination bonding, forming a CS-metal-Cur architecture. The sizes of these nanocomplexes can be tuned by the feed concentrations of CS as well as the kind of metal ions. Time dependent absorption spectral measurements demonstrated the significant increase in hydrolytic stability of Cur after forming nanoparticles (NPs) especially for the CS-Cu-Cur NPs. Particularly, the formed CS-metal-Cur NPs can be efficiently triggered by pH, which was stable under physiological conditions while releasing encapsulated drugs under low pH conditions in a sustained manner. Based on cellular uptake study and cytotoxicity experiments, CS-metal-Cur NPs were shown to possess highly efficient internalization and an apparent cytotoxic effect. The high drug-loading capacities and responses to pH value, substantially enhanced antitumor activity of Cur provided this nanocomplex with promising properties for biomimetic and biomedical applications.
Collapse
|
43
|
van Asbeck AH, Dieker J, Boswinkel M, van der Vlag J, Brock R. Kidney-targeted therapies: A quantitative perspective. J Control Release 2020; 328:762-775. [DOI: 10.1016/j.jconrel.2020.09.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 01/23/2023]
|
44
|
Wei S, Xu C, Zhang Y, Shi Z, Wu M, Yang B. Ultrasound Assisted a Peroxisome Proliferator-Activated Receptor (PPAR)γ Agonist-Loaded Nanoparticle-Microbubble Complex to Attenuate Renal Interstitial Fibrosis. Int J Nanomedicine 2020; 15:7315-7327. [PMID: 33061383 PMCID: PMC7537998 DOI: 10.2147/ijn.s262052] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/15/2020] [Indexed: 12/25/2022] Open
Abstract
Objective To investigate the antifibrotic effect of the combination of a PPARγ agonist-loaded nanoparticle-microbubble complex with ultrasound (US) exposure on renal interstitial fibrosis (RIF). Materials and Methods Polylactide-co-glycolide (PLGA) nanoparticles were used to load PPARγ agonist (rosiglitazone, RSG) and prepare PLGA-RSG nanoparticles (PLNPs-RSG); then, a novel complex between PLNPs-RSG and SonoVue microbubbles (MBs) (PLNPs-RSG-MBs) was prepared. The size distribution, zeta potentials, RSG-loading capacity and entrapment efficiency were measured, and the release of RSG was assessed using a UV-vis spectrophotometer. The in vitro cytotoxicity and in vivo systemic toxicity assays were performed. The cellular uptake assessment was performed using a confocal laser scanning microscope (CLSM). The in vivo biodistribution assessment was performed using fluorescence imaging with a near-infrared (NIR) imaging system. Furthermore, this complex was administered to a unilateral ureteral obstruction (UUO) rat model with the assistance of US exposure to investigate the antifibrotic effect. Results This PLNPs-RSG-MBs complex had a size of 2199.5± 988.1 nm and a drug-loading efficiency of 28.5%. In vitro cytotoxicity and in vivo systemic toxicity assays indicated that the PLNPs-RSG-MBs complex displayed excellent biocompatibility. In addition, the complex showed high cellular uptake efficiency in vitro and kidney-targeting ability in vivo. In a UUO rat model, the combination of the PLNPs-RSG-MBs complex with US exposure significantly reduced collagen deposition and successfully attenuated renal fibrosis. Conclusion The combination of the PLNPs-RSG-MBs complex with US exposure may be a promising approach for the treatment of RIF.
Collapse
Affiliation(s)
- Shuping Wei
- Department of Ultrasound, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Chaoli Xu
- Department of Ultrasound, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, People's Republic of China
| | - Yidan Zhang
- Department of Ultrasound, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Zhongqing Shi
- Department of Cardiac Function, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Min Wu
- Department of Ultrasound, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, People's Republic of China
| | - Bin Yang
- Department of Ultrasound, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, People's Republic of China
| |
Collapse
|
45
|
Acharya A, Patial V. Nanotechnological interventions for the treatment of renal diseases: Current scenario and future prospects. J Drug Deliv Sci Technol 2020; 59:101917. [DOI: 10.1016/j.jddst.2020.101917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
46
|
Tan L, Deng X, Lai X, Zeng T, Li A, Hu J, Xiong Z. Mesoscale nanoparticles encapsulated with emodin for targeting antifibrosis in animal models. OPEN CHEM 2020. [DOI: 10.1515/chem-2020-0163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
AbstractThe aim of this study is to explore the kidney-targeting capability of mesoscale nanoparticles (MNPs)-emodin (Em-MNPs) and its potential antifibrosis in the animal model. First, MNPs and Em-MNPs were synthesized via nanoprecipitation method, and their diameters were both ∼400 nm with the uniform size. The entrapment efficiency of MNPs was 45.1% when adding emodin at the concentration of 12 mg/mL. Moreover, cytotoxicity assay showed that Em-MNPs presented excellent biocompatibility in rat proximal tubular cells. Cellular uptake assay demonstrated that Em-MNPs had high-efficiency uptake, especially in the cytoplasm. Ex vivo organ fluorescence imaging revealed that Em-MNPs possessed specific kidney-targeting ability with relative long retention time in the kidney (∼24 h). In the renal unilateral ureteral obstruction model, Em-MNPs treatment could significantly alleviate kidney tubule injury and reduce extracellular matrix deposition compared with free MNPs. Herein, Em-MNPs with specific kidney-targeting and preferable antifibrosis effects in animal model may pave an avenue for treating renal diseases.
Collapse
Affiliation(s)
- Lishan Tan
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
| | - Xiulong Deng
- Department of Chemical and Chemical Engineering, Key Laboratory of Organo-Pharmaceutical Chemistry, Gannan Normal University, Ganzhou, Jiangxi Province, 341000, China
| | - Xuandi Lai
- Department of Oncology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China
| | - Tao Zeng
- Department of Nephrology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Aiqing Li
- Department of Nephrology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jianqiang Hu
- Department of Nephrology, State Key Laboratory of Organ Failure Research, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zuying Xiong
- Department of Nephrology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, 518036, China
| |
Collapse
|
47
|
Giménez VMM, Fuentes LB, Kassuha DE, Manucha W. Current Drug Nano-targeting Strategies for Improvement in the Diagnosis and Treatment of Prevalent Pathologies such as Cardiovascular and Renal Diseases. Curr Drug Targets 2020; 20:1496-1504. [PMID: 31267869 DOI: 10.2174/1389450120666190702162533] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/10/2019] [Accepted: 06/17/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND The kidney and cardiovascular system are closely related to each other during the modulation of the cardiovascular homeostasis. However, the search for new alternatives for the treatment and diagnosis of cardiovascular diseases does not take into account this relationship, so their evaluation results and the advantages offered by their global and integrative analysis are wasted. For example, a variety of receptors that are overexpressed in both pathologies is large enough to allow expansion in the search for new molecular targets and ligands. Nanotechnology offers pharmacological targeting strategies to kidney, heart, and blood vessels for overcoming one of the essential restrictions of traditional cardiovascular therapies the ones related to their unspecific pharmacodynamics distribution in these critical organs. RECENT FINDINGS Drug or contrast agent nano-targeting for treatment or diagnosis of atherosclerosis, thrombosis, renal cancer or fibrosis, glomerulonephritis, among other renal, cardiac and blood vessels pathologies would allow an increase in their efficacy and a reduction of their side effects. Such effects are possible because, through pharmacological targeting, the drug is mainly found at the desired site. Review Purpose: In this mini-review, active, passive, and physical targeting strategies of several nanocarriers that have been assessed and proposed for the treatment and diagnosis of different cardiovascular diseases, are being addressed.
Collapse
Affiliation(s)
- Virna Margarita Martín Giménez
- Instituto de Investigaciones en Ciencias Quimicas, Facultad de Ciencias Quimicas y Tecnologicas, Universidad Catolica de Cuyo, San Juan, Argentina
| | - Lucía Beatriz Fuentes
- Facultad de Quimica, Bioquimica y Farmacia, Universidad Nacional de San Luis, San Luis, Argentina
| | - Diego Enrique Kassuha
- Instituto de Investigaciones en Ciencias Quimicas, Facultad de Ciencias Quimicas y Tecnologicas, Universidad Catolica de Cuyo, San Juan, Argentina
| | - Walter Manucha
- Instituto de Medicina y Biologia Experimental de Cuyo, Consejo Nacional de Investigacion Científica y Tecnologica (IMBECU-CONICET), Mendoza, Argentina.,Laboratorio de Farmacología Experimental Básica y Traslacional. Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
48
|
Lai X, Geng X, Tan L, Hu J, Wang S. A pH-Responsive System Based on Fluorescence Enhanced Gold Nanoparticles for Renal Targeting Drug Delivery and Fibrosis Therapy. Int J Nanomedicine 2020; 15:5613-5627. [PMID: 32884257 PMCID: PMC7440925 DOI: 10.2147/ijn.s260069] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/17/2020] [Indexed: 11/23/2022] Open
Abstract
Background Stimuli-responsive gold nano-assemblies have attracted attention as drug delivery systems in the biomedical field. However, there are challenges achieving targeted delivery and controllable drug release for specific diseases. Materials and Methods In this study, a glutathione (GSH)-modified fluorescent gold nanoparticle termed AuLA-GSH was prepared and a Co2+-induced self-assembly drug delivery platform termed AuLA-GSH-Co was constructed. Both the pH-responsive character and drug loading behavior of AuLA-GSH-Co were studied in vitro. Kidney-targeting capability was investigated in vitro and in vivo. Finally, the anti-fibrosis efficiency of AuLA-GSH-Co in a mouse model of unilateral ureteral obstruction (UUO) was explored. Results AuLA-GSH-Co was sensitive to pH changes and released Co2+ in acidic conditions, allowing it to have controllable drug release abilities. AuLA-GSH-Co was found to improve cellular uptake of Co2+ ions compared to CoCl2 in vitro. AuLA-GSH exhibited specific renal targeting and prolonged renal retention time with low non-specific accumulation in vivo. Moreover, the anti-fibrosis efficiency of AuLA-GSH-Co was higher compared to CoCl2 in a mouse model of unilateral ureteral obstruction (UUO). Conclusion AuLA-GSH-Co could greatly enhance drug delivery efficiency with renal targeting capability and obviously relieve renal fibrosis, providing a promising strategy for renal fibrosis therapy.
Collapse
Affiliation(s)
- Xuandi Lai
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, People's Republic of China
| | - Xinran Geng
- Nanobiological Medicine Center, Key Laboratory of Fuel Cell Technology of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, People's Republic of China
| | - Lishan Tan
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, People's Republic of China
| | - Jianqiang Hu
- Nanobiological Medicine Center, Key Laboratory of Fuel Cell Technology of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, People's Republic of China
| | - Shubin Wang
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, People's Republic of China
| |
Collapse
|
49
|
Kim CS, Mathew AP, Uthaman S, Moon MJ, Bae EH, Kim SW, Park IK. Glycol chitosan-based renal docking biopolymeric nanomicelles for site-specific delivery of the immunosuppressant. Carbohydr Polym 2020; 241:116255. [DOI: 10.1016/j.carbpol.2020.116255] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 03/25/2020] [Accepted: 04/02/2020] [Indexed: 01/26/2023]
|
50
|
Chen Z, Peng H, Zhang C. Advances in kidney-targeted drug delivery systems. Int J Pharm 2020; 587:119679. [PMID: 32717283 DOI: 10.1016/j.ijpharm.2020.119679] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/28/2020] [Accepted: 07/18/2020] [Indexed: 12/19/2022]
Abstract
The management and treatment of kidney diseases currently have caused a huge global burden. Although the application of nanotechnology for the therapy of kidney diseases is still at an early stages, it has profound potential of development. More and more nano-based drug delivery systems provide novel solutions for the treatment of kidney diseases. This article summarizes the physiological and anatomical properties of the kidney and the biological and physicochemical characters of drug delivery systems, which affects the ability of drug to target the kidney, and highlights the prospects, opportunities, and challenges of nanotechnology in the therapy of kidney diseases.
Collapse
Affiliation(s)
- Zhong Chen
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, 1 Xinyang Rd, Daqing 163319, China
| | - Haisheng Peng
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, 1 Xinyang Rd, Daqing 163319, China.
| | - Changmei Zhang
- Department of Pharmaceutics, Daqing Campus of Harbin Medical University, 1 Xinyang Rd, Daqing 163319, China.
| |
Collapse
|