1
|
Lu C, Li C, Gu N, Yang F. Emerging Elastic Micro-Nano Materials for Diagnosis and Treatment of Thrombosis. RESEARCH (WASHINGTON, D.C.) 2025; 8:0614. [PMID: 40028043 PMCID: PMC11868703 DOI: 10.34133/research.0614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 03/05/2025]
Abstract
Thrombus is a blood clot that forms in a blood vessel at the point of flaking. Thrombosis is closely associated with cardiovascular diseases caused by different sources and factors. However, the current clinical methods of thrombus diagnosis and treatment still have problems with targeting, permeability, stability, and biosafety. Therefore, in recent years, based on the development of micro/nano technology, researchers have tried to develop some new strategies for the diagnosis and treatment of thrombosis. Due to the unique structural characteristics, the micro-nano materials in physiological environments show excellent transport and delivery properties such as better in vivo circulation, longer life span, better targeting ability, and controllable cellular internalization. Especially, elasticity and stiffness are inherent mechanical properties of some well-designed micro-nano materials, which can make them better adapted to the needs of thrombosis diagnosis and treatment. Herein, this review first introduces the thrombotic microenvironment to characterize the thrombus development process. Then, to fine-tune the pathological occurrence and development of thrombosis, the role of elastic micro-nano materials for thrombus diagnosis and treatment is summarized. The properties, preparation methods, and biological fate of these materials have been discussed in detail. Following, the applications of elastic micro-nano materials in biomedical imaging, drug delivery, and therapy of thrombosis are highlighted. Last, the shortcomings and future design strategies of elastic micro-nano materials in diagnosis and treatment of clinical thrombosis are discussed. This review will provide new ideas for the use of nanotechnology in clinical diagnosis and treatment of thrombus in the future.
Collapse
Affiliation(s)
- Chenxin Lu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering,
Southeast University, Nanjing 210096, P. R. China
| | - Chunjian Li
- Department of Cardiology,
The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Ning Gu
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, Institute of Clinical Medicine, Nanjing Drum Tower Hospital, Medical School,
Nanjing University, Nanjing 210093, P. R. China
| | - Fang Yang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering,
Southeast University, Nanjing 210096, P. R. China
| |
Collapse
|
2
|
Zhao Z, Song H, Qi M, Liu Y, Zhang Y, Li S, Zhang H, Sun Y, Sun Y, Gao Z. Brain targeted polymeric micelles as drug carriers for ischaemic stroke treatment. J Drug Target 2025; 33:232-248. [PMID: 39403962 DOI: 10.1080/1061186x.2024.2417190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/30/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Ischaemic stroke is a central nervous system disease with high morbidity, recurrence and mortality rates. Thrombolytic and neuroprotective therapies are the main therapeutic strategies for ischaemic stroke, however, the poor delivery efficiency of thrombolytic and neuroprotective drugs to the brain limits their clinical application. So far, the development of nanomedicine has brought opportunities for the above challenges, which can not only realise the effective accumulation of drugs in the target site, but also improve the pharmacokinetic behaviour of the drugs. Among the most rapidly developing nanoparticles, micelles gradually emerging as an effective strategy for ischaemic stroke treatment due to their own unique advantages. This review provided an overview of targeted and response-release micelles based on the physicochemical properties of the ischaemic stroke microenvironment, summarised the targeting strategies for delivering micellar formulations to the thrombus, blood-brain barrier, and brain parenchyma, and finally described the potentials and challenges of polymeric micelles in the treatment of ischaemic stroke.
Collapse
Affiliation(s)
- Zirui Zhao
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Huijia Song
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Mengge Qi
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yurong Liu
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yanchao Zhang
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Shuo Li
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Huimin Zhang
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yongjun Sun
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Yanping Sun
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| | - Zibin Gao
- Department of Pharmacy, School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
- State Key Laboratory Breeding Base - Hebei Province Key Laboratory of Molecular Chemistry for Drugs, Hebei University of Science and Technology, Shijiazhuang, China
- Hebei Research Center of Pharmaceutical and Chemical Engineering, Hebei University of Science and Technology, Shijiazhuang, China
| |
Collapse
|
3
|
Martín‐Morales C, Caspani S, Desco M, Tavares de Sousa C, Gómez‐Gaviro MV. Controlled Drug Release Systems for Cerebrovascular Diseases. ADVANCED THERAPEUTICS 2024. [DOI: 10.1002/adtp.202400239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Indexed: 01/06/2025]
Abstract
AbstractThis review offers a comprehensive exploration of optimized drug delivery systems tailored for controlled release and their crucial role in addressing cerebrovascular diseases. Through an in‐depth analysis, various controlled release methods, including nanoparticles, liposomes, hydrogels, and other emerging technologies are examined. Highlighting the importance of precise drug targeting, it is delved into the underlying mechanisms of these delivery systems and their potential to improve therapeutic outcomes while minimizing adverse effects. Additionally, the specific applications of these optimized drug delivery systems in treating cerebrovascular disorders such as ischemic stroke, cerebral aneurysms, and intracranial hemorrhage are discussed. By shedding light on the advancements in drug delivery techniques and their implications in cerebrovascular medicine, this review offers valuable insights into the future of therapeutic interventions in neurology.
Collapse
Affiliation(s)
- Celia Martín‐Morales
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM) Doctor Esquerdo 46 Madrid 28007 Spain
| | - Sofia Caspani
- IFIMUP – Institute of Physics for Advanced Materials Departamento de Física e Astronomia, Nanotechnology and Photonics of University of Porto Faculdade de Ciências Universidade do Porto, Rua do Campo Alegre s/n Porto 4169‐007 Portugal
| | - Manuel Desco
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM) Doctor Esquerdo 46 Madrid 28007 Spain
- Departamento de Bioingeniería Universidad Carlos III de Madrid Leganés 28911 Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM) Madrid 28029 Spain
- Centro de Investigaciones Cardiovasculares (CNIC) Melchor Fernández Almagro Madrid 28029 Spain
| | - Célia Tavares de Sousa
- Departamento de Física Aplicada and IAdChem Facultad de Ciencias Universidad Autonoma de Madrid (UAM) Campus de Cantoblanco, C/ Francisco Tomas y Valiente, 7 Madrid 28049 Spain
| | - María Victoria Gómez‐Gaviro
- Unidad de Medicina y Cirugía Experimental, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM) Doctor Esquerdo 46 Madrid 28007 Spain
- Departamento de Bioingeniería Universidad Carlos III de Madrid Leganés 28911 Spain
- Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM) Madrid 28029 Spain
| |
Collapse
|
4
|
Huang Y, Wang J, Guo Y, Shen L, Li Y. Fibrinogen binding to activated platelets and its biomimetic thrombus-targeted thrombolytic strategies. Int J Biol Macromol 2024; 274:133286. [PMID: 38908635 DOI: 10.1016/j.ijbiomac.2024.133286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
Thrombosis is associated with various fatal arteriovenous syndromes including ischemic stroke, myocardial infarction, and pulmonary embolism. However, current clinical thrombolytic treatment strategies still have many problems in targeting and safety to meet the thrombolytic therapy needs. Understanding the molecular mechanism that underlies thrombosis is critical in developing effective thrombolytic strategies. It is well known that platelets play a central role in thrombosis and the binding of fibrinogen to activated platelets is a common pathway in the process of clot formation. Based on this, a concept of biomimetic thrombus-targeted thrombolytic strategy inspired from fibrinogen binding to activated platelets in thrombosis was proposed, which could selectively bind to activated platelets at a thrombus site, thus enabling targeted delivery and local release of thrombolytic agents for effective thrombolysis. In this review, we first summarized the main characteristics of platelets and fibrinogen, and then introduced the classical molecular mechanisms of thrombosis, including platelet adhesion, platelet activation and platelet aggregation through the interactions of activated platelets with fibrinogen. In addition, we highlighted the recent advances in biomimetic thrombus-targeted thrombolytic strategies which inspired from fibrinogen binding to activated platelets in thrombosis. The possible future directions and perspectives in this emerging area are briefly discussed.
Collapse
Affiliation(s)
- Yu Huang
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China.
| | - Jiahua Wang
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China
| | - Yuanyuan Guo
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China
| | - Lingyue Shen
- Department of Oral & Maxillofacial-Head & Neck Oncology, Department of Laser and Aesthetic Medicine, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stoma-tology & Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai 200011, PR China.
| | - Yuehua Li
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China.
| |
Collapse
|
5
|
Ye Y, Chen Z, Zhang S, Slezak P, Lu F, Xie R, Lee D, Lan G, Hu E. pH-Responsive Theranostic Colloidosome Drug Carriers Enable Real-Time Imaging of Targeted Thrombolytic Process with Near-Infrared-II for Deep Venous Thrombosis. RESEARCH (WASHINGTON, D.C.) 2024; 7:0388. [PMID: 38812529 PMCID: PMC11136571 DOI: 10.34133/research.0388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 04/21/2024] [Indexed: 05/31/2024]
Abstract
Thrombosis can cause life-threatening disorders. Unfortunately, current therapeutic methods for thrombosis using injecting thrombolytic medicines systemically resulted in unexpected bleeding complications. Moreover, the absence of practical imaging tools for thrombi raised dangers of undertreatment and overtreatment. This study develops a theranostic drug carrier, Pkr(IR-Ca/Pda-uPA)-cRGD, that enables real-time monitoring of the targeted thrombolytic process of deep vein thrombosis (DVT). Pkr(IR-Ca/Pda-uPA)-cRGD, which is prepared from a Pickering-emulsion-like system, encapsulates both near-infrared-II (NIR-II) contrast agent (IR-1048 dye, loading capacity: 28%) and urokinase plasminogen activators (uPAs, encapsulation efficiency: 89%), pioneering the loading of multiple drugs with contrasting hydrophilicity into one single-drug carrier. Upon intravenous injection, Pkr(IR-Ca/Pda-uPA)-cRGD considerably targets to thrombi selectively (targeting rate: 91%) and disintegrates in response to acidic thrombi to release IR-1048 dye and uPA for imaging and thrombolysis, respectively. Investigations indicate that Pkr(IR-Ca/Pda-uPA)-cRGD enabled real-time visualization of targeted thrombolysis using NIR-II imaging in DVT models, in which thrombi were eliminated (120 min after drug injection) without bleeding complications. This may be the first study using convenient NIR-II imaging for real-time visualization of targeted thrombolysis. It represents the precision medicine that enables rapid response to acquire instantaneous medical images and make necessary real-time adjustments to diagnostic and therapeutic protocols during treatment.
Collapse
Affiliation(s)
- Yaxin Ye
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences,
Southwest University, Chongqing 400715, China
| | - Zhechang Chen
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences,
Southwest University, Chongqing 400715, China
| | - Shengzhang Zhang
- Department of Cardiovascular Medicine,
Yueqing People's Hospital, Wenzhou 325699, China
| | - Paul Slezak
- Ludwig Boltzmann Institute for Traumatology,
AUVA Research Center, 1200 Vienna, Austria
| | - Fei Lu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences,
Southwest University, Chongqing 400715, China
| | - Ruiqi Xie
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences,
Southwest University, Chongqing 400715, China
- Ludwig Boltzmann Institute for Traumatology,
AUVA Research Center, 1200 Vienna, Austria
| | - Dongwon Lee
- Department of Bionanotechnology and Bioconvergence Engineering and Department of Polymer·Nano Science and Technology,
Jeonbuk National University, Jeonju, Chonbuk 54896, Republic of Korea
| | - Guangqian Lan
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences,
Southwest University, Chongqing 400715, China
| | - Enling Hu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences,
Southwest University, Chongqing 400715, China
- School of Fashion and Textiles,
The Hong Kong Polytechnic University, Hong Kong
| |
Collapse
|
6
|
Shen Y, Yu Y, Zhang X, Hu B, Wang N. Progress of nanomaterials in the treatment of thrombus. Drug Deliv Transl Res 2024; 14:1154-1172. [PMID: 38006448 DOI: 10.1007/s13346-023-01478-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2023] [Indexed: 11/27/2023]
Abstract
Thrombus has long been the major contributor of death and disability because it can cause adverse effects to varying degrees on the body, resulting in vascular blockage, embolism, heart valve deformation, widespread bleeding, etc. However, clinically, conventional thrombolytic drug treatments have hemorrhagic complication risks and easy to miss the best time of treatment window. Thus, it is an urgent need to investigate newly alternative treatment strategies that can reduce adverse effects and improve treatment effectiveness. Drugs based on nanomaterials act as a new biomedical strategy and promising tools, and have already been investigated for both diagnostic and therapeutic purposes in thrombus therapy. Recent studies have some encouraging progress. In the present review, we primarily concern with the latest developments in the areas of nanomedicines targeting thrombosis therapy. We present the thrombus' formation, characteristics, and biomarkers for diagnosis, overview recent emerging nanomedicine strategies for thrombus therapy, and focus on the future design directions, challenges, and prospects in the nanomedicine application in thrombus therapy.
Collapse
Affiliation(s)
- Yetong Shen
- Department of Biochemistry and Molecular Biology, China Medical University, No. 77 Puhe Road, Shenyang, 110122, China
- College of Life and Health Sciences, Northeastern University, Shenyang, 110167, China
| | - Yang Yu
- Department of Biochemistry and Molecular Biology, China Medical University, No. 77 Puhe Road, Shenyang, 110122, China
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Xin Zhang
- Department of Biochemistry and Molecular Biology, China Medical University, No. 77 Puhe Road, Shenyang, 110122, China
| | - Bo Hu
- Department of Biochemistry and Molecular Biology, China Medical University, No. 77 Puhe Road, Shenyang, 110122, China.
| | - Ning Wang
- Department of Biochemistry and Molecular Biology, China Medical University, No. 77 Puhe Road, Shenyang, 110122, China.
- Department of Forensic Medicine, China Medical University, No.77 Puhe Road, Shenyang, 110122, China.
| |
Collapse
|
7
|
Mao Y, Ren J, Yang L. Advances of nanomedicine in treatment of atherosclerosis and thrombosis. ENVIRONMENTAL RESEARCH 2023; 238:116637. [PMID: 37482129 DOI: 10.1016/j.envres.2023.116637] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/17/2023] [Accepted: 07/10/2023] [Indexed: 07/25/2023]
Abstract
Atherosclerosis (AS) is a chronic inflammatory vascular disease. Myocardial ischemia originated from AS is the main cause of cardiovascular diseases, one of the major factors contributing to the global disease burden. AS is typically quiescent until occurrence of plaque rupture and thrombosis, leading to acute coronary syndrome and sudden death. Currently, clinical diagnostic techniques suffer from major pitfalls including lack of accuracy and specificity, which makes it rather difficult for drugs to directly target plaques to achieve therapeutic effect. Therefore, how to accurately diagnose and effectively intervene vulnerable AS plaques to achieve accurate delivery of drugs has become an urgent and evolving clinical problem. With the rapid development of nanomedicine and nanomaterials, nanotechnology has shown unique advantages in monitoring vulnerable plaques and thrombus and improving drug efficacy. Recent studies have shown that application of nanoparticle drug delivery system can booster the safety and effectiveness of drug therapy, and molecular imaging technology and nanomedicine also exhibit high clinical application potentials in disease diagnosis. Therefore, nanotechnology provides another promising avenue for diagnosis and treatment of AS and thrombosis, and has shown excellent performance in the development of targeted drug therapy and biomaterials. In this review, the research progress, challenges and prospects of nanotechnology in AS and thrombosis are discussed, expecting to provide new ideas for the prevention, diagnosis and treatment of AS and thrombosis.
Collapse
Affiliation(s)
- Yu Mao
- Department of Cardiovascular Surgery, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| | - Lifang Yang
- Department of Anesthesiology, Xi'an Children Hospital, Xi'an, China.
| |
Collapse
|
8
|
Xiang S, Wang X, Peng S, Kang X, Wang J, Peng L, Ma X, Huang J, Sun X. Washout-Resistant, pH-Responsive Anti-TMV Nanoimmune Inducer Based on Cellulose Nanocrystals. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:16542-16553. [PMID: 37877141 DOI: 10.1021/acs.jafc.3c05733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
The application of antiplant virus agents on leaf surfaces faces challenges due to their vulnerability to wear, instability, and limited duration, which in turn jeopardizes plant health and yield. In recent years, high-aspect-ratio nanomaterials have gained prominence as powerful carriers for disease treatment, thanks to their exceptional penetrability and precise drug delivery capabilities. Here, we synthesized a pH-responsive nanoimmune inducer (CNC-AMO) with strong leaf adhesion through a Schiff base reaction, achieved by grafting amino-oligosaccharides (AMOs) on the surface of aldehyde-based CNC (CNC-CHO). Fourier transform infrared spectrometry, zeta potential, X-ray photoelectron spectroscopy, X-ray diffraction, transmission electron microscopy, atomic force microscopy, scanning electron microscopy, thermogravimetric analysis, and elemental analysis were used to characterize the CNC-AMO. The CNC-AMO displayed the capability for pH-responsive AMO release, showcasing its potential for targeted and controlled delivery. When applied to plants, the CNC-AMO exhibited impressive anti-TMV efficacy during a weeklong observation period. Meanwhile, the CNC-AMO exhibited remarkable adhesion and scouring resistance on the surfaces of the plant leaves. We strongly believe that the synergy of environmentally friendly synthetic materials, efficient plant virus control, and streamlined scalability positions CNC-AMOs as a promising pesticide for plant virus therapy.
Collapse
Affiliation(s)
- Shunyu Xiang
- College of Plant Protection, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China
| | - Xiaoyan Wang
- College of Plant Protection, Southwest University, Chongqing 400715, China
| | - Shiqi Peng
- College of Plant Protection, Southwest University, Chongqing 400715, China
| | - Xinke Kang
- School of Minerals Processing and Bioengineering, Central South University, Changsha 410083, China
| | - Jing Wang
- College of Plant Protection, Southwest University, Chongqing 400715, China
| | - Liyuan Peng
- College of Plant Protection, Southwest University, Chongqing 400715, China
| | - Xiaozhou Ma
- College of Plant Protection, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China
| | - Jin Huang
- School of Chemistry and Chemical Engineering, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China
| | - Xianchao Sun
- College of Plant Protection, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Soft-Matter Material Chemistry and Function Manufacturing, Southwest University, Chongqing 400715, China
| |
Collapse
|
9
|
Zhou S, Zhao W, Hu J, Mao C, Zhou M. Application of Nanotechnology in Thrombus Therapy. Adv Healthc Mater 2023; 12:e2202578. [PMID: 36507827 DOI: 10.1002/adhm.202202578] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/26/2022] [Indexed: 12/14/2022]
Abstract
A thrombus is a blood clot that forms in the lumen of an artery or vein, restricting blood flow and causing clinical symptoms. Thrombosis is associated with many life-threatening cardiovascular diseases. However, current clinical therapeutic technologies still have many problems in targeting, enrichment, penetration, and safety to meet the thrombosis treatment needs. Therefore, researchers devote themselves to developing nanosystems loaded with antithrombotic drugs to address this paradox in recent years. Herein, the existing thrombosis treatment technologies are first reviewed; and then, their advantages and disadvantages are outlined based on a brief discussion of thrombosis's definition and formation mechanism. Furthermore, the need and application cases for introducing nanotechnology are discussed, focusing on thrombus-specific targeted ligand modification technology and microenvironment-triggered responsive drug release technology. Then, nanomaterials that can be used to design antithrombotic nanotherapeutic systems are summarized. Moreover, a variety of drug delivery technologies driven by nanomotors in thrombosis therapy is also introduced. Last of all, a prospective discussion on the future development of nanotechnology for thrombosis therapy is highlighted.
Collapse
Affiliation(s)
- Shuyin Zhou
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China.,Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Wenbo Zhao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Jinglei Hu
- Kuang Yaming Honors School, Nanjing University, Nanjing, 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Min Zhou
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| |
Collapse
|
10
|
Nguyen AB, Iqbal O, Block RC, Mousa SA. Prevention and treatment of atherothrombosis: Potential impact of nanotechnology. Vascul Pharmacol 2023; 148:107127. [PMID: 36375733 DOI: 10.1016/j.vph.2022.107127] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/29/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Complications with atherosclerosis can often lead to fatal clot formation and blood vessel occlusion - also known as atherothrombosis. A key component to the development of atherosclerosis and atherothrombosis is the endothelium and its ability to regulate the balance between prothrombotic and antithrombotic activities. Endothelial surface glycocalyx has a critical role in maintenance of vascular integrity. The endothelial glycocalyx, nitric oxide, prostacyclins, heparan sulfate, thrombomodulin, and tissue factor pathway inhibitor all prevent thrombosis, while P-selectin, among many other factors, favors thrombosis. However, endothelial dysfunction gives rise to the acceleration of thrombotic development and eventually the requirement of antithrombotic therapy. Most FDA-approved anticoagulant and antiplatelet therapies today carry a side effect profile of major bleed. Within the past five years, several preclinical studies using different endothelial targets and nanotechnology as a drug delivery method have emerged to target the endothelium and to enhance current antithrombosis without increasing bleed risk. While clinical studies are required, this review illustrates the proof-of-concept of nanotechnology in promoting a greater safety and efficacy profile through multiple in vitro and in vivo studies.
Collapse
Affiliation(s)
- Anthony B Nguyen
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York 12144, United States of America
| | - Omer Iqbal
- Stritch School of Medicine, Loyola University, Chicago, IL, United States of America
| | - Robert C Block
- University of Rochester, 601 Elmwood Ave, Rochester, NY 14642, United States of America
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York 12144, United States of America.
| |
Collapse
|
11
|
Tapeinos C, Gao H, Bauleth-Ramos T, Santos HA. Progress in Stimuli-Responsive Biomaterials for Treating Cardiovascular and Cerebrovascular Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200291. [PMID: 35306751 DOI: 10.1002/smll.202200291] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/15/2022] [Indexed: 06/14/2023]
Abstract
Cardiovascular and cerebrovascular diseases (CCVDs) describe abnormal vascular system conditions affecting the brain and heart. Among these, ischemic heart disease and ischemic stroke are the leading causes of death worldwide, resulting in 16% and 11% of deaths globally. Although several therapeutic approaches are presented over the years, the continuously increasing mortality rates suggest the need for more advanced strategies for their treatment. One of these strategies lies in the use of stimuli-responsive biomaterials. These "smart" biomaterials can specifically target the diseased tissue, and after "reading" the altered environmental cues, they can respond by altering their physicochemical properties and/or their morphology. In this review, the progress in the field of stimuli-responsive biomaterials for CCVDs in the last five years, aiming at highlighting their potential as early-stage therapeutics in the preclinical scenery, is described.
Collapse
Affiliation(s)
- Christos Tapeinos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Han Gao
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Tomás Bauleth-Ramos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| |
Collapse
|
12
|
Liao J, Li Y, Luo Y, Meng S, Zhang C, Xiong L, Wang T, Lu Y. Recent Advances in Targeted Nanotherapies for Ischemic Stroke. Mol Pharm 2022; 19:3026-3041. [PMID: 35905397 DOI: 10.1021/acs.molpharmaceut.2c00383] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Ischemic stroke (IS) is a severe neurological disease caused by the narrowing or occlusion of cerebral blood vessels and is known for high morbidity, disability, and mortality rates. Clinically available treatments of stroke include the surgical removal of the thrombus and thrombolysis with tissue fibrinogen activator. Pharmaceuticals targeting IS are uncommon, and the development of new therapies is hindered by the low bioavailability and stability of many drugs. Nanomedicine provides new opportunities for the development of novel neuroprotective and thrombolytic strategies for the diagnosis and treatment of IS. Numerous nanotherapeutics with different physicochemical properties are currently being developed to facilitate drug delivery by accumulation and controlled release and to improve their restorative properties. In this review, we discuss recent developments in IS therapy, including assisted drug delivery and targeting, neuroprotection through regulation of the neuron environment, and sources of endogenous biomimetic specific targeting. In addition, we discuss the role and neurotoxic effects of inorganic metal nanoparticles in IS therapy. This study provides a theoretical basis for the utilization of nano-IS therapies that may contribute to the development of new strategies for a range of embolic diseases.
Collapse
Affiliation(s)
- Jun Liao
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Yi Li
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Yunchun Luo
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Sha Meng
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Chuan Zhang
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Liyan Xiong
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Tingfang Wang
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Ying Lu
- School of Pharmacy, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
13
|
Wang D, Wang X. Diosgenin and Its Analogs: Potential Protective Agents Against Atherosclerosis. Drug Des Devel Ther 2022; 16:2305-2323. [PMID: 35875677 PMCID: PMC9304635 DOI: 10.2147/dddt.s368836] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/09/2022] [Indexed: 11/23/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the artery wall associated with lipid metabolism imbalance and maladaptive immune response, which mediates most cardiovascular events. First-line drugs such as statins and antiplatelet drug aspirin have shown good effects against atherosclerosis but may lead to certain side effects. Thus, the development of new, safer, and less toxic agents for atherosclerosis is urgently needed. Diosgenin and its analogs have gained importance for their efficacy against life-threatening diseases, including cardiovascular, endocrine, nervous system diseases, and cancer. Diosgenin and its analogs are widely found in the rhizomes of Dioscore, Solanum, and other species and share similar chemical structures and pharmacological effects. Recent data suggested diosgenin plays an anti-atherosclerosis role through its anti-inflammatory, antioxidant, plasma cholesterol-lowering, anti-proliferation, and anti-thrombotic effects. However, a review of the effects of diosgenin and its natural structure analogs on AS is still lacking. This review summarizes the effects of diosgenin and its analogs on vascular endothelial dysfunction, vascular smooth muscle cell (VSMC) proliferation, migration and calcification, lipid metabolism, and inflammation, and provides a new overview of its anti-atherosclerosis mechanism. Besides, the structures, sources, safety, pharmacokinetic characteristics, and biological availability are introduced to reveal the limitations and challenges of current studies, hoping to provide a theoretical basis for the clinical application of diosgenin and its analogs and provide a new idea for developing new agents for atherosclerosis.
Collapse
Affiliation(s)
- Dan Wang
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai, People’s Republic of China
| | - Xiaolong Wang
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai, People’s Republic of China
- Correspondence: Xiaolong Wang, Tel +86 13501991450, Fax +86 21 51322445, Email
| |
Collapse
|
14
|
Advanced drug delivery system against ischemic stroke. J Control Release 2022; 344:173-201. [PMID: 35248645 DOI: 10.1016/j.jconrel.2022.02.036] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 02/06/2023]
|
15
|
|
16
|
Nguyen A, Böttger R, Li SD. Recent trends in bioresponsive linker technologies of Prodrug-Based Self-Assembling nanomaterials. Biomaterials 2021; 275:120955. [PMID: 34130143 DOI: 10.1016/j.biomaterials.2021.120955] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 05/19/2021] [Accepted: 05/29/2021] [Indexed: 12/15/2022]
Abstract
Prodrugs are designed to improve pharmaceutical properties of potent compounds and represent a central approach in drug development. The success of the prodrug strategy relies on incorporation of a reversible linkage facilitating controlled release of the parent drug. While prodrug approaches enhance pharmacokinetic properties over their parent drug, they still face challenges in absorption, distribution, metabolism, elimination, and toxicity (ADMET). Conjugating a drug to a carrier molecule such as a polymer can create an amphiphile that self-assembles into nanoparticles. These nanoparticles display prolonged blood circulation and passive targeting ability. Furthermore, the drug release can be tailored using a variety of linkers between the parent drug and the carrier molecule. In this review, we introduce the concept of self-assembling prodrugs and summarize different approaches for controlling the drug release with a focus on the linker technology. We also summarize recent clinical trials, discuss the emerging challenges, and provide our perspective on the utility and future potential of this technology.
Collapse
Affiliation(s)
- Anne Nguyen
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Roland Böttger
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, British Columbia, V6T 1Z3, Canada.
| |
Collapse
|
17
|
New Approaches in Nanomedicine for Ischemic Stroke. Pharmaceutics 2021; 13:pharmaceutics13050757. [PMID: 34065179 PMCID: PMC8161190 DOI: 10.3390/pharmaceutics13050757] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/20/2022] Open
Abstract
Ischemic stroke, caused by the interruption of blood flow to the brain and subsequent neuronal death, represents one of the main causes of disability in developed countries. Therapeutic methods such as recanalization approaches, neuroprotective drugs, or recovery strategies have been widely developed to improve the patient's outcome; however, important limitations such as a narrow therapeutic window, the ability to reach brain targets, or drug side effects constitute some of the main aspects that limit the clinical applicability of the current treatments. Nanotechnology has emerged as a promising tool to overcome many of these drug limitations and improve the efficacy of treatments for neurological diseases such as stroke. The use of nanoparticles as a contrast agent or as drug carriers to a specific target are some of the most common approaches developed in nanomedicine for stroke. Throughout this review, we have summarized our experience of using nanotechnology tools for the study of stroke and the search for novel therapies.
Collapse
|
18
|
Priya V, Viswanadh MK, Mehata AK, Jain D, Singh SK, Muthu MS. Targeted nanotherapeutics in the prophylaxis and treatment of thrombosis. Nanomedicine (Lond) 2021; 16:1153-1176. [PMID: 33973818 DOI: 10.2217/nnm-2021-0058] [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: 01/01/2023] Open
Abstract
Currently available anti-thrombotic therapy for the prophylaxis and treatment of arterial and venous thrombosis includes intravenous administration of anti-thrombotic drugs which lead to severe bleeding risks such as cerebral hemorrhage and stroke. Targeting approaches that utilize nanosystems to reach the thrombus sites are emerging to increase the local effect of anti-thrombotic drugs, as well as to decrease these severe bleeding complications by diminishing the systemic availability of these drugs. This review emphasizes the emerging targeted nanomedicines (liposomes, micelles, polymeric nanoparticles, material bases nanoparticles and other biological vectors) for the prophylaxis and treatment of thrombotic events as well as multifunctional nanomedicines for theranostic applications. Nanomedicine offers a promising platform for a smart, safe, and effective approach for the management of thrombosis.
Collapse
Affiliation(s)
- Vishnu Priya
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Matte Kasi Viswanadh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Abhishesh Kumar Mehata
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Dharmendra Jain
- Department of Cardiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Sanjeev K Singh
- Department of Physiology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Madaswamy S Muthu
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| |
Collapse
|
19
|
He H, Adili R, Liu L, Hong K, Holinstat M, Schwendeman A. Synthetic high-density lipoproteins loaded with an antiplatelet drug for efficient inhibition of thrombosis in mice. SCIENCE ADVANCES 2020; 6:6/49/eabd0130. [PMID: 33277254 PMCID: PMC7821904 DOI: 10.1126/sciadv.abd0130] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/21/2020] [Indexed: 05/12/2023]
Abstract
Antiplatelet agents offer a desirable approach to thrombosis prevention through the reduction of platelet reactivity. However, major bleeding events greatly attenuate the clinical outcomes of most antithrombotic agents. Therefore, the development of safer and more effective strategies to prevent vascular occlusion and avoid bleeding is urgently needed. A reconstituted nanoparticle, synthetic high-density lipoprotein (sHDL), which mimics the native HDL, has been established as clinically safe and is easily manufactured on a large scale. In this study, we propose that the delivery of the antiplatelet drug ML355, a selective inhibitor of 12(S)-lipoxygenase (12-LOX), by sHDL will efficiently inhibit thrombosis by targeting ML355 to the intended site of action, improving the pharmaceutical profile and harnessing the innate antithrombotic efficacy of the sHDL carrier. Our data show that ML355-sHDL exhibits more potent inhibition of thrombus formation in both small arterioles and larger arteries in mice without impairing the normal hemostasis in vivo.
Collapse
Affiliation(s)
- Hongliang He
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA
| | - Reheman Adili
- Department of Pharmacology, University of Michigan Medical School, 1150 W. Medical Center Dr., Room 2220D, Medical Sciences Research Building III, Ann Arbor, MI 48109, USA
| | - Lisha Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA
| | - Kristen Hong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan Medical School, 1150 W. Medical Center Dr., Room 2220D, Medical Sciences Research Building III, Ann Arbor, MI 48109, USA
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, 1150 W. Medical Center Dr., Room 2220D, Medical Sciences Research Building III, Ann Arbor, MI 48109, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA.
- Biointerfaces Institute, NCRC, 2800 Plymouth Rd., Ann Arbor, MI 48109, USA
| |
Collapse
|
20
|
Wei Z, Wang H, Xin G, Zeng Z, Li S, Ming Y, Zhang X, Xing Z, Li L, Li Y, Zhang B, Zhang J, Niu H, Huang W. A pH-Sensitive Prodrug Nanocarrier Based on Diosgenin for Doxorubicin Delivery to Efficiently Inhibit Tumor Metastasis. Int J Nanomedicine 2020; 15:6545-6560. [PMID: 32943867 PMCID: PMC7480473 DOI: 10.2147/ijn.s250549] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 07/17/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The metastasis, one of the biggest barriers in cancer therapy, is the leading cause of tumor deterioration and recurrence. The anti.-metastasis has been considered as a feasible strategy for clinical cancer management. It is well known that diosgenin could inhibit tumor metastasis and doxorubicin (DOX) could induce tumor apoptosis. However, their efficient delivery remains challenging. PURPOSE To address these issues, a novel pH-sensitive polymer-prodrug based on diosgenin nanoparticles (NPs) platform was developed to enhance the efficiency of DOX delivery (DOX/NPs) for synergistic therapy of cutaneous melanoma, the most lethal form of skin cancer with high malignancy, early metastasis and high mortality. METHODS AND RESULTS The inhibitory effect of DOX/NPs on tumor proliferation and migration was superior to that of NPs or free DOX. What is more, DOX/NPs could combine mitochondria-associated metastasis and apoptosis with unique internalization pathway of carrier to fight tumors. In addition, biodistribution experiments proved that DOX/NPs could efficiently accumulate in tumor sites through enhancing permeation and retention (EPR) effect compared with free DOX. Importantly, the data from in vivo experiment revealed that DOX/NPs without heart toxicity significantly inhibited tumor metastasis by exerting synergistic therapeutic effect, and reduced tumor volume and weight by inducing apoptosis. CONCLUSION The nanocarrier DOX/NPs with satisfying pharmaceutical characteristics based on the establishment of two different functional agents is a promising strategy for synergistically enhancing effects of cancer therapy.
Collapse
Affiliation(s)
- Zeliang Wei
- Laboratory of Ethnopharmacology, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Haibo Wang
- Textile Institute, College of Light Industry, Textile and Food Engineering, Sichuan University, Chengdu, People’s Republic of China
| | - Guang Xin
- Laboratory of Ethnopharmacology, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Zhi Zeng
- Laboratory of Ethnopharmacology, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Shiyi Li
- Laboratory of Ethnopharmacology, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Yue Ming
- Laboratory of Ethnopharmacology, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Xiaoyu Zhang
- Laboratory of Ethnopharmacology, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Zhihua Xing
- Laboratory of Ethnopharmacology, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Li Li
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Youping Li
- Laboratory of Ethnopharmacology, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Boli Zhang
- Laboratory of Ethnopharmacology, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
| | - Junhua Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
| | - Hai Niu
- Laboratory of Ethnopharmacology, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- College of Mathematics, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Wen Huang
- Laboratory of Ethnopharmacology, West China Medical School, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
21
|
Parama D, Boruah M, Yachna K, Rana V, Banik K, Harsha C, Thakur KK, Dutta U, Arya A, Mao X, Ahn KS, Kunnumakkara AB. Diosgenin, a steroidal saponin, and its analogs: Effective therapies against different chronic diseases. Life Sci 2020; 260:118182. [PMID: 32781063 DOI: 10.1016/j.lfs.2020.118182] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Chronic diseases are a major cause of mortality worldwide, and despite the recent development in treatment modalities, synthetic drugs have continued to show toxic side effects and development of chemoresistance, thereby limiting their application. The use of phytochemicals has gained attention as they show minimal side effects. Diosgenin is one such phytochemical which has gained importance for its efficacy against the life-threatening diseases, such as cardiovascular diseases, cancer, nervous system disorders, asthma, arthritis, diabetes, and many more. AIM To evaluate the literature available on the potential of diosgenin and its analogs in modulating different molecular targets leading to the prevention and treatment of chronic diseases. METHOD A detailed literature search has been carried out on PubMed for gathering information related to the sources, biosynthesis, physicochemical properties, biological activities, pharmacokinetics, bioavailability and toxicity of diosgenin and its analogs. KEY FINDINGS The literature search resulted in many in vitro, in vivo and clinical trials that reported the efficacy of diosgenin and its analogs in modulating important molecular targets and signaling pathways such as PI3K/AKT/mTOR, JAK/STAT, NF-κB, MAPK, etc., which play a crucial role in the development of most of the diseases. Reports have also revealed the safety of the compound and the adaptation of nanotechnological approaches for enhancing its bioavailability and pharmacokinetic properties. SIGNIFICANCE Thus, the review summarizes the efficacy of diosgenin and its analogs for developing as a potent drug against several chronic diseases.
Collapse
Affiliation(s)
- Dey Parama
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Monikongkona Boruah
- Cell and Molecular Biology Lab, Department of Zoology, Cotton University, Guwahati, Assam 781001, India
| | - Kumari Yachna
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Varsha Rana
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Kishore Banik
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Choudhary Harsha
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Krishan Kumar Thakur
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Uma Dutta
- Cell and Molecular Biology Lab, Department of Zoology, Cotton University, Guwahati, Assam 781001, India
| | - Aditya Arya
- Department of Pharmacology and Therapeutics, School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Xinliang Mao
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, China; Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India.
| |
Collapse
|
22
|
Ming Y, Xin G, Ji B, Ji C, Wei Z, Zhang B, Zhang J, Yu K, Zhang X, Li S, Li Y, Xing Z, Niu H, Huang W. Entecavir as a P2X7R antagonist ameliorates platelet activation and thrombus formation. J Pharmacol Sci 2020; 144:43-51. [PMID: 32653340 DOI: 10.1016/j.jphs.2020.07.002] [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: 11/06/2019] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 02/08/2023] Open
Abstract
Platelet activation is the primary cause of thrombosis. The P2X7 receptor (P2X7R) is a therapeutic target of thrombosis. However, it is still unknown whether P2X7R activation affects platelet thrombus. Our molecular docking results showed that entecavir as a P2X7R antagonist interacted perfectly with the human P2X7R (hP2X7R) in silico simulation studies. Furthermore, our experimental data revealed that entecavir could act as a P2X7R antagonist to exert cytoprotective effects against platelet activation via protecting mitochondrial function, improving lipid peroxidation and increasing antioxidant activity. Correlated with this, entecavir inhibited platelet aggregation, dense-granule secretion, P-selectin expression, integrin activation and Ca2+ increase. In experimental mouse model, entecavir could significantly inhibit arteriovenous thrombosis and prolong the bleeding time. Furthermore, we found that entecavir had no significant effect on prothrombin time (PT), activated partial thrombin time (APTT), thrombin time (TT), fibrinogen (FIB), mean platelet volume (MPV) and platelet counts (PLT). This study demonstrates that entecavir markedly prevents platelet activation and thrombosis through inhibiting P2X7R without affecting coagulation system. Therefore, entecavir may be a potential candidate for treating thrombosis disease.
Collapse
Affiliation(s)
- Yue Ming
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guang Xin
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Beihong Ji
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Pittsburgh, Pennsylvania, United States
| | - Chengji Ji
- Clinical Laboratory, Hospital of University of Electronic Science and Technology of China and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Zeliang Wei
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Boli Zhang
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junhua Zhang
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Kui Yu
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoyu Zhang
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shiyi Li
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Youping Li
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhihua Xing
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hai Niu
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Wen Huang
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
23
|
Emerging nanotherapeutics for antithrombotic treatment. Biomaterials 2020; 255:120200. [PMID: 32563945 DOI: 10.1016/j.biomaterials.2020.120200] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 06/03/2020] [Accepted: 06/09/2020] [Indexed: 12/20/2022]
Abstract
Thrombus causes insufficient blood flow and ischemia damages to brain and heart, leading to life-threatening cardio-cerebrovascular diseases. Development of efficient antithrombotic strategies has long been a high priority, owing to the high morbidity and mortality of thrombotic diseases. With the rapid development of biomedical nanotechnology in diagnosis and treatment of thrombotic disorder, remarkable progresses have been made in antithrombotic nanomedicines in recent years. Herein, we outline the recent advances in this field at the intersection of thrombus theranostics and biomedical nanotechnology. First, thrombus diagnosis techniques based on biomedical nanotechnology are presented. Then, emerging antithrombotic nanotherapeutics are overviewed, including thrombus-targeting strategies, thrombus stimuli-responsive nanosystems and phase transition-driven nanotherapeutics. Furthermore, multifunctional nanosystems for combination theranostics of thrombotic diseases are discussed. Finally, the design considerations, advantages and challenges of these biomedical nanotechnology-driven therapeutics in clinical translation are highlighted.
Collapse
|
24
|
Xin G, Ming Y, Ji C, Wei Z, Li S, Morris-Natschke SL, Zhang X, Yu K, Li Y, Zhang B, Zhang J, Xing Z, He Y, Chen Z, Yang X, Niu H, Lee KH, Huang W. Novel potent antiplatelet thrombotic agent derived from biguanide for ischemic stroke. Eur J Med Chem 2020; 200:112462. [PMID: 32464472 DOI: 10.1016/j.ejmech.2020.112462] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 02/08/2023]
Abstract
Platelet thrombosis is the main pathogeny resulting in the low curability of ischemic stroke, a leading cause of mortality and disability worldwide. Metformin, a biguanide derivative that is the first-line oral medicine for type 2 diabetes, alleviates the severity of ischemic stroke in diabetic patients and suppresses platelet activation in experimental animal model. However, the clinical implementation of commercial biguanide analogs for stroke related to platelet thrombosis remains challenging due to its weak potency, poor pharmacokinetic characteristics and possible hypoglycemia. Here, twenty-three biguanide derivatives were designed and synthesized based on the principles of bioisosteres. These derivatives were evaluated for the activity of antiplatelet thrombosis in vivo. We found that N-trifluoromethanesulfonyl biguanide derivative, compound b10, uniquely prevented cerebral infarction as well as neuronal function injury, and significantly decrease the mortality rate of ischemic stroke in the middle cerebral artery occlusion mice without significant side effects. We verified that b10 directly inhibited platelets thrombus formation and decreased the compactness of stroke thrombi. Particularly, b10 exhibited good potency to inhibit human platelet activation including platelet aggregation, adhesion, pseudopodia formation, integrin GPIIb/IIIa activation, CD62P expression and clot retraction. Meanwhile, the pharmacokinetics assessment showed that b10 had satisfying pharmacological characteristics including a longer duration and a higher oral absorption ratio than its parent compound. In addition, b10 remarkably ameliorated not only stroke related to platelet thrombosis but also carotid artery thrombus formation. It is concluded that the novel potent antiplatelet thrombotic agent derived from biguanide is a promising candidate for stroke treatment.
Collapse
Affiliation(s)
- Guang Xin
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yue Ming
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chengjie Ji
- Clinical Laboratory, Hospital of University of Electronic Science and Technology of China, Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Zeliang Wei
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shiyi Li
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Susan L Morris-Natschke
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Xiaoyu Zhang
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kui Yu
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Youping Li
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Boli Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junhua Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhihua Xing
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yarong He
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhen Chen
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xijing Yang
- Animal Experiment Center, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hai Niu
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China; College of Mathematics, Sichuan University, Chengdu, Sichuan, China.
| | - Kuo-Hsiung Lee
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| | - Wen Huang
- Laboratory of Ethnopharmacology, West China School of Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
25
|
Li J, Yang X, Li X, Zhang Z, Wei Z, Xing Z, Deng S, Duan F. Okra polysaccharides/gelatin complex coacervate as pH-responsive and intestine-targeting delivery protects isoquercitin bioactivity. Int J Biol Macromol 2020; 159:487-496. [PMID: 32422271 DOI: 10.1016/j.ijbiomac.2020.05.067] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 04/30/2020] [Accepted: 05/10/2020] [Indexed: 02/08/2023]
Abstract
Okra polysaccharides (OPs) belong to RG I pectin branched with neutral saccharide side chains, which possesses distinctive structure and physicochemical properties from the commonly used HG pectin. Until now, the application of RG I pectin as wall material of microcapsule remains unclear. Here, we obtained OPs/gelatin complex coacervate at the maximum yield of 86.8% (pH 3.5, gelatin/OPs ratio 9:1 and 2% (w/v) total polymer concentration) by response surface methodology. Isoquercitin (IQ)-loaded OPs/gelatin complex coacervate (OGIQ) showed porous spongy-like surface structure with average particle size, encapsulation efficiency and surface porosity at 334 nm, 81.6% and 31.9%, respectively. OGIQ was found to be pH-responsive and intestine-targeting. The IQ-release rate of OGIQ was assayed to be 89.4% in intestine fluid and below 2% in acidic and simulated gastric digestion, respectively. Accordingly, embedding in OGIQ protected IQ in digestion and improved its postdigestive α-glucosidase inhibitory rate by 88.7%. The differential scanning calorimetry curves showed that OGIQ effectively prevented IQ from thermal decomposition. The XRD, FT-IR and CD spectra indicated that IQ was embedded in OGIQ in amorphous state by hydrogen bonds and electrostatic interaction. Compared with HG, the neutral saccharide side chains of OPs could induce different secondary conformation change of gelatin during complex coacervation.
Collapse
Affiliation(s)
- Jingwen Li
- Department of Food Engineering, College of Biomass Science and Engineering & Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, PR China
| | - Xiaoran Yang
- Department of Food Engineering, College of Biomass Science and Engineering & Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, PR China
| | - Xiao Li
- Department of Food Engineering, College of Biomass Science and Engineering & Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, PR China
| | - Zihan Zhang
- Department of Food Engineering, College of Biomass Science and Engineering & Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, PR China
| | - Zeliang Wei
- Laboratory of Ethnopharmacology, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China
| | - Zhihua Xing
- Laboratory of Ethnopharmacology, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China
| | - Sha Deng
- Department of Food Engineering, College of Biomass Science and Engineering & Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, PR China
| | - Feixia Duan
- Department of Food Engineering, College of Biomass Science and Engineering & Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, PR China.
| |
Collapse
|
26
|
Cai B, Zhang Y, Wang Z, Xu D, Jia Y, Guan Y, Liao A, Liu G, Chun C, Li J. Therapeutic Potential of Diosgenin and Its Major Derivatives against Neurological Diseases: Recent Advances. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:3153082. [PMID: 32215172 PMCID: PMC7079249 DOI: 10.1155/2020/3153082] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/16/2019] [Accepted: 12/30/2019] [Indexed: 12/15/2022]
Abstract
Diosgenin (DG), a well-known steroidal sapogenin, is present abundantly in medicinal herbs such as Dioscorea rhizome, Dioscorea villosa, Trigonella foenum-graecum, Smilax China, and Rhizoma polgonati. DG is utilized as a major starting material for the production of steroidal drugs in the pharmaceutical industry. Due to its wide range of pharmacological activities and medicinal properties, it has been used in the treatment of cancers, hyperlipidemia, inflammation, and infections. Numerous studies have reported that DG is useful in the prevention and treatment of neurological diseases. Its therapeutic mechanisms are based on the mediation of different signaling pathways, and targeting these pathways might lead to the development of effective therapeutic agents for neurological diseases. The present review mainly summarizes recent progress using DG and its derivatives as therapeutic agents for multiple neurological disorders along with their various mechanisms in the central nervous system. In particular, those related to therapeutic efficacy for Parkinson's disease, Alzheimer's disease, brain injury, neuroinflammation, and ischemia are discussed. This review article also critically evaluates existing limitations associated with the solubility and bioavailability of DG and discusses imperatives for translational clinical research. It briefly recapitulates recent advances in structural modification and novel formulations to increase the therapeutic efficacy and brain levels of DG. In the present review, databases of PubMed, Web of Science, and Scopus were used for studies of DG and its derivatives in the treatment of central nervous system diseases published in English until December 10, 2019. Three independent researchers examined articles for eligibility. A total of 150 articles were screened from the above scientific literature databases. Finally, a total of 46 articles were extracted and included in this review. Keywords related to glioma, ischemia, memory, aging, cognitive impairment, Alzheimer, Parkinson, and neurodegenerative disorders were searched in the databases based on DG and its derivatives.
Collapse
Affiliation(s)
- Bangrong Cai
- Henan Research Center for Special Processing Technology of Chinese Medicine, School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Ying Zhang
- Department of Biochemistry, Department of Biomedical Sciences, Research Center for Aging and Geriatrics, Research Institute of Medical Sciences, Chonnam National University Medical School, Gwangju 501-190, Republic of Korea
| | - Zengtao Wang
- Department of Medicinal Chemistry, College of Pharmacy JiangXi University of Traditional Chinese Medicine, Nanchang 330004, China
| | - Dujuan Xu
- Henan Research Center for Special Processing Technology of Chinese Medicine, School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yongyan Jia
- Henan Research Center for Special Processing Technology of Chinese Medicine, School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yanbin Guan
- Henan Research Center for Special Processing Technology of Chinese Medicine, School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Aimei Liao
- College of Biological Engineering, Henan University of Technology, Zhengzhou 450001, China
| | - Gaizhi Liu
- Henan Research Center for Special Processing Technology of Chinese Medicine, School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - ChangJu Chun
- Research Institute of Drug Development, College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Jiansheng Li
- Collaborative Innovation Center for Respiratory Disease Diagnosis and Treatment and Chinese Medicine Development of Henan Province, Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Henan University of Chinese Medicine, China
| |
Collapse
|
27
|
Ilkar Erdagi S, Ngwabebhoh FA, Yildiz U. Pickering stabilized nanocellulose-alginate: A diosgenin-mediated delivery of quinalizarin as a potent cyto-inhibitor in human lung/breast cancer cell lines. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 109:110621. [PMID: 32228903 DOI: 10.1016/j.msec.2019.110621] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/30/2019] [Accepted: 12/31/2019] [Indexed: 01/29/2023]
Abstract
The current study explores the facile fabrication of multilayer self-assembled electrostatic oil-in-water Pickering emulsions (PEs) using quaternized nanocellulose (Q-NC) and diosgenin-conjugate alginate (DGN-ALG) particles as stabilizers to form hydrocolloid nanocarriers. The conditions of formulation such as storage time, pH, temperature and salt effect on the emulsion stability were evaluated. The results deduced showed good emulsion droplet stability over a period of 30 days. Morphological analysis revealed the hydrodynamic sizes of the PE droplets to be spherically shaped with an average diameter of 150 ± 3.51 nm. Creaming index, wettability and critical aggregation concentrations (CAC) as well as chemical characterization of the PEs were examined. In vitro release kinetics of encapsulated quinalizarin as a model drug was investigated with a determined cumulative drug release (CDR) of 89 ± 1.21% in simulated pH blood medium of pH 7.4. In addition, cellular internalization of the PEs was studied via confocal microscopy imaging and showed high cellular uptake. Also, evaluated in vitro cytotoxicity by MTT assay demonstrated excellent anticancer activity in human lung (A549) and breast (MCF-7) cancer cell lines.
Collapse
Affiliation(s)
| | | | - Ufuk Yildiz
- Department of Chemistry, Kocaeli University, 41380 Kocaeli, Turkey
| |
Collapse
|
28
|
Nguyen A, Ando H, Böttger R, DurgaRao Viswanadham KK, Rouhollahi E, Ishida T, Li SD. Utilization of click chemistry to study the effect of poly(ethylene)glycol molecular weight on the self-assembly of PEGylated gambogic acid nanoparticles for the treatment of rheumatoid arthritis. Biomater Sci 2020; 8:4626-4637. [DOI: 10.1039/d0bm00711k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Click chemistry was used to study the effect of varied PEG molecular weights on the self-assembly of PEG-gambogic acid (GA) conjugates into nanoparticles.
Collapse
Affiliation(s)
- Anne Nguyen
- Faculty of Pharmaceutical Sciences
- University of British Columbia
- Vancouver
- Canada
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics
- Subdivision of Biopharmaceutical Sciences
- Institute of Health Biosciences
- The University of Tokushima
- Tokushima
| | - Roland Böttger
- Faculty of Pharmaceutical Sciences
- University of British Columbia
- Vancouver
- Canada
| | | | - Elham Rouhollahi
- Faculty of Pharmaceutical Sciences
- University of British Columbia
- Vancouver
- Canada
| | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics
- Subdivision of Biopharmaceutical Sciences
- Institute of Health Biosciences
- The University of Tokushima
- Tokushima
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences
- University of British Columbia
- Vancouver
- Canada
| |
Collapse
|
29
|
Cai D, Qi J, Yang Y, Zhang W, Zhou F, Jia X, Guo W, Huang X, Gao F, Chen H, Li T, Li G, Wang P, Zhang Y, Lei H. Design, Synthesis and Biological Evaluation of Diosgenin-Amino Acid Derivatives with Dual Functions of Neuroprotection and Angiogenesis. Molecules 2019; 24:molecules24224025. [PMID: 31703284 PMCID: PMC6891328 DOI: 10.3390/molecules24224025] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 12/20/2022] Open
Abstract
Diosgenin, a natural product with steroidal structure, has a wide range of clinical applications in China. It also shows great potential in the treatment of blood clots and nerve damage. To enhance the bioavailability as well as efficacy of diosgenin, eighteen diosgenin-amino acid derivatives were designed and synthesized. The neuroprotective effects of these compounds were evaluated by SH-SY5Y cell line and the biosafety was evaluated by H9c2 cell line. The results displayed that part of the derivatives' activities (EC50 < 20 μM) were higher than positive control edaravone (EC50 = 21.60 ± 3.04 μM), among which, DG-15 (EC50 = 6.86 ± 0.69 μM) exhibited the best neuroprotection. Meanwhile, biosafety evaluation showed that DG-15 had no cytotoxicity on H9c2 cell lines. Interestingly, combined neuroprotective and cytotoxic results, part of the derivatives without their protecting group were superior to compounds with protecting group. Subsequently, Giemsa staining and DAPI (4',6-diamidino-2-phenylindole) staining indicated that DG-15 had a protective effect on damaged SH-SY5Y cells by reducing apoptosis. Moreover, DG-15 showed a higher role in promoting angiogenesis at high concentrations (4 mg/mL) on the chorioallantoic membrane model. This finding displayed that DG-15 had dual functions of neuroprotection and angiogenesis, which provided further insight into designing agent for the application in treatment of ischemic stroke.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Yuzhong Zhang
- Correspondence: (Y.Z.); (H.L.); Tel.: +86-10-8473-8645 (H.L.)
| | - Haimin Lei
- Correspondence: (Y.Z.); (H.L.); Tel.: +86-10-8473-8645 (H.L.)
| |
Collapse
|
30
|
Huang H, Nie C, Qin X, Zhou J, Zhang L. Diosgenin inhibits the epithelial-mesenchymal transition initiation in osteosarcoma cells via the p38MAPK signaling pathway. Oncol Lett 2019; 18:4278-4287. [PMID: 31579425 DOI: 10.3892/ol.2019.10780] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 06/13/2019] [Indexed: 01/01/2023] Open
Abstract
Diosgenin is an important basic raw material for the production of steroid hormone drugs. It can be isolated and purified from a variety of traditional Chinese medicines or plants. Modern molecular biological studies have shown that diosgenin inhibits various tumor cells migration and invasion ability to varying degrees in vitro and in vivo. The aim of the present study was to observe the inhibitory effects of diosgenin on the invasive and metastatic capabilities of osteosarcoma cells and to determine the association between the effects of diosgenin on the epithelial-mesenchymal transition (EMT). Wound healing and Transwell assays were used to observe the inhibitory effects of diosgenin on the invasion and migration of two osteosarcoma cell lines. Immunofluorescence was used to observe changes in transforming growth factor β1 (TGF-β1) protein expression levels in the osteosarcoma cells following drug administration. EMT-associated proteins, including TGFβ1, E-cadherin and vimentin were detected by western blotting, which demonstrated that the drug may inhibit the initiation of EMT in osteosarcoma cells. Western blot analysis of the expression of all the proteins in the mitogen-activated protein kinase (MAPK) pathway demonstrated that the drug inhibited the MAPK signaling pathway. The primary mechanism of action of diosgenin was the inhibition of the phosphorylated p38 (pP38) protein. Through a combination of inhibitors of the p38MAPK signaling pathway and detection of the downstream EMT marker protein E-cadherin by quantitative PCR, pP38 was confirmed to be a target of diosgenin in the inhibition of EMT in the osteosarcoma cells via the MAPK molecular signaling pathway. Diosgenin may exhibit utility as an auxiliary drug for the clinical reduction of metastasis in patients with osteosarcoma.
Collapse
Affiliation(s)
- Huaming Huang
- Department of Research Office, Jiangsu Health Vocational College, Nanjing, Jiangsu 211800, P.R. China.,Department of Orthopedics, Xishan People's Hospital of Wuxi, Wuxi, Jiangsu 214015, P.R. China
| | - Chao Nie
- Department of Research Office, Jiangsu Health Vocational College, Nanjing, Jiangsu 211800, P.R. China
| | - Xiaokang Qin
- Jiangsu KeyGEN BioTECH Co., Ltd., Nanjing, Jiangsu 211100, P.R. China
| | - Jie Zhou
- Department of Research Office, Jiangsu Health Vocational College, Nanjing, Jiangsu 211800, P.R. China
| | - Lei Zhang
- Department of Research Office, Jiangsu Health Vocational College, Nanjing, Jiangsu 211800, P.R. China
| |
Collapse
|
31
|
Zhuang M, Xin G, Wei Z, Li S, Xing Z, Ji C, Du J, Niu H, Huang W. Dihydrodiosgenin inhibits endothelial cell-derived factor VIII and platelet-mediated hepatocellular carcinoma metastasis. Cancer Manag Res 2019; 11:4871-4882. [PMID: 31239763 PMCID: PMC6554527 DOI: 10.2147/cmar.s202225] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/20/2019] [Indexed: 02/05/2023] Open
Abstract
Background: Our previous studies have demonstrated that diosgenin and diosgenin derivatives exhibit excellent antithrombotic activity via regulating platelet function and coagulation factor level. Platelets and blood coagulation system are highly associated with tumor hematogenous metastasis. Therefore, the purpose of this study was to evaluate whether dihydrodiosgenin (dydio) mediated-platelet inhibition or coagulation factor level modulation is involved in hepatocellular carcinoma cell (HCC) metastasis. Methods: Cell viability was examined by MTT and colony formation assays. Platelet aggregation text and morphology were used to assess dydio's role on tumor cell-induced platelet activation (TCIPA). Scratch assay, adhesion assay and Western blot were used to evaluate dydio's role on platelet-mediated metastasis. Western blot and fluorescence detection were performed to clarify dydio's role on endothelial cell (EC) function. The mice lung metastasis model was constructed to investigated dydio's function on coagulation factor and platelet-mediated metastasis. Results: This study found that pretreatment with dydio caused a significant inhibition of TCIPA. Platelets exposed to dydio significantly inhibited their adhesion to tumor cells, meanwhile, releasates of platelets that pretreated with dydio led to diminished cancer cell proliferation and migration along with the increase of epithelial markers E-cadherin and loss of mesenchymal phenotype. Additionally, ECs pretreated with dydio suppressed factor VIII (FVIII) level which in turn restrained the activation of platelets and the adhesion of cancer cells or platelets to ECs. Interestingly, our study demonstrated that FVIII could promote HCC proliferation. In vivo study revealed that mice intragastrical (i.g.) administration with dydio significantly inhibited the lung metastasis of hepal-6 cells which is highly correlated with the altered platelet function and coagulation level. Conclusion: Taken together, these results demonstrated that dydio altered platelet function and coagulation FVIII level, resulting in decreased metastatic potential of HCC. Thus, our study reveals that dydio exerts novel mechanisms of antitumor action beside its direct antitumor activity.
Collapse
Affiliation(s)
- Manjiao Zhuang
- Laboratory of Ethnopharmacology, West China School of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Guang Xin
- Laboratory of Ethnopharmacology, West China School of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Zeliang Wei
- Laboratory of Ethnopharmacology, West China School of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Shiyi Li
- Laboratory of Ethnopharmacology, West China School of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Zhihua Xing
- Laboratory of Ethnopharmacology, West China School of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Chengjie Ji
- Laboratory of Ethnopharmacology, West China School of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, Chengdu, Sichuan, People's Republic of China
| | - Junrong Du
- Laboratory of Ethnopharmacology, West China School of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Hai Niu
- Laboratory of Ethnopharmacology, West China School of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
- College of Mathematics, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| | - Wen Huang
- Laboratory of Ethnopharmacology, West China School of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, People’s Republic of China
| |
Collapse
|
32
|
Hajizadeh MR, Parvaz N, Barani M, Khoshdel A, Fahmidehkar MA, Mahmoodi M, Torkzadeh-Mahani M. Diosgenin-loaded niosome as an effective phytochemical nanocarrier: physicochemical characterization, loading efficiency, and cytotoxicity assay. ACTA ACUST UNITED AC 2019; 27:329-339. [PMID: 31134490 DOI: 10.1007/s40199-019-00277-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/22/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The use of phytochemicals to prevent or suppress tumours is known as chemoprevention. Numerous plant-derived agents have been reported to have anticancer potentials. As one such anticancer phytochemical, diosgenin has several applications which are nevertheless limited due to its low solubility in water. METHODS We loaded diosgenin into niosome to increase its solubility and hence efficiency. Diosgenin-niosome (diosgenin loaded into niosome) was prepared by thin-film hydration method and characterised by optical microscopy, dynamic light scattering (DLS), scanning electron microscopy (SEM), and UV-visible spectrophotometry. Also, loading efficiency, in vitro drug release, and cytotoxicity assay were performed on HepG2 cell line. RESULTS AND DISCUSSION Diosgenin-niosome has a nanometric size with a normal size distribution and spherical morphology. The loading efficiency of diosgenin was about 89% with a sustainable and controllable release rate. Finally, the viability of free diosgenin was 61.25%, and after loading into niosomes, it was improved to 28.32%. CONCLUSION The results demonstrated that niosomes increase the solubility of naturally derived hydrophobic chemicals and thus enhance their anticancer effect. Graphical abstract.
Collapse
Affiliation(s)
- Mohammad Reza Hajizadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Najmeh Parvaz
- Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mahmood Barani
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Alireza Khoshdel
- Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Ali Fahmidehkar
- Research Center of Advanced Technologies in Medicine, Torbat Heydariyeh University of Medical Sciences, Torbat Heydariyeh, Iran
| | - Mehdi Mahmoodi
- Department of Clinical Biochemistry, Afzalipoor Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Masoud Torkzadeh-Mahani
- Department of Biotechnology, Institute of Science, High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran.
| |
Collapse
|
33
|
Abstract
The effects of diosgenin are discussed with respect to endothelial dysfunction, lipid profile, macrophage foam cell formation, VSMC viability, thrombosis and inflammation during the formation of atherosclerosis.
Collapse
Affiliation(s)
- Fang-Chun Wu
- College of Food and Bioengineering
- South China University of Technology
- Guangzhou
- China
| | - Jian-Guo Jiang
- College of Food and Bioengineering
- South China University of Technology
- Guangzhou
- China
| |
Collapse
|
34
|
Ilkar Erdagi S, Yildiz U. Diosgenin-conjugated PCL–MPEG polymeric nanoparticles for the co-delivery of anticancer drugs: design, optimization, in vitro drug release and evaluation of anticancer activity. NEW J CHEM 2019. [DOI: 10.1039/c9nj00659a] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study, a polymeric nanoparticle-mediated dual anti-cancer drug delivery system was designed and developed.
Collapse
Affiliation(s)
| | - Ufuk Yildiz
- Department of Chemistry, Kocaeli University
- Kocaeli
- Turkey
| |
Collapse
|
35
|
Nanobiotechnology medical applications: Overcoming challenges through innovation. THE EUROBIOTECH JOURNAL 2018. [DOI: 10.2478/ebtj-2018-0019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Abstract
Biomedical Nanotechnology (BNT) has rapidly become a revolutionary force that is driving innovation in the medical field. BNT is a subclass of nanotechnology (NT), and often operates in cohort with other subclasses, such as mechanical or electrical NT for the development of diagnostic assays, therapeutic implants, nano-scale imaging systems, and medical machinery. BNT is generating solutions to many conventional challenges through the development of enhanced therapeutic delivery systems, diagnostic techniques, and theranostic therapies. Therapeutically, BNT has generated many novel nanocarriers (NCs) that each express specifically designed physiochemical properties that optimize their desired pharmacokinetic profile. NCs are also being integrated into nanoscale platforms that further enhance their delivery by controlling and prolonging their release profile. Nano-platforms are also proving to be highly efficient in tissue regeneration when combined with the appropriate growth factors. Regarding diagnostics, NCs are being designed to perform targeted delivery of luminescent tags and contrast agents that enhance the NC -aided imaging capabilities and resulting diagnostic accuracy of the presence of diseased cells. This technology has also been advancing the ability for surgeons to practice true precision surgical techniques. Incorporating therapeutic and diagnostic NC-components within a single NC can facilitate both functions, referred to as theranostics, which facilitates real-time in vivo tracking and observation of drug release events via enhanced imaging. Additionally, stimuli-responsive theranostic NCs are quickly developing as vectors for tumor ablation therapies by providing a model that facilitates the location of cancer cells for the application of an external stimulus. Overall, BNT is an interdisciplinary approach towards health care, and has the potential to significantly improve the quality of life for humanity by significantly decreasing the treatment burden for patients, and by providing non-invasive therapeutics that confer enhanced therapeutic efficiency and safety
Collapse
|