1
|
Gong Z, Zhou D, Wu D, Han Y, Yu H, Shen H, Feng W, Hou L, Chen Y, Xu T. Challenges and material innovations in drug delivery to central nervous system tumors. Biomaterials 2025; 319:123180. [PMID: 39985979 DOI: 10.1016/j.biomaterials.2025.123180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 01/28/2025] [Accepted: 02/08/2025] [Indexed: 02/24/2025]
Abstract
Central nervous system (CNS) tumors, encompassing a diverse array of neoplasms in the brain and spinal cord, pose significant therapeutic challenges due to their intricate anatomy and the protective presence of the blood-brain barrier (BBB). The primary treatment obstacle is the effective delivery of therapeutics to the tumor site, which is hindered by multiple physiological, biological, and technical barriers, including the BBB. This comprehensive review highlights recent advancements in material science and nanotechnology aimed at surmounting these delivery challenges, with a focus on the development and application of nanomaterials. Nanomaterials emerge as potent tools in designing innovative drug delivery systems that demonstrate the potential to overcome the limitations posed by CNS tumors. The review delves into various strategies, including the use of lipid nanoparticles, polymeric nanoparticles, and inorganic nanoparticles, all of which are engineered to enhance drug stability, BBB penetration, and targeted tumor delivery. Additionally, this review highlights the burgeoning role of theranostic nanoparticles, integrating therapeutic and diagnostic functionalities to optimize treatment efficacy. The exploration extends to biocompatible materials like biodegradable polymers, liposomes, and advanced material-integrated delivery systems such as implantable drug-eluting devices and microfabricated devices. Despite promising preclinical results, the translation of these material-based strategies into clinical practice necessitates further research and optimization.
Collapse
Affiliation(s)
- Zhenyu Gong
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China; Department of Neurosurgery, Klinikum rechts der Isar, Technical University of Munich, Munich, 81675, Germany
| | - Dairan Zhou
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Dejun Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, 230601, PR China
| | - Yaguang Han
- Department of Orthopedics, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Hao Yu
- National Engineering Research Center of Ophthalmology and Optometry, School of Ophthalmology & Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, PR China
| | - Haotian Shen
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Lijun Hou
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| | - Tao Xu
- Department of Neurosurgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China.
| |
Collapse
|
2
|
Wen Y, Su Z, Zhu H, Liu M, Li Z, Zhang S, Cai S, Tang J, Ding H, Zeng H. Mannitol-facilitated entry of vancomycin into the central nervous system inhibits neuroinflammation in a rat model of MRSA intracranial infection by modulating brain endothelial cells. World J Emerg Med 2025; 16:239-247. [PMID: 40406305 PMCID: PMC12093437 DOI: 10.5847/wjem.j.1920-8642.2025.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/26/2025] [Indexed: 05/26/2025] Open
Abstract
BACKGROUND The present study aims to investigate whether mannitol facilitates central nervous system (CNS) entry of vancomycin and alleviates methicillin-resistant Staphylococcus aureus (MRSA) intracranial infection. METHODS Blood-brain barrier (BBB) permeability was assessed by measuring the concentration of sodium fluorescein (NaF) in the brain tissues of rats and fluorescein isothiocyanate-dextran (FITC-dextran) in a single-cell layer model. Neutrophil infiltration in the brain tissue, inflammatory cytokine levels in the serum, neurological function, and 7-day survival rates were used to evaluate therapeutic effects of mannitol and vancomycin in MRSA-infected rats. Syndecan-1 and filamentous actin (F-actin) levels were measured, and the relationship between F-actin and the endothelial glycocalyx layer (EGL) was explored via the depolymerization agent cytochalasin D and the polymerization agent jasplakinolide. RESULTS Following mannitol administration, the NaF and vancomycin concentrations in the brain tissue increased rapidly within 5 min and remained stable for 30 min, indicating that mannitol increased BBB permeability for 30 min. In vitro, mannitol treatment led to significantly greater FITC-dextran permeation through a single-cell layer compared to controls. In the MRSA intracranial infection model, rats treated with mannitol and vancomycin simultaneously presented less inflammation, improved neurological function, and increased 7-day survival rate compared to rats treated with vancomycin and mannitol at 10-hour intervals. Further experiments revealed that mannitol decreased the expression of syndecan-1 in brain tissues, which was confirmed by in vitro experiments showing that mannitol significantly decreased syndecan-1 via F-actin depolymerization. CONCLUSION Mannitol may enhance the therapeutic efficacy of vancomycin against intracranial MRSA infection by decreasing the endothelial glycocalyx of the BBB via F-actin depolymerization.
Collapse
Affiliation(s)
- Yin Wen
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Zhiwei Su
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Huishan Zhu
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Mengting Liu
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zhuo Li
- Department of Critical Care Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shiying Zhang
- Department of Emergency Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Shuangming Cai
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Jiaqi Tang
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Hongguang Ding
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Hongke Zeng
- Department of Critical Care Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| |
Collapse
|
3
|
Meller SJ, Greer CA. Olfactory Development and Dysfunction: Involvement of Microglia. Physiology (Bethesda) 2025; 40:0. [PMID: 39499248 DOI: 10.1152/physiol.00037.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/07/2024] Open
Abstract
Olfactory deficits are increasingly recognized in a variety of neurological, neurodevelopmental, psychiatric, and viral diseases. While the pathology underlying olfactory loss is likely to differ across diseases, one shared feature may be an immune response mediated by microglia. Microglia orchestrate the brain's response to environmental insults and maintain neurodevelopmental homeostasis. Here, we explore the potential involvement of microglia in olfactory development and loss in disease. The effects of microglia-mediated immune response during development may be of special relevance to the olfactory system, which is unique in both its vulnerability to environmental insults as well as its extended period of neurogenesis and neuronal migration.
Collapse
Affiliation(s)
- Sarah J Meller
- Departments of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, United States
- Neurosurgery, Yale University School of Medicine, New Haven, Connecticut, United States
- The Interdepartmental Neuroscience Graduate Program, Yale University School of Medicine, New Haven, Connecticut, United States
| | - Charles A Greer
- Departments of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, United States
- Neurosurgery, Yale University School of Medicine, New Haven, Connecticut, United States
- The Interdepartmental Neuroscience Graduate Program, Yale University School of Medicine, New Haven, Connecticut, United States
| |
Collapse
|
4
|
Rennie C, Morshed N, Faria M, Collins-Praino L, Care A. Nanoparticle Association with Brain Cells Is Augmented by Protein Coronas Formed in Cerebrospinal Fluid. Mol Pharm 2025; 22:940-957. [PMID: 39805033 DOI: 10.1021/acs.molpharmaceut.4c01179] [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] [Indexed: 01/16/2025]
Abstract
Neuronanomedicine harnesses nanoparticle technology for the treatment of neurological disorders. An unavoidable consequence of nanoparticle delivery to biological systems is the formation of a protein corona on the nanoparticle surface. Despite the well-established influence of the protein corona on nanoparticle behavior and fate, as well as FDA approval of neuro-targeted nanotherapeutics, the effect of a physiologically relevant protein corona on nanoparticle-brain cell interactions is insufficiently explored. Indeed, less than 1% of protein corona studies have investigated protein coronas formed in cerebrospinal fluid (CSF), the fluid surrounding the brain. Herein, we utilize two clinically relevant polymeric nanoparticles (PLGA and PLGA-PEG) to evaluate the formation of serum and CSF protein coronas. LC-MS analysis revealed distinct protein compositions, with selective enrichment/depletion profiles. Enhanced association of CSF precoated particles with brain cells demonstrates the importance of selecting physiologically relevant biological fluids to more accurately study protein corona formation and subsequent nanoparticle-cell interactions, paving the way for improved nanoparticle engineering for in vivo applications.
Collapse
Affiliation(s)
- Claire Rennie
- School of Life Sciences, University of Technology Sydney, Sydney 2007, New South Wales, Australia
- Australian Institute for Microbiology and Infection, Sydney 2007, New South Wales, Australia
| | - Nabila Morshed
- School of Life Sciences, University of Technology Sydney, Sydney 2007, New South Wales, Australia
| | - Matthew Faria
- Department of Biomedical Engineering, The University of Melbourne, Melbourne 3010, Victoria, Australia
| | - Lyndsey Collins-Praino
- School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, South Australia, Australia
| | - Andrew Care
- School of Life Sciences, University of Technology Sydney, Sydney 2007, New South Wales, Australia
| |
Collapse
|
5
|
Duan Z, Zhou W, He S, Wang W, Huang H, Yi L, Zhang R, Chen J, Zan X, You C, Gao X. Intranasal Delivery of Curcumin Nanoparticles Improves Neuroinflammation and Neurological Deficits in Mice with Intracerebral Hemorrhage. SMALL METHODS 2024; 8:e2400304. [PMID: 38577823 DOI: 10.1002/smtd.202400304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 03/24/2024] [Indexed: 04/06/2024]
Abstract
Intracerebral hemorrhage (ICH) represents one of the most severe subtypes of stroke. Due to the complexity of the brain injury mechanisms following ICH, there are currently no effective treatments to significantly improve patient functional outcomes. Curcumin, as a potential therapeutic agent for ICH, is limited by its poor water solubility and oral bioavailability. In this study, mPEG-PCL is used to encapsulate curcumin, forming curcumin nanoparticles, and utilized the intranasal administration route to directly deliver curcumin nanoparticles from the nasal cavity to the brain. By inhibiting pro-inflammatory neuroinflammation of microglia following ICH in mice, reprogramming pro-inflammatory microglia toward an anti-inflammatory function, and consequently reducing neuronal inflammatory death and hematoma volume, this approach improved blood-brain barrier damage in ICH mice and promoted the recovery of neurological function post-stroke. This study offers a promising therapeutic strategy for ICH to mediate neuroinflammatory microenvironments.
Collapse
Affiliation(s)
- Zhongxin Duan
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Wenjie Zhou
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, 610041, China
| | - Shi He
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Wanyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Hongyi Huang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Linbin Yi
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Rui Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Junli Chen
- Department of Pathophysiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xin Zan
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Chao You
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
6
|
Ferrão R, Rai A. Advanced Polymeric Nanoparticles for the Treatment of Neurodegenerative Diseases. CHEMICAL PHYSICS OF POLYMER NANOCOMPOSITES 2024:843-885. [DOI: 10.1002/9783527837021.ch27] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
7
|
Sierri G, Patrucco M, Ferrario D, Renda A, Comi S, Ciprandi M, Fontanini V, Sica FS, Sesana S, Costa Verdugo M, Kravicz M, Salassa L, Busnelli M, Re F. Targeting specific brain districts for advanced nanotherapies: A review from the perspective of precision nanomedicine. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1991. [PMID: 39251878 PMCID: PMC11670049 DOI: 10.1002/wnan.1991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 09/11/2024]
Abstract
Numerous studies are focused on nanoparticle penetration into the brain functionalizing them with ligands useful to cross the blood-brain barrier. However, cell targeting is also crucial, given that cerebral pathologies frequently affect specific brain cells or areas. Functionalize nanoparticles with the most appropriate targeting elements, tailor their physical parameters, and consider the brain's complex anatomy are essential aspects for precise therapy and diagnosis. In this review, we addressed the state of the art on targeted nanoparticles for drug delivery in diseased brain regions, outlining progress, limitations, and ongoing challenges. We also provide a summary and overview of general design principles that can be applied to nanotherapies, considering the areas and cell types affected by the most common brain disorders. We then emphasize lingering uncertainties that hinder the translational possibilities of nanotherapies for clinical use. Finally, we offer suggestions for continuing preclinical investigations to enhance the overall effectiveness of precision nanomedicine in addressing neurological conditions. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Giulia Sierri
- School of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Michela Patrucco
- School of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
- PhD program in Neuroscience, School of Medicine and Surgery, University of Milano‐Bicocca, Italy
| | - Davide Ferrario
- School of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Antonio Renda
- School of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Susanna Comi
- School of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Matilde Ciprandi
- Department of Biotechnology and BiosciencesUniversity of Milano‐BicoccaMilanItaly
| | | | | | - Silvia Sesana
- School of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | | | - Marcelo Kravicz
- School of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Luca Salassa
- Donostia International Physics Center (DIPC)DonostiaEuskadiSpain
- Polimero eta Material Aurreratuak: Fisika, Kimika eta Teknologia, Kimika FakultateaEuskal Herriko Unibertsitatea UPV/EHUDonostiaSpain
- Basque Foundation for ScienceIkerbasqueBilbaoSpain
| | - Marta Busnelli
- Institute of Neuroscience, National Research CouncilItaly
| | - Francesca Re
- School of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| |
Collapse
|
8
|
Madadi AK, Sohn MJ. Advances in Intrathecal Nanoparticle Delivery: Targeting the Blood-Cerebrospinal Fluid Barrier for Enhanced CNS Drug Delivery. Pharmaceuticals (Basel) 2024; 17:1070. [PMID: 39204177 PMCID: PMC11357388 DOI: 10.3390/ph17081070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 09/03/2024] Open
Abstract
The blood-cerebrospinal fluid barrier (BCSFB) tightly regulates molecular exchanges between the bloodstream and cerebrospinal fluid (CSF), creating challenges for effective central nervous system (CNS) drug delivery. This review assesses intrathecal (IT) nanoparticle (NP) delivery systems that aim to enhance drug delivery by circumventing the BCSFB, complementing approaches that target the blood-brain barrier (BBB). Active pharmaceutical ingredients (APIs) face hurdles like restricted CNS distribution and rapid clearance, which diminish the efficacy of IT therapies. NPs can be engineered to extend drug circulation times, improve CNS penetration, and facilitate sustained release. This review discusses key pharmacokinetic (PK) parameters essential for the effectiveness of these systems. NPs can quickly traverse the subarachnoid space and remain within the leptomeninges for extended periods, often exceeding three weeks. Some designs enable deeper brain parenchyma penetration. Approximately 80% of NPs in the CSF are cleared through the perivascular glymphatic pathway, with microglia-mediated transport significantly contributing to their paravascular clearance. This review synthesizes recent progress in IT-NP delivery across the BCSFB, highlighting critical findings, ongoing challenges, and the therapeutic potential of surface modifications and targeted delivery strategies.
Collapse
Affiliation(s)
- Ahmad Khalid Madadi
- Department of Biomedical Science, Graduate School of Medicine, Inje University, 75, Bokji-ro, Busanjingu, Busan 47392, Republic of Korea;
| | - Moon-Jun Sohn
- Department of Biomedical Science, Graduate School of Medicine, Inje University, 75, Bokji-ro, Busanjingu, Busan 47392, Republic of Korea;
- Department of Neurosurgery, Neuroscience & Radiosurgery Hybrid Research Center, Inje University Ilsan Paik Hospital, College of Medicine, Juhwa-ro 170, Ilsanseo-gu, Goyang City 10380, Republic of Korea
| |
Collapse
|
9
|
Borrajo ML, Quijano A, Lapuhs P, Rodriguez-Perez AI, Anthiya S, Labandeira-Garcia JL, Valenzuela R, Alonso MJ. Ionizable nanoemulsions for RNA delivery into the central nervous system - importance of diffusivity. J Control Release 2024; 372:295-303. [PMID: 38909703 DOI: 10.1016/j.jconrel.2024.06.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
Lipid nanoparticles (LNPs) currently dominate the RNA delivery landscape; however their limited diffusivity hampers targeted tissue dissemination, and, hence, their capacity for intracellular drug delivery. This is especially relevant for tissues such as the central nervous system (CNS), where overcoming proactive brain barriers is crucial for the efficacy of genetic therapeutics. This research aimed to create ionizable nanoemulsions (iNEs), a new generation of RNA delivery systems with enhanced diffusivity. The developed iNEs (consisting of the combination of C12-200, DOPE, Vitamin E, and DMG-PEG) with a size below 100 nm, neutral surface charge, and high RNA loading capacity, showed excellent cell viability and transfection efficiency in various cellular models, including neurons, astrocytes, and microglia. Subsequently, iNEs containing mRNA GFP were tested for CNS transfection, highlighting their exceptional diffusivity and selective transfection of neurons following intra-parenchymal administration.
Collapse
Affiliation(s)
- Mireya L Borrajo
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Av. Barcelona s/n, Campus Vida, University de Santiago de Compostela, 15782 Santiago de Compostela, Spain; Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; IDIS Research Institute, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Aloia Quijano
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Av. Barcelona s/n, Campus Vida, University de Santiago de Compostela, 15782 Santiago de Compostela, Spain; IDIS Research Institute, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Philipp Lapuhs
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Av. Barcelona s/n, Campus Vida, University de Santiago de Compostela, 15782 Santiago de Compostela, Spain; Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; IDIS Research Institute, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ana I Rodriguez-Perez
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Av. Barcelona s/n, Campus Vida, University de Santiago de Compostela, 15782 Santiago de Compostela, Spain; IDIS Research Institute, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Networking Research Center of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Shubaash Anthiya
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Av. Barcelona s/n, Campus Vida, University de Santiago de Compostela, 15782 Santiago de Compostela, Spain; Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; IDIS Research Institute, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - José L Labandeira-Garcia
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Av. Barcelona s/n, Campus Vida, University de Santiago de Compostela, 15782 Santiago de Compostela, Spain; IDIS Research Institute, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Networking Research Center of Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Rita Valenzuela
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Av. Barcelona s/n, Campus Vida, University de Santiago de Compostela, 15782 Santiago de Compostela, Spain; IDIS Research Institute, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; Networking Research Center of Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Av. Barcelona s/n, Campus Vida, University de Santiago de Compostela, 15782 Santiago de Compostela, Spain; Department of Pharmacy and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain; IDIS Research Institute, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| |
Collapse
|
10
|
Gao Z. Strategies for enhanced gene delivery to the central nervous system. NANOSCALE ADVANCES 2024; 6:3009-3028. [PMID: 38868835 PMCID: PMC11166101 DOI: 10.1039/d3na01125a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/12/2024] [Indexed: 06/14/2024]
Abstract
The delivery of genes to the central nervous system (CNS) has been a persistent challenge due to various biological barriers. The blood-brain barrier (BBB), in particular, hampers the access of systemically injected drugs to parenchymal cells, allowing only a minimal percentage (<1%) to pass through. Recent scientific insights highlight the crucial role of the extracellular space (ECS) in governing drug diffusion. Taking into account advancements in vectors, techniques, and knowledge, the discussion will center on the most notable vectors utilized for gene delivery to the CNS. This review will explore the influence of the ECS - a dynamically regulated barrier-on drug diffusion. Furthermore, we will underscore the significance of employing remote-control technologies to facilitate BBB traversal and modulate the ECS. Given the rapid progress in gene editing, our discussion will also encompass the latest advances focused on delivering therapeutic editing in vivo to the CNS tissue. In the end, a brief summary on the impact of Artificial Intelligence (AI)/Machine Learning (ML), ultrasmall, soft endovascular robots, and high-resolution endovascular cameras on improving the gene delivery to the CNS will be provided.
Collapse
Affiliation(s)
- Zhenghong Gao
- Mechanical Engineering, The University of Texas at Dallas USA
| |
Collapse
|
11
|
Huang Y, Guo X, Wu Y, Chen X, Feng L, Xie N, Shen G. Nanotechnology's frontier in combatting infectious and inflammatory diseases: prevention and treatment. Signal Transduct Target Ther 2024; 9:34. [PMID: 38378653 PMCID: PMC10879169 DOI: 10.1038/s41392-024-01745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/27/2023] [Accepted: 01/11/2024] [Indexed: 02/22/2024] Open
Abstract
Inflammation-associated diseases encompass a range of infectious diseases and non-infectious inflammatory diseases, which continuously pose one of the most serious threats to human health, attributed to factors such as the emergence of new pathogens, increasing drug resistance, changes in living environments and lifestyles, and the aging population. Despite rapid advancements in mechanistic research and drug development for these diseases, current treatments often have limited efficacy and notable side effects, necessitating the development of more effective and targeted anti-inflammatory therapies. In recent years, the rapid development of nanotechnology has provided crucial technological support for the prevention, treatment, and detection of inflammation-associated diseases. Various types of nanoparticles (NPs) play significant roles, serving as vaccine vehicles to enhance immunogenicity and as drug carriers to improve targeting and bioavailability. NPs can also directly combat pathogens and inflammation. In addition, nanotechnology has facilitated the development of biosensors for pathogen detection and imaging techniques for inflammatory diseases. This review categorizes and characterizes different types of NPs, summarizes their applications in the prevention, treatment, and detection of infectious and inflammatory diseases. It also discusses the challenges associated with clinical translation in this field and explores the latest developments and prospects. In conclusion, nanotechnology opens up new possibilities for the comprehensive management of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Yujing Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xiaohan Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yi Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Xingyu Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Lixiang Feng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Na Xie
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Guobo Shen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
12
|
Tian M, Ma Z, Yang GZ. Micro/nanosystems for controllable drug delivery to the brain. Innovation (N Y) 2024; 5:100548. [PMID: 38161522 PMCID: PMC10757293 DOI: 10.1016/j.xinn.2023.100548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/26/2023] [Indexed: 01/03/2024] Open
Abstract
Drug delivery to the brain is crucial in the treatment for central nervous system disorders. While significant progress has been made in recent years, there are still major challenges in achieving controllable drug delivery to the brain. Unmet clinical needs arise from various factors, including controlled drug transport, handling large drug doses, methods for crossing biological barriers, the use of imaging guidance, and effective models for analyzing drug delivery. Recent advances in micro/nanosystems have shown promise in addressing some of these challenges. These include the utilization of microfluidic platforms to test and validate the drug delivery process in a controlled and biomimetic setting, the development of novel micro/nanocarriers for large drug loads across the blood-brain barrier, and the implementation of micro-intervention systems for delivering drugs through intraparenchymal or peripheral routes. In this article, we present a review of the latest developments in micro/nanosystems for controllable drug delivery to the brain. We also delve into the relevant diseases, biological barriers, and conventional methods. In addition, we discuss future prospects and the development of emerging robotic micro/nanosystems equipped with directed transportation, real-time image guidance, and closed-loop control.
Collapse
Affiliation(s)
- Mingzhen Tian
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhichao Ma
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guang-Zhong Yang
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
13
|
Chen Y, Liang Z, Wang Q, Xiao L, Xie S, Yang S, Liu X, Ling D, Li F. Alpha-Synuclein Oligomers Driven T1-T2 Switchable Nanoprobes for Early and Accurate Diagnosis of Parkinson's Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2310404. [PMID: 38149464 DOI: 10.1002/adma.202310404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/28/2023] [Indexed: 12/28/2023]
Abstract
The alpha-synuclein (α-syn) oligomers hold a central role in the pathology of Parkinson's disease (PD). Achieving accurate detection of α-syn oligomers in vivo presents a promising avenue for early and accurate diagnosis of PD. Magnetic resonance imaging (MRI), with non-invasion and exceptional tissue penetration, offers a potent tool for visualizing α-syn oligomers in vivo. Nonetheless, ensuring diagnostic specificity remains a formidable challenge. Herein, a novel MRI probe (ASOSN) is introduced, which encompasses highly sensitive antiferromagnetic nanoparticles functionalized with single-chain fragment variable antibodies, endowing it with the capacity for discerning recognition and binding to α-syn oligomers and triggering a switchable T1-T2 MRI signal. Significantly, ASOSN possesses the unique capability to accurately discriminate α-syn oligomers from neuroinflammation in vivo. Moreover, ASOSN facilitates the non-invasive and precise visualizing of endogenous α-syn oligomers in living systems. This innovative design heralds the development of a non-invasive visualization strategy for α-syn oligomers, marking a pivotal advancement for early and accurate diagnosis of PD.
Collapse
Affiliation(s)
- Ying Chen
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Zeyu Liang
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Qiyue Wang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai, 201203, China
| | - Lin Xiao
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shangzhi Xie
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shengfei Yang
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xun Liu
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai, 201203, China
| | - Daishun Ling
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University, Shanghai, 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai, 201203, China
| | - Fangyuan Li
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- World Laureates Association (WLA) Laboratories, Shanghai, 201203, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou, 310009, China
| |
Collapse
|
14
|
Kung Y, Wu CH, Lin MT, Liao WH, Chen WS, Hsiao MY. Blood-cerebrospinal fluid barrier opening by modified single pulse transcranial focused shockwave. Drug Deliv 2023; 30:97-107. [PMID: 36533878 PMCID: PMC9769131 DOI: 10.1080/10717544.2022.2157068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Transcranial focused shockwave (FSW) is a novel noninvasive brain stimulation that can open blood-brain barriers (BBB) and blood-cerebrospinal fluid barriers (BCSFB) with a single low-energy (energy flux density 0.03 mJ/mm2) pulse and low-dose microbubbles (2 × 106/kg). Similar to focused ultrasound, FSW deliver highly precise stimulation of discrete brain regions with adjustable focal lengths that essentially covers the whole brain. By opening the BCSFB, it allows for rapid widespread drug delivery to the whole brain by cerebrospinal fluid (CSF) circulation. Although no definite adverse effect or permeant injury was noted in our previous study, microscopic hemorrhage was infrequently observed. Safety concerns remain the major obstacle to further application of FSW in brain. To enhance its applicability, a modified single pulse FSW technique was established that present 100% opening rate but much less risk of adverse effect than previous methods. By moving the targeting area 2.5 mm more superficially on the left lateral ventricle as compared with the previous methods, the microscopic hemorrhage rate was reduced to zero. We systemically examine the safety profiles of the modified FSW-BCSFB opening regarding abnormal behavior and brain injury or hemorrhage 72 hr after 0, 1, and 10 pulses of FSW-treatment. Animal behavior, physiological monitor, and brain MRI were examined and recorded. Brain section histology was examined for hemorrhage, apoptosis, inflammation, oxidative stress related immunohistochemistry and biomarkers. The single pulse FSW group demonstrated no mortality or gross/microscopic hemorrhage (N = 30), and no observable changes in all examined outcomes, while 10 pulses of FSW was found to be associated with microscopic and temporary RBC extravasation (N = 6/30), and abnormal immunohistochemistry biomarkers which showed a trend of recovery at 72 hrs. The results suggest that single pulse low-energy FSW-BCSFB opening is effective, safe and poses minimal risk of injury to brain tissue (Sprague Dawley, SD rats).
Collapse
Affiliation(s)
- Yi Kung
- Department of Biomechatronic Engineering, National Chiayi University, Chiayi City, Taiwan
| | - Chueh-Hung Wu
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Meng-Ting Lin
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Wei-Hao Liao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Wen-Shiang Chen
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan,Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| | - Ming-Yen Hsiao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan,CONTACT Ming-Yen Hsiao
| |
Collapse
|
15
|
Fan Y, Xu C, Deng N, Gao Z, Jiang Z, Li X, Zhou Y, Pei H, Li L, Tang B. Understanding drug nanocarrier and blood-brain barrier interaction based on a microfluidic microphysiological model. LAB ON A CHIP 2023; 23:1935-1944. [PMID: 36891748 DOI: 10.1039/d2lc01077a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
As many nanoparticles (NPs) have been exploited as drug carriers to overcome the resistance of the blood-brain barrier (BBB), reliable in vitro BBB models are urgently needed to help researchers to comprehensively understand drug nanocarrier-BBB interaction during penetration, which can prompt pre-clinical nanodrug exploitation. Herein, we developed a microfluidic microphysiological model, allowing the analysis of BBB homeostasis and NP penetration. We found that the BBB penetrability of gold nanoparticles (AuNPs) was size- and modification-dependent, which might be caused by a distinct transendocytosis pathway. Notably, transferrin-modified 13 nm AuNPs held the strongest BBB penetrability and induced the slightest BBB dysfunction, while bare 80 nm and 120 nm AuNPs showed opposite results. Moreover, further analysis of the protein corona showed that PEGylation reduced the protein absorption, and some proteins facilitated the BBB penetration of NPs. The developed microphysiological model provides a powerful tool for understanding the drug nanocarrier-BBB interaction, which is vital for exploiting high-efficiency and biocompatible nanodrugs.
Collapse
Affiliation(s)
- Yuanyuan Fan
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Chang Xu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Ning Deng
- Shandong Institute for Product Quality Inspection, Jinan 250101, P. R. China
| | - Ze Gao
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Zhongyao Jiang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Xiaoxiao Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Yingshun Zhou
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Haimeng Pei
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Lu Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education Shandong Normal University, Jinan 250014, P. R. China.
| |
Collapse
|
16
|
Kim TY, Lee GH, Mun J, Cheong S, Choi I, Kim H, Hahn SK. Smart Contact Lens Systems for Ocular Drug Delivery and Therapy. Adv Drug Deliv Rev 2023; 196:114817. [PMID: 37004938 DOI: 10.1016/j.addr.2023.114817] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
Ocular drug delivery and therapy systems have been extensively investigated with various methods including direct injections, eye drops and contact lenses. Nowadays, smart contact lens systems are attracting a lot of attention for ocular drug delivery and therapy due to their minimally invasive or non-invasive characteristics, highly enhanced drug permeation, high bioavailability, and on-demand drug delivery. Furthermore, smart contact lens systems can be used for direct light delivery into the eyes for biophotonic therapy replacing the use of drugs. Here, we review smart contact lens systems which can be classified into two groups of drug-eluting contact lens and ocular device contact lens. More specifically, this review covers smart contact lens systems with nanocomposite-laden systems, polymeric film-incorporated systems, micro and nanostructure systems, iontophoretic systems, electrochemical systems, and phototherapy systems for ocular drug delivery and therapy. After that, we discuss the future opportunities, challenges and perspectives of smart contact lens systems for ocular drug delivery and therapy.
Collapse
Affiliation(s)
- Tae Yeon Kim
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Geon-Hui Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Jonghwan Mun
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Sunah Cheong
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Inhoo Choi
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Hyemin Kim
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - Sei Kwang Hahn
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 77 Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea; PHI BIOMED Co., 168 Yeoksam-ro, Gangnamgu, Seoul 06248, Republic of Korea.
| |
Collapse
|
17
|
Metzger JM, Wang Y, Neuman SS, Snow KJ, Murray SA, Lutz CM, Bondarenko V, Felton J, Gimse K, Xie R, Li D, Zhao Y, Flowers MT, Simmons HA, Roy S, Saha K, Levine JE, Emborg ME, Gong S. Efficient in vivo neuronal genome editing in the mouse brain using nanocapsules containing CRISPR-Cas9 ribonucleoproteins. Biomaterials 2023; 293:121959. [PMID: 36527789 PMCID: PMC9868115 DOI: 10.1016/j.biomaterials.2022.121959] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
Genome editing of somatic cells via clustered regularly interspaced short palindromic repeats (CRISPR) offers promise for new therapeutics to treat a variety of genetic disorders, including neurological diseases. However, the dense and complex parenchyma of the brain and the post-mitotic state of neurons make efficient genome editing challenging. In vivo delivery systems for CRISPR-Cas proteins and single guide RNA (sgRNA) include both viral vectors and non-viral strategies, each presenting different advantages and disadvantages for clinical application. We developed non-viral and biodegradable PEGylated nanocapsules (NCs) that deliver preassembled Cas9-sgRNA ribonucleoproteins (RNPs). Here, we show that the RNP NCs led to robust genome editing in neurons following intracerebral injection into the healthy mouse striatum. Genome editing was predominantly observed in medium spiny neurons (>80%), with occasional editing in cholinergic, calretinin, and parvalbumin interneurons. Glial activation was minimal and was localized along the needle tract. Our results demonstrate that the RNP NCs are capable of safe and efficient neuronal genome editing in vivo.
Collapse
Affiliation(s)
- Jeanette M Metzger
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Yuyuan Wang
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Samuel S Neuman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Kathy J Snow
- The Jackson Laboratory, Bar Harbor, ME, 04609, USA
| | | | | | - Viktoriya Bondarenko
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Jesi Felton
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Kirstan Gimse
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Ruosen Xie
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Dongdong Li
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Yi Zhao
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Matthew T Flowers
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Heather A Simmons
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Subhojit Roy
- Departments of Pathology and Neuroscience, University of California-San Diego, San Diego, CA, 92093, USA
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Jon E Levine
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Marina E Emborg
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, 53715, USA.
| | - Shaoqin Gong
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, 53715, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA.
| |
Collapse
|
18
|
Huang T, Wu J, Mu J, Gao J. Advanced Therapies for Traumatic Central Nervous System Injury: Delivery Strategy Reinforced Efficient Microglial Manipulation. Mol Pharm 2023; 20:41-56. [PMID: 36469398 DOI: 10.1021/acs.molpharmaceut.2c00605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Traumatic central nervous system (CNS) injuries, including spinal cord injury and traumatic brain injury, are challenging enemies of human health. Microglia, the main component of the innate immune system in CNS, can be activated postinjury and are key participants in the pathological procedure and development of CNS trauma. Activated microglia can be typically classified into pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes. Reducing M1 polarization while promoting M2 polarization is thought to be promising for CNS injury treatment. However, obstacles such as the low permeability of the blood-brain barrier and short retention time in circulation limit the therapeutic outcomes of administrated drugs, and rational delivery strategies are necessary for efficient microglial regulation. To this end, proper administration methods and delivery systems like nano/microcarriers and scaffolds are investigated to augment the therapeutic effects of drugs, while some of these delivery systems have self-efficacies in microglial manipulation. Besides, systems based on cell and cell-derived exosomes also show impressive effects, and some underlying targeting mechanisms of these delivery systems have been discovered. In this review, we introduce the roles of microglia play in traumatic CNS injuries, discuss the potential targets for the polarization regulation of microglial phenotype, and summarize recent studies and clinical trials about delivery strategies on enhancing the effect of microglial regulation and therapeutic outcome, as well as targeting mechanisms post CNS trauma.
Collapse
Affiliation(s)
- Tianchen Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiahe Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer, Pharmacology and Toxicology Research of Zhejiang Province, Affiliated, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Jiafu Mu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Jinhua Institute of Zhejiang University, Jinhua 321002, China
| |
Collapse
|
19
|
Jin L, Zhu Z, Hong L, Qian Z, Wang F, Mao Z. ROS-responsive 18β-glycyrrhetic acid-conjugated polymeric nanoparticles mediate neuroprotection in ischemic stroke through HMGB1 inhibition and microglia polarization regulation. Bioact Mater 2023; 19:38-49. [PMID: 35415314 PMCID: PMC8980441 DOI: 10.1016/j.bioactmat.2022.03.040] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/26/2022] [Accepted: 03/26/2022] [Indexed: 01/01/2023] Open
Abstract
Ischemic stroke is an acute and serious cerebral vascular disease, which greatly affects people's health and brings huge economic burden to society. Microglia, as important innate immune components in central nervous system (CNS), are double-edged swords in the battle of nerve injury, considering their polarization between pro-inflammatory M1 or anti-inflammatory M2 phenotypes. High mobility group box 1 (HMGB1) is one of the potent pro-inflammatory mediators that promotes the M1 polarization of microglia. 18β-glycyrrhetinic acid (GA) is an effective intracellular inhibitor of HMGB1, but of poor water solubility and dose-dependent toxicity. To overcome the shortcomings of GA delivery and to improve the efficacy of cerebral ischemia therapy, herein, we designed reactive oxygen species (ROS) responsive polymer-drug conjugate nanoparticles (DGA) to manipulate microglia polarization by suppressing the translocation of nuclear HMGB1. DGA presented excellent therapeutic efficacy in stroke mice, as evidenced by the reduction of infarct volume, recovery of motor function, suppressed of M1 microglia activation and enhanced M2 activation, and induction of neurogenesis. Altogether, our work demonstrates a close association between HMGB1 and microglia polarization, suggesting potential strategies for coping with inflammatory microglia-related diseases. We synthesized GA-boronate ester-conjugated diethylaminoethylen-dextran polymer-drug conjugate nanoparticles. The DGA nanoparticles achieve ROS-responsive drug release. The DGA nanoparticles inhibit cytoplasmic translocation of nuclear HMGB1, thus modulate microglia to M2 phenotype. The DGA nanoparticles effectively alleviate the pathology of stroke, reduce infarct volume, and enhance neurogenesis.
Collapse
Affiliation(s)
- Lulu Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhixin Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Liangjie Hong
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Zhefeng Qian
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Fang Wang
- The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, 310058, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Corresponding author.
| |
Collapse
|
20
|
Bioactive 2D nanomaterials for neural repair and regeneration. Adv Drug Deliv Rev 2022; 187:114379. [PMID: 35667464 DOI: 10.1016/j.addr.2022.114379] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/12/2022] [Accepted: 05/27/2022] [Indexed: 12/18/2022]
Abstract
Biomaterials have provided promising strategies towards improving the functions of injured tissues of the nervous system. Recently, 2D nanomaterials, such as graphene, layered double hydroxides (LDHs), and black phosphorous, which are characterized by ultrathin film structures, have attracted much attention in the fields of neural repair and regeneration. 2D nanomaterials have extraordinary physicochemical properties and excellent biological activities, such as a large surface-area-to-thickness ratio, high levels of adhesion, and adjustable flexibility. In addition, they can be designed to have superior biocompatibility and electrical or nano-carrier properties. To date, many 2D nanomaterials have been used for synaptic modulation, neuroinflammatory reduction, stem cell fate regulation, and injured neural cell/tissue repair. In this review, we discuss the advances in 2D nanomaterial technology towards novel neurological applications and the mechanisms underlying their unique features. In addition, the future outlook of functional 2D nanomaterials towards addressing the difficult issues of neuropathy has been explored to introduce a promising strategy towards repairing and regenerating the injured nervous system.
Collapse
|
21
|
Targeting neuroinflammation by intranasal delivery of nanoparticles in neurological diseases: a comprehensive review. Naunyn Schmiedebergs Arch Pharmacol 2022; 395:133-148. [PMID: 34982185 DOI: 10.1007/s00210-021-02196-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 12/15/2021] [Indexed: 12/17/2022]
Abstract
Neuroinflammation (NIF) plays an essential role in the pathology of neurological disorders like Parkinson's disease, Alzheimer's disease, multiple sclerosis, and epilepsy. Despite progress in the drug discovery and development of new drugs, drug delivery to the central nervous system (CNS) still represents the challenge due to the presence of the blood-brain barrier (BBB). Targeting NIF may require an adequate amount of drug to cross the BBB. Recently, the intranasal (IN) drug administration has attracted increasing attention as a reliable method to cross the BBB and treat neurological disorders. On the other hand, using optimized nanoparticles may improve the IN delivery limitations, increase the mucoadhesive properties, and prevent drug degradation. NPs can carry and deliver drugs to the CNS by bypassing the BBB. In this review, we described briefly the NIF as a pathologic feature of CNS diseases. The potential treatment possibilities with IN transfer of NP-loaded drugs will enhance the establishment of more efficient nanoformulations and delivery systems.
Collapse
|
22
|
Kung Y, Chen KY, Liao WH, Hsu YH, Wu CH, Hsiao MY, Huang APH, Chen WS. Facilitating drug delivery in the central nervous system by opening the blood-cerebrospinal fluid barrier with a single low energy shockwave pulse. Fluids Barriers CNS 2022; 19:3. [PMID: 34991647 PMCID: PMC8740485 DOI: 10.1186/s12987-021-00303-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Background The blood-cerebrospinal fluid (CSF) barrier (BCSFB) is critically important to the pathophysiology of the central nervous system (CNS). However, this barrier prevents the safe transmission of beneficial drugs from the blood to the CSF and thus the spinal cord and brain, limiting their effectiveness in treating a variety of CNS diseases. Methods This study demonstrates a method on SD rats for reversible and site-specific opening of the BCSFB via a noninvasive, low-energy focused shockwave (FSW) pulse (energy flux density 0.03 mJ/mm2) with SonoVue microbubbles (2 × 106 MBs/kg), posing a low risk of injury. Results By opening the BCSFB, the concentrations of certain CNS-impermeable indicators (70 kDa Evans blue and 500 kDa FITC-dextran) and drugs (penicillin G, doxorubicin, and bevacizumab) could be significantly elevated in the CSF around both the brain and the spinal cord. Moreover, glioblastoma model rats treated by doxorubicin with this FSW-induced BCSFB (FSW-BCSFB) opening technique also survived significantly longer than untreated controls. Conclusion This is the first study to demonstrate and validate a method for noninvasively and selectively opening the BCSFB to enhance drug delivery into CSF circulation. Potential applications may include treatments for neurodegenerative diseases, CNS infections, brain tumors, and leptomeningeal carcinomatosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-021-00303-x.
Collapse
Affiliation(s)
- Yi Kung
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Kuan-Yu Chen
- Division of Pulmonology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei City, Taiwan
| | - Wei-Hao Liao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Yi-Hua Hsu
- Department of Surgery, National Taiwan University Hospital, Taipei City, Taiwan
| | - Chueh-Hung Wu
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Ming-Yen Hsiao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Abel P-H Huang
- Department of Surgery, National Taiwan University Hospital, Taipei City, Taiwan.
| | - Wen-Shiang Chen
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital & National Taiwan University College of Medicine, Taipei City, Taiwan. .,Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan.
| |
Collapse
|
23
|
Khan S, Hussain A, Attar F, Bloukh SH, Edis Z, Sharifi M, Balali E, Nemati F, Derakhshankhah H, Zeinabad HA, Nabi F, Khan RH, Hao X, Lin Y, Hua L, Ten Hagen TLM, Falahati M. A review of the berberine natural polysaccharide nanostructures as potential anticancer and antibacterial agents. Biomed Pharmacother 2021; 146:112531. [PMID: 34906771 DOI: 10.1016/j.biopha.2021.112531] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/02/2021] [Accepted: 12/08/2021] [Indexed: 12/20/2022] Open
Abstract
Despite the promising medicinal properties, berberine (BBR), due to its relatively poor solubility in plasma, low bio-stability and limited bioavailability is not used broadly in clinical stages. Due to these drawbacks, drug delivery systems (DDSs) based on nanoscale natural polysaccharides, are applied to address these concerns. Natural polymers are biodegradable, non-immunogenic, biocompatible, and non-toxic agents that are capable of trapping large amounts of hydrophobic compounds in relatively small volumes. The use of nanoscale natural polysaccharide improves the stability and pharmacokinetics of the small molecules and, consequently, increases the therapeutic effects and reduces the side effects of the small molecules. Therefore, this paper presents an overview of the different methods used for increasing the BBR solubility and bioavailability. Afterwards, the pharmacodynamic and pharmacokinetic of BBR nanostructures were discussed followed by the introduction of natural polysaccharides of plant (cyclodextrines, glucomannan), the shells of crustaceans (chitosan), and the cell wall of brown marine algae (alginate)-based origins used to improve the dissolution rate of poorly soluble BBR and their anticancer and antibacterial properties. Finally, the anticancer and antibacterial mechanisms of free BBR and BBR nanostructures were surveyed. In conclusion, this review may pave the way for providing some useful data in the development of BBR-based platforms for clinical applications.
Collapse
Affiliation(s)
- Suliman Khan
- Advanced Medical Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Arif Hussain
- School of Life Sciences, Manipal Academy of Higher Education, Dubai, United Arab Emirates
| | - Farnoosh Attar
- Department of Biology, Faculty of Food Industry & Agriculture, Standard Research Institute, Karaj, Iran
| | - Samir Haj Bloukh
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, PO Box 346, Ajman, United Arab Emirates; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Zehra Edis
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Ajman University, PO Box 346, Ajman, United Arab Emirates
| | - Majid Sharifi
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran; Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Ebrahim Balali
- Department of Organic Chemistry, Faculty of Pharmaceutical Chemistry, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fahimeh Nemati
- Department of Biotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hossein Derakhshankhah
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hojjat Alizadeh Zeinabad
- Apoptosis Research Centre, Department of Biochemistry, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland; Institute of Pathology, Univesity of Berne, Berne, Switzerland
| | - Faisal Nabi
- Biotechnology Unit, Aligarh Muslim University, India
| | | | - Xiao Hao
- Advanced Medical Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Yueting Lin
- High Level Talent Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China
| | - Linlin Hua
- Advanced Medical Center, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P.R. China.
| | - Timo L M Ten Hagen
- Laboratory Experimental Oncology, Department of Pathology, Erasmus MC, 3015GD Rotterdam, the Netherlands.
| | - Mojtaba Falahati
- Laboratory Experimental Oncology, Department of Pathology, Erasmus MC, 3015GD Rotterdam, the Netherlands.
| |
Collapse
|
24
|
Feng Q, Xu X, Wei C, Li Y, Wang M, Lv C, Wu J, Dai Y, Han Y, Lesniak MS, Fan H, Zhang L, Cheng Y. The Dynamic Interactions between Nanoparticles and Macrophages Impact Their Fate in Brain Tumors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2103600. [PMID: 34643042 DOI: 10.1002/smll.202103600] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/10/2021] [Indexed: 06/13/2023]
Abstract
Functional nanomaterials such as iron oxide nanoparticles have been extensively explored for the diagnosis and treatment of central nervous system diseases. However, an insufficient understanding of the comprehensive nanomaterial-biological interactions in the brain hinders the nanomaterials from meeting the medical requirements for translational research. Here, FDA-approved ferumoxytol, an iron oxide nanoparticle, is chosen as the model nanomaterial for a systematic study of the dynamic interactions between ferumoxytol and immune cells, including microglia and macrophages, in the brain tumors. Strikingly, up to 90% of intratumorally injected ferumoxytol nanoparticles are recognized and phagocytized by tumor-associated microglia and macrophages. The dynamic trafficking progress of ferumoxytol in microglia and macrophages, including scavenger receptor-mediated endocytosis, lysosomal internalization, and extracellular vesicle-dominated excretion, is further studied. Importantly, the results demonstrate that extracellular vesicle-encapsulated nanoparticles could be gradually eliminated from the brain along with cerebrospinal fluid circulation over 21 days. Moreover, ferumoxytol exhibits no obvious long-term neurological toxicity after its injection. The study suggests that the dynamic biointeractions of nanoparticles with immune cells in the brain exert a key rate-limiting impact on the efficiency of targeting tumor cells and their in vivo fate and thus provide a deeper understanding of the nanomaterials in the brain for clinical applications.
Collapse
Affiliation(s)
- Qishuai Feng
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Collaborative Innovation Center for Brain Science, Tongji University, 1800 Yuntai Road, Shanghai, 200123, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200092, China
| | - Xianyun Xu
- The First Rehabilitation Hospital of Shanghai, Tongji University School of Medicine, 349 Hangzhou Road, Shanghai, 200090, China
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Department of Pediatric Surgery, The First Affiliated Hospital of Gannan Medical University, Gannan Medical University, 1 Yixueyuan Road, Ganzhou, Jiangxi, 341000, China
| | - Chen Wei
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Collaborative Innovation Center for Brain Science, Tongji University, 1800 Yuntai Road, Shanghai, 200123, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200092, China
| | - Yingze Li
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Collaborative Innovation Center for Brain Science, Tongji University, 1800 Yuntai Road, Shanghai, 200123, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200092, China
| | - Min Wang
- Shanghai Key Lab of Tuberculofsis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China
| | - Cheng Lv
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Collaborative Innovation Center for Brain Science, Tongji University, 1800 Yuntai Road, Shanghai, 200123, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200092, China
| | - Jiaojiao Wu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Collaborative Innovation Center for Brain Science, Tongji University, 1800 Yuntai Road, Shanghai, 200123, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200092, China
| | - Yalei Dai
- Shanghai Key Lab of Tuberculofsis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China
| | - Yu Han
- Feinberg School of Medicine, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, IL, 60611, USA
| | - Maciej S Lesniak
- Feinberg School of Medicine, Northwestern University, 676 North Saint Clair Street, Suite 2210, Chicago, IL, 60611, USA
| | - Haiming Fan
- College of Chemistry and Materials Science, Northwest University, Xi'an, 710127, China
| | - Ling Zhang
- The First Rehabilitation Hospital of Shanghai, Tongji University School of Medicine, 349 Hangzhou Road, Shanghai, 200090, China
| | - Yu Cheng
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Medicine, Collaborative Innovation Center for Brain Science, Tongji University, 1800 Yuntai Road, Shanghai, 200123, China
- Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai, 200092, China
| |
Collapse
|
25
|
Picone P, Palumbo FS, Federico S, Pitarresi G, Adamo G, Bongiovanni A, Chaves A, Cancemi P, Muccilli V, Giglio V, Vetri V, Anselmo S, Sancataldo G, Di Liberto V, Nuzzo D. Nano-structured myelin: new nanovesicles for targeted delivery to white matter and microglia, from brain-to-brain. Mater Today Bio 2021; 12:100146. [PMID: 34761196 PMCID: PMC8567303 DOI: 10.1016/j.mtbio.2021.100146] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 01/04/2023] Open
Abstract
Neurodegenerative diseases affect millions of people worldwide and the presence of various physiological barriers limits the accessibility to the brain and reduces the efficacy of various therapies. Moreover, new carriers having targeting properties to specific brain regions and cells are needed in order to improve therapies for the brain disorder treatment. In this study, for the first time, Myelin nanoVesicles (hereafter defined MyVes) from brain-extracted myelin were produced. The MyVes have an average diameter of 100–150 nm, negative zeta potential, spheroidal morphology, and contain lipids and the key proteins of the myelin sheath. Furthermore, they exhibit good cytocompatibility. The MyVes were able to target the white matter and interact mainly with the microglia cells. The preliminary results here presented allow us to suppose the employment of MyVes as potential carrier to target the white matter and microglia in order to counteract white matter microglia-related diseases. Bio-fabrication of brain tissue derived nanovesicles: myelin nanovesicles. Myelin nanovesicles contain the main proteins of the myelin sheath (myelin basic protein and myelin proteolipid protein). Myelin nanovesicles can lade a drug/molecule and cross a blood–brain barrier model. Myelin nanovesicles target white matter and microglia cells.
Collapse
Affiliation(s)
- Pasquale Picone
- Istituto per la Ricerca e l’Innovazione Biomedica, CNR, via U. La Malfa 153, 90146, Palermo, Italy
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy
- Corresponding author.
| | - Fabio Salvatore Palumbo
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy
| | - Salvatore Federico
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy
| | - Giovanna Pitarresi
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy
| | - Giorgia Adamo
- Istituto per la Ricerca e l’Innovazione Biomedica, CNR, via U. La Malfa 153, 90146, Palermo, Italy
| | - Antonella Bongiovanni
- Istituto per la Ricerca e l’Innovazione Biomedica, CNR, via U. La Malfa 153, 90146, Palermo, Italy
| | - Antonio Chaves
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Festa del Perdono 7, 20122, Milano, Italy
| | - Patrizia Cancemi
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy
| | - Vera Muccilli
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria, 6, I-95125, Catania, Italy
| | - Valentina Giglio
- Dipartimento di Scienze Chimiche, Università degli Studi di Catania, Viale A. Doria, 6, I-95125, Catania, Italy
| | - Valeria Vetri
- Dipartimento di Fisica e Chimica-Emilio Segrè, Università degli studi di Palermo, Viale delle Scienze edificio 18, 90128, Palermo, Italy
| | - Sara Anselmo
- Dipartimento di Fisica e Chimica-Emilio Segrè, Università degli studi di Palermo, Viale delle Scienze edificio 18, 90128, Palermo, Italy
| | - Giuseppe Sancataldo
- Dipartimento di Fisica e Chimica-Emilio Segrè, Università degli studi di Palermo, Viale delle Scienze edificio 18, 90128, Palermo, Italy
| | - Valentina Di Liberto
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Corso Tukory 129, 90134, Palermo, Italy
| | - Domenico Nuzzo
- Istituto per la Ricerca e l’Innovazione Biomedica, CNR, via U. La Malfa 153, 90146, Palermo, Italy
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche, Università di Palermo, Viale delle Scienze, 90128, Palermo, Italy
- Corresponding author.
| |
Collapse
|
26
|
Meneghini V, Peviani M, Luciani M, Zambonini G, Gritti A. Delivery Platforms for CRISPR/Cas9 Genome Editing of Glial Cells in the Central Nervous System. Front Genome Ed 2021; 3:644319. [PMID: 34713256 PMCID: PMC8525379 DOI: 10.3389/fgeed.2021.644319] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 01/21/2021] [Indexed: 12/14/2022] Open
Abstract
Glial cells (astrocytes, oligodendrocytes, and microglia) are emerging as key players in several physiological and pathological processes of the central nervous system (CNS). Astrocytes and oligodendrocytes are not only supportive cells that release trophic factors or regulate energy metabolism, but they also actively modulate critical neuronal processes and functions in the tripartite synapse. Microglia are defined as CNS-resident cells that provide immune surveillance; however, they also actively contribute to shaping the neuronal microenvironment by scavenging cell debris or regulating synaptogenesis and pruning. Given the many interconnected processes coordinated by glial cells, it is not surprising that both acute and chronic CNS insults not only cause neuronal damage but also trigger complex multifaceted responses, including neuroinflammation, which can critically contribute to the disease progression and worsening of symptoms in several neurodegenerative diseases. Overall, this makes glial cells excellent candidates for targeted therapies to treat CNS disorders. In recent years, the application of gene editing technologies has redefined therapeutic strategies to treat genetic and age-related neurological diseases. In this review, we discuss the advantages and limitations of clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-based gene editing in the treatment of neurodegenerative disorders, focusing on the development of viral- and nanoparticle-based delivery methods for in vivo glial cell targeting.
Collapse
Affiliation(s)
- Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Peviani
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Marco Luciani
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giada Zambonini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
27
|
Md S, Alhakamy NA, Alfaleh MA, Afzal O, Altamimi ASA, Iqubal A, Shaik RA. Mechanisms Involved in Microglial-Interceded Alzheimer's Disease and Nanocarrier-Based Treatment Approaches. J Pers Med 2021; 11:1116. [PMID: 34834468 PMCID: PMC8619529 DOI: 10.3390/jpm11111116] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disorder accountable for dementia and cognitive dysfunction. The etiology of AD is complex and multifactorial in origin. The formation and deposition of amyloid-beta (Aβ), hyperphosphorylated tau protein, neuroinflammation, persistent oxidative stress, and alteration in signaling pathways have been extensively explored among the various etiological hallmarks. However, more recently, the immunogenic regulation of AD has been identified, and macroglial activation is considered a limiting factor in its etiological cascade. Macroglial activation causes neuroinflammation via modulation of the NLRP3/NF-kB/p38 MAPKs pathway and is also involved in tau pathology via modulation of the GSK-3β/p38 MAPK pathways. Additionally, microglial activation contributes to the discrete release of neurotransmitters and an altered neuronal synaptic plasticity. Therefore, activated microglial cells appear to be an emerging target for managing and treating AD. This review article discussed the pathology of microglial activation in AD and the role of various nanocarrier-based anti-Alzeihmenr's therapeutic approaches that can either reverse or inhibit this activation. Thus, as a targeted drug delivery system, nanocarrier approaches could emerge as a novel means to overcome existing AD therapy limitations.
Collapse
Affiliation(s)
- Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (M.A.A.)
- Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (M.A.A.)
- Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohamed A. Alfaleh
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (N.A.A.); (M.A.A.)
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (O.A.); (A.S.A.A.)
| | - Abdulmalik S. A. Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (O.A.); (A.S.A.A.)
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India;
| | - Rasheed A. Shaik
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| |
Collapse
|
28
|
Auriemma R, Sponchioni M, Capasso Palmiero U, Manfredini N, Razavi Dinani FS, Moscatelli D. Synthesis of a Diapocynin Prodrug for Its Prolonged Release from Zwitterionic Biodegradable Nanoparticles. MACROMOL CHEM PHYS 2021. [DOI: 10.1002/macp.202100223] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Renato Auriemma
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta” Politecnico di Milano Via Mancinelli 7 Milano 20131 Italy
| | - Mattia Sponchioni
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta” Politecnico di Milano Via Mancinelli 7 Milano 20131 Italy
| | - Umberto Capasso Palmiero
- Department of Chemistry and Applied Biosciences ETH Zürich Vladimir‐Prelog‐Weg 1‐5/10 Zürich 8093 Switzerland
| | - Nicolò Manfredini
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta” Politecnico di Milano Via Mancinelli 7 Milano 20131 Italy
| | - Fatemeh Sadat Razavi Dinani
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta” Politecnico di Milano Via Mancinelli 7 Milano 20131 Italy
| | - Davide Moscatelli
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta” Politecnico di Milano Via Mancinelli 7 Milano 20131 Italy
| |
Collapse
|
29
|
Shen K, Sun G, Chan L, He L, Li X, Yang S, Wang B, Zhang H, Huang J, Chang M, Li Z, Chen T. Anti-Inflammatory Nanotherapeutics by Targeting Matrix Metalloproteinases for Immunotherapy of Spinal Cord Injury. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2102102. [PMID: 34510724 DOI: 10.1002/smll.202102102] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/26/2021] [Indexed: 05/24/2023]
Abstract
Neuroinflammation is critically involved in the repair of spinal cord injury (SCI), and macrophages associated with inflammation propel the degeneration or recovery in the pathological process. Currently, efforts have been focused on obtaining efficient therapeutic anti-inflammatory drugs to treat SCI. However, these drugs are still unable to penetrate the blood spinal cord barrier and lack the ability to target lesion areas, resulting in unsatisfactory clinical efficacy. Herein, a polymer-based nanodrug delivery system is constructed to enhance the targeting ability. Because of increased expression of matrix metalloproteinases (MMPs) in injured site after SCI, MMP-responsive molecule, activated cell-penetrating peptides (ACPP), is introduced into the biocompatible polymer PLGA-PEI-mPEG (PPP) to endow the nanoparticles with the ability for diseased tissue-targeting. Meanwhile, etanercept (ET), a clinical anti-inflammation treatment medicine, is loaded on the polymer to regulate the polarization of macrophages, and promote locomotor recovery. The results show that PPP-ACPP nanoparticles possess satisfactory lesion targeting effects. Through inhibited consequential production of proinflammation cytokines and promoted anti-inflammation cytokines, ET@PPP-ACPP could decrease the percentage of M1 macrophages and increase M2 macrophages. As expected, ET@PPP-ACPP accumulates in lesion area and achieves effective treatment of SCI; this confirmed the potential of nano-drug loading systems in SCI immunotherapy.
Collapse
Affiliation(s)
- Kui Shen
- Department of Orthopedics, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Guodong Sun
- Department of Orthopedics, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou, Guangdong, 510632, China
- The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, 517000, China
| | - Leung Chan
- Department of Orthopedics, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Lizhen He
- Department of Orthopedics, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Xiaowei Li
- Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong, 519000, P. R. China
- The Biomedical Translational Research Institute, Jinan University Faculty of Medical Science, Jinan University, Guangzhou, 510632, P. R. China
| | - Shuxian Yang
- Department of Orthopedics, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou, Guangdong, 510632, China
- The Biomedical Translational Research Institute, Jinan University Faculty of Medical Science, Jinan University, Guangzhou, 510632, P. R. China
| | - Baocheng Wang
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, 518055, China
| | - Hua Zhang
- Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong, 519000, P. R. China
- The Biomedical Translational Research Institute, Jinan University Faculty of Medical Science, Jinan University, Guangzhou, 510632, P. R. China
| | - Jiarun Huang
- Department of Orthopedics, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Minmin Chang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Zhizhong Li
- Department of Orthopedics, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou, Guangdong, 510632, China
- The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, 517000, China
| | - Tianfeng Chen
- Department of Orthopedics, The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou, Guangdong, 510632, China
| |
Collapse
|
30
|
Auriemma R, Sponchioni M, Lotti S, Morosi L, Zucchetti M, Lupi M, Moscatelli D, Capasso Palmiero U. Preformed Biodegradable Zwitterionic Nanoparticles as Tunable Excipients for the Formulation of Therapeutics Directly at the Point of Care. Ind Eng Chem Res 2021. [DOI: 10.1021/acs.iecr.1c00238] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Renato Auriemma
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milano, 20131, Italy
| | - Mattia Sponchioni
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milano, 20131, Italy
| | - Sophia Lotti
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milano, 20131, Italy
| | - Lavinia Morosi
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, 20156, Italy
| | - Massimo Zucchetti
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, 20156, Italy
| | - Monica Lupi
- Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano, 20156, Italy
| | - Davide Moscatelli
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Milano, 20131, Italy
| | - Umberto Capasso Palmiero
- Department of Chemistry and Applied Biosciences, Institute for Chemical and Bioengineering, ETH Zürich, Zürich, 8093, Switzerland
| |
Collapse
|
31
|
Auriemma R, Sponchioni M, Capasso Palmiero U, Rossino G, Rossetti A, Marsala A, Collina S, Sacchetti A, Moscatelli D, Peviani M. Synthesis and Characterization of a "Clickable" PBR28 TSPO-Selective Ligand Derivative Suitable for the Functionalization of Biodegradable Polymer Nanoparticles. NANOMATERIALS 2021; 11:nano11071693. [PMID: 34203263 PMCID: PMC8308144 DOI: 10.3390/nano11071693] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/21/2021] [Accepted: 06/22/2021] [Indexed: 02/04/2023]
Abstract
Reactive microgliosis is a pathological hallmark that accompanies neuronal demise in many neurodegenerative diseases, ranging from acute brain/spinal cord injuries to chronic diseases, such as amyotrophic lateral sclerosis (ALS), Alzheimer’s disease (AD) and age-related dementia. One strategy to assess and monitor microgliosis is to use positron emission tomography (PET) by exploiting radioligands selective for the 18 kDa translocator protein (TSPO) which is highly upregulated in the brain in pathological conditions. Several TSPO ligands have been developed and validated, so far. Among these, PBR28 has been widely adopted for PET imaging at both preclinical and clinical levels, thanks to its high brain penetration and high selectivity. For this reason, PBR28 represents a good candidate for functionalization strategies, where this ligand could be exploited to drive selective targeting of TSPO-expressing cells. Since the PBR28 structure lacks functional moieties that could be exploited for derivatization, in this work we explored a synthetic pathway for the synthesis of a PBR28 derivative carrying an alkyne group (PBR-alkyne), enabling the fast conjugation of the ligand through azide-alkyne cycloaddition, also known as click-chemistry. As a proof of concept, we demonstrated in silico that the derivatized PBR28 ligand maintains the capability to fit into the TSPO binding pocked, and we successfully exploited PBR-alkyne to decorate zwitterionic biodegradable polymer nanoparticles (NPs) resulting in efficient internalization in cultured microglia-like cell lines.
Collapse
Affiliation(s)
- Renato Auriemma
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy; (R.A.); (A.R.); (A.S.); (D.M.)
| | - Mattia Sponchioni
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy; (R.A.); (A.R.); (A.S.); (D.M.)
- Correspondence: (M.S.); (M.P.)
| | - Umberto Capasso Palmiero
- Department of Chemistry and Applied Biosciences, ETH Zürich, Vladimir-Prelog-Weg 1-5/10, 8093 Zürich, Switzerland;
| | - Giacomo Rossino
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (G.R.); (S.C.)
| | - Arianna Rossetti
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy; (R.A.); (A.R.); (A.S.); (D.M.)
| | - Andrea Marsala
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy;
| | - Simona Collina
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (G.R.); (S.C.)
| | - Alessandro Sacchetti
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy; (R.A.); (A.R.); (A.S.); (D.M.)
| | - Davide Moscatelli
- Department of Chemistry, Materials and Chemical Engineering “Giulio Natta”, Politecnico di Milano, Via Mancinelli 7, 20131 Milano, Italy; (R.A.); (A.R.); (A.S.); (D.M.)
| | - Marco Peviani
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, Via Ferrata 9, 27100 Pavia, Italy;
- Gene Therapy Program, Dana Farber/Boston Children’s Cancer and Blood Disorders Center, 450 Brookline Ave., Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02115, USA
- Correspondence: (M.S.); (M.P.)
| |
Collapse
|
32
|
Islam Y, Leach AG, Smith J, Pluchino S, Coxon CR, Sivakumaran M, Downing J, Fatokun AA, Teixidò M, Ehtezazi T. Physiological and Pathological Factors Affecting Drug Delivery to the Brain by Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2002085. [PMID: 34105297 PMCID: PMC8188209 DOI: 10.1002/advs.202002085] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/06/2021] [Indexed: 05/04/2023]
Abstract
The prevalence of neurological/neurodegenerative diseases, such as Alzheimer's disease is known to be increasing due to an aging population and is anticipated to further grow in the decades ahead. The treatment of brain diseases is challenging partly due to the inaccessibility of therapeutic agents to the brain. An increasingly important observation is that the physiology of the brain alters during many brain diseases, and aging adds even more to the complexity of the disease. There is a notion that the permeability of the blood-brain barrier (BBB) increases with aging or disease, however, the body has a defense mechanism that still retains the separation of the brain from harmful chemicals in the blood. This makes drug delivery to the diseased brain, even more challenging and complex task. Here, the physiological changes to the diseased brain and aged brain are covered in the context of drug delivery to the brain using nanoparticles. Also, recent and novel approaches are discussed for the delivery of therapeutic agents to the diseased brain using nanoparticle based or magnetic resonance imaging guided systems. Furthermore, the complement activation, toxicity, and immunogenicity of brain targeting nanoparticles as well as novel in vitro BBB models are discussed.
Collapse
Affiliation(s)
- Yamir Islam
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Andrew G. Leach
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- Division of Pharmacy and OptometryThe University of ManchesterStopford Building, Oxford RoadManchesterM13 9PTUK
| | - Jayden Smith
- Cambridge Innovation Technologies Consulting (CITC) LimitedSt. John's Innovation CentreCowley RoadCambridgeCB4 0WSUK
| | - Stefano Pluchino
- Department of Clinical NeurosciencesClifford Allbutt Building – Cambridge Biosciences Campus and NIHR Biomedical Research CentreUniversity of CambridgeHills RoadCambridgeCB2 0HAUK
| | - Christopher R. Coxon
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- School of Engineering and Physical SciencesHeriot‐Watt UniversityWilliam Perkin BuildingEdinburghEH14 4ASUK
| | - Muttuswamy Sivakumaran
- Department of HaematologyPeterborough City HospitalEdith Cavell CampusBretton Gate PeterboroughPeterboroughPE3 9GZUK
| | - James Downing
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Amos A. Fatokun
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Meritxell Teixidò
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)Baldiri Reixac 10Barcelona08028Spain
| | - Touraj Ehtezazi
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| |
Collapse
|
33
|
Khan NH, Mir M, Ngowi EE, Zafar U, Khakwani MMAK, Khattak S, Zhai YK, Jiang ES, Zheng M, Duan SF, Wei JS, Wu DD, Ji XY. Nanomedicine: A Promising Way to Manage Alzheimer's Disease. Front Bioeng Biotechnol 2021; 9:630055. [PMID: 33996777 PMCID: PMC8120897 DOI: 10.3389/fbioe.2021.630055] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/08/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is a devastating disease of the aging population characterized by the progressive and slow brain decay due to the formation of extracellular plaques in the hippocampus. AD cells encompass tangles of twisted strands of aggregated microtubule binding proteins surrounded by plaques. Delivering corresponding drugs in the brain to deal with these clinical pathologies, we face a naturally built strong, protective barrier between circulating blood and brain cells called the blood-brain barrier (BBB). Nanomedicines provide state-of-the-art alternative approaches to overcome the challenges in drug transport across the BBB. The current review presents the advances in the roles of nanomedicines in both the diagnosis and treatment of AD. We intend to provide an overview of how nanotechnology has revolutionized the approaches used to manage AD and highlight the current key bottlenecks and future perspective in this field. Furthermore, the emerging nanomedicines for managing brain diseases like AD could promote the booming growth of research and their clinical availability.
Collapse
Affiliation(s)
- Nazeer Hussain Khan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Maria Mir
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
| | - Ebenezeri Erasto Ngowi
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Department of Biological Sciences, Faculty of Sciences, Dar es Salaam University College of Education, Dar es Salaam, Tanzania
| | - Ujala Zafar
- School of Natural Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | | | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Yuan-Kun Zhai
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
- School of Stomatology, Henan University, Kaifeng, China
| | - En-She Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Institutes of Nursing and Health, School of Nursing and Health, Henan University, Kaifeng, China
| | - Meng Zheng
- International Joint Center for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, China
| | - Shao-Feng Duan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Institute for Innovative Drug Design and Evaluation, School of Pharmacy, Henan University, Kaifeng, China
| | - Jian-She Wei
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Brain Research Laboratory, School of Life Sciences, Henan University, Kaifeng, China
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
- School of Stomatology, Henan University, Kaifeng, China
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences, Henan University, Kaifeng, China
| |
Collapse
|
34
|
Janjua TI, Rewatkar P, Ahmed-Cox A, Saeed I, Mansfeld FM, Kulshreshtha R, Kumeria T, Ziegler DS, Kavallaris M, Mazzieri R, Popat A. Frontiers in the treatment of glioblastoma: Past, present and emerging. Adv Drug Deliv Rev 2021; 171:108-138. [PMID: 33486006 DOI: 10.1016/j.addr.2021.01.012] [Citation(s) in RCA: 142] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/13/2020] [Accepted: 01/09/2021] [Indexed: 12/13/2022]
Abstract
Glioblastoma (GBM) is one of the most aggressive cancers of the brain. Despite extensive research over the last several decades, the survival rates for GBM have not improved and prognosis remains poor. To date, only a few therapies are approved for the treatment of GBM with the main reasons being: 1) significant tumour heterogeneity which promotes the selection of resistant subpopulations 2) GBM induced immunosuppression and 3) fortified location of the tumour in the brain which hinders the delivery of therapeutics. Existing therapies for GBM such as radiotherapy, surgery and chemotherapy have been unable to reach the clinical efficacy necessary to prolong patient survival more than a few months. This comprehensive review evaluates the current and emerging therapies including those in clinical trials that may potentially improve both targeted delivery of therapeutics directly to the tumour site and the development of agents that may specifically target GBM. Particular focus has also been given to emerging delivery technologies such as focused ultrasound, cellular delivery systems nanomedicines and immunotherapy. Finally, we discuss the importance of developing novel materials for improved delivery efficacy of nanoparticles and therapeutics to reduce the suffering of GBM patients.
Collapse
|
35
|
Lipophilic dye-compatible brain clearing technique allowing correlative magnetic resonance/high-resolution fluorescence imaging in rat models of glioblastoma. Sci Rep 2020; 10:17974. [PMID: 33087842 PMCID: PMC7578790 DOI: 10.1038/s41598-020-75137-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/09/2020] [Indexed: 12/12/2022] Open
Abstract
In this work we optimized a novel approach for combining in vivo MRI and ex vivo high-resolution fluorescence microscopy that involves: (i) a method for slicing rat brain tissue into sections with the same thickness and spatial orientation as in in vivo MRI, to better correlate in vivo MRI analyses with ex-vivo imaging via scanning confocal microscope and (ii) an improved clearing protocol compatible with lipophilic dyes that highlight the neurovascular network, to obtain high tissue transparency while preserving tissue staining and morphology with no significant tissue shrinkage or expansion. We applied this methodology in two rat models of glioblastoma (GBM; U87 human glioma cells and patient-derived human glioblastoma cancer stem cells) to demonstrate how vital the information retrieved from the correlation between MRI and confocal images is and to highlight how the increased invasiveness of xenografts derived from cancer stem cells may not be clearly detected by standard in vivo MRI approaches. The protocol studied in this work could be implemented in pre-clinical GBM research to further the development and validation of more predictive and translatable MR imaging protocols that can be used as critical diagnostic and prognostic tools. The development of this protocol is part of the quest for more efficacious treatment approaches for this devastating and still uncurable disease. In particular, this approach could be instrumental in validating novel MRI-based techniques to assess cellular infiltration beyond the macroscopic tumor margins and to quantify neo-angiogenesis.
Collapse
|
36
|
Cerqueira SR, Ayad NG, Lee JK. Neuroinflammation Treatment via Targeted Delivery of Nanoparticles. Front Cell Neurosci 2020; 14:576037. [PMID: 33192321 PMCID: PMC7555434 DOI: 10.3389/fncel.2020.576037] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022] Open
Abstract
The lack of effective treatments for most neurological diseases has prompted the search for novel therapeutic options. Interestingly, neuroinflammation is emerging as a common feature to target in most CNS pathologies. Recent studies suggest that targeted delivery of small molecules to reduce neuroinflammation can be beneficial. However, suboptimal drug delivery to the CNS is a major barrier to modulate inflammation because neurotherapeutic compounds are currently being delivered systemically without spatial or temporal control. Emerging nanomaterial technologies are providing promising and superior tools to effectively access neuropathological tissue in a controlled manner. Here we highlight recent advances in nanomaterial technologies for drug delivery to the CNS. We propose that state-of-the-art nanoparticle drug delivery platforms can significantly impact local CNS bioavailability of pharmacological compounds and treat neurological diseases.
Collapse
Affiliation(s)
- Susana R Cerqueira
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States.,The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Nagi G Ayad
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States.,The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, United States.,Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Jae K Lee
- Department of Neurological Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States.,The Miami Project to Cure Paralysis, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
37
|
Zhao N, Francis NL, Calvelli HR, Moghe PV. Microglia-targeting nanotherapeutics for neurodegenerative diseases. APL Bioeng 2020; 4:030902. [PMID: 32923843 PMCID: PMC7481010 DOI: 10.1063/5.0013178] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022] Open
Abstract
Advances in nanotechnology have enabled the design of nanotherapeutic platforms that could address the challenges of targeted delivery of active therapeutic agents to the central nervous system (CNS). While the majority of previous research studies on CNS nanotherapeutics have focused on neurons and endothelial cells, the predominant resident immune cells of the CNS, microglia, are also emerging as a promising cellular target for neurodegeneration considering their prominent role in neuroinflammation. Under normal physiological conditions, microglia protect neurons by removing pathological agents. However, long-term exposure of microglia to stimulants will cause sustained activation and lead to neuronal damage due to the release of pro-inflammatory agents, resulting in neuroinflammation and neurodegeneration. This Perspective highlights criteria to be considered when designing microglia-targeting nanotherapeutics for the treatment of neurodegenerative disorders. These criteria include conjugating specific microglial receptor-targeting ligands or peptides to the nanoparticle surface to achieve targeted delivery, leveraging microglial phagocytic properties, and utilizing biocompatible and biodegradable nanomaterials with low immune reactivity and neurotoxicity. In addition, certain therapeutic agents for the controlled inhibition of toxic protein aggregation and for modulation of microglial activation pathways can also be incorporated within the nanoparticle structure without compromising stability. Overall, considering the multifaceted disease mechanisms of neurodegeneration, microglia-targeted nanodrugs and nanotherapeutic particles may have the potential to resolve multiple pathological determinants of the disease and to guide a shift in the microglial phenotype spectrum toward a more neuroprotective state.
Collapse
Affiliation(s)
- Nanxia Zhao
- Department of Chemical and Biochemical Engineering, 98 Brett Rd., Rutgers University, Piscataway, New Jersey 08854, USA
| | - Nicola L. Francis
- Department of Biomedical Engineering, 599 Taylor Rd., Rutgers University, Piscataway, New Jersey 08854, USA
| | - Hannah R. Calvelli
- Department of Molecular Biology and Biochemistry, 604 Allison Rd., Rutgers University, Piscataway, New Jersey 08854, USA
| | | |
Collapse
|
38
|
Taking advantage of cellular uptake of ferritin nanocages for targeted drug delivery. J Control Release 2020; 325:176-190. [DOI: 10.1016/j.jconrel.2020.06.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/16/2022]
|
39
|
Phạm TL, Kim DW. Poly(lactic-co-glycolic acid) nanomaterial-based treatment options for pain management: a review. Nanomedicine (Lond) 2020; 15:1897-1913. [PMID: 32757701 DOI: 10.2217/nnm-2020-0114] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neuropathic pain is one of the most intense types of chronic pain; it constitutes a pervasive complaint throughout the public health system. With few effective treatments, it remains a significant challenge. Commercially available drugs for neuropathic pain are still limited and have disappointing efficacy. Therefore, chronic neuropathic pain imposes a tremendous burden on patients' quality of life. Recently, the introduction and application of nanotechnology in multiple fields has accelerated the development of new drugs. This review highlights the application of poly(lactic-co-glycolic acid) nanomaterial-based vehicles for drug delivery and how they improve the therapeutic outcomes for neuropathic pain treatment. Finally, future developments for pain research and effective management are presented.
Collapse
Affiliation(s)
- Thuỳ Linh Phạm
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Anatomy, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Histology & Embryology, Hai Phong University of Medicine & Pharmacy Hospital, Hai Phong, 042-12, Vietnam
| | - Dong Woon Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
- Department of Anatomy, Brain Research Institute, Chungnam National University College of Medicine, Daejeon, 35015, Republic of Korea
| |
Collapse
|
40
|
Ren C, Li D, Zhou Q, Hu X. Mitochondria-targeted TPP-MoS 2 with dual enzyme activity provides efficient neuroprotection through M1/M2 microglial polarization in an Alzheimer's disease model. Biomaterials 2019; 232:119752. [PMID: 31923845 DOI: 10.1016/j.biomaterials.2019.119752] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/07/2019] [Accepted: 12/29/2019] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is one of the most common age-associated brain diseases and is induced by the accumulation of amyloid beta (Aβ) and oxidative stress. Many studies have focused on eliminating Aβ by nanoparticle affinity; however, nanoparticles are taken up mainly by microglia rather than neurons, leading poor control of AD. Herein, mitochondria-targeted nanozymes known as (3-carboxypropyl)triphenyl-phosphonium bromide-conjugated 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000]-functionalized molybdenum disulfide quantum dots (TPP-MoS2 QDs) were designed. TPP-MoS2 QDs mitigate Aβ aggregate-mediated neurotoxicity and eliminate Aβ aggregates in AD mice by switching microglia from the proinflammatory M1 phenotype to the anti-inflammatory M2 phenotype. TPP-MoS2 QDs cross the blood-brain barrier, escape from lysosomes, target mitochondria and exhibit the comprehensive activity of a bifunctional nanozyme, thus preventing spontaneous neuroinflammation by regulating the proinflammatory substances interleukin-1β, interleukin-6 and tumor necrosis factors as well as the anti-inflammatory substance transforming growth factor-β. In contrast to the low efficacy of eliminating Aβ by nanoparticle affinity, the present study provides a new pathway to mitigate AD pathology through mitochondria-targeted nanozymes and M1/M2 microglial polarization.
Collapse
Affiliation(s)
- Chaoxiu Ren
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education), Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China
| | - Dandan Li
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education), Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China
| | - Qixing Zhou
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education), Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China
| | - Xiangang Hu
- Key Laboratory of Pollution Processes and Environmental Criteria (Ministry of Education), Tianjin Key Laboratory of Environmental Remediation and Pollution Control, College of Environmental Science and Engineering, Nankai University, Tianjin, 300350, China.
| |
Collapse
|
41
|
Gonda A, Zhao N, Shah JV, Calvelli HR, Kantamneni H, Francis NL, Ganapathy V. Engineering Tumor-Targeting Nanoparticles as Vehicles for Precision Nanomedicine. MED ONE 2019; 4:e190021. [PMID: 31592196 PMCID: PMC6779336 DOI: 10.20900/mo.20190021] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
As a nascent and emerging field that holds great potential for precision oncology, nanotechnology has been envisioned to improve drug delivery and imaging capabilities through precise and efficient tumor targeting, safely sparing healthy normal tissue. In the clinic, nanoparticle formulations such as the first-generation Abraxane® in breast cancer, Doxil® for sarcoma, and Onivyde® for metastatic pancreatic cancer, have shown advancement in drug delivery while improving safety profiles. However, effective accumulation of nanoparticles at the tumor site is sub-optimal due to biological barriers that must be overcome. Nanoparticle delivery and retention can be altered through systematic design considerations in order to enhance passive accumulation or active targeting to the tumor site. In tumor niches where passive targeting is possible, modifications in the size and charge of nanoparticles play a role in their tissue accumulation. For niches in which active targeting is required, precision oncology research has identified targetable biomarkers, with which nanoparticle design can be altered through bioconjugation using antibodies, peptides, or small molecule agonists and antagonists. This review is structured to provide a better understanding of nanoparticle engineering design principles with emphasis on overcoming tumor-specific biological barriers.
Collapse
Affiliation(s)
- Amber Gonda
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ 08854, USA
| | - Nanxia Zhao
- Department of Chemical and Biochemical Engineering, Rutgers, The State University of New Jersey, 98 Brett Road, Piscataway, NJ 08854, USA
| | - Jay V. Shah
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ 08854, USA
| | - Hannah R. Calvelli
- Department of Molecular Biology and Biochemistry, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854, USA
| | - Harini Kantamneni
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ 08854, USA
| | - Nicola L. Francis
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ 08854, USA
| | - Vidya Ganapathy
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Road, Piscataway, NJ 08854, USA
| |
Collapse
|
42
|
Sitia L, Catelani T, Guarnieri D, Pompa PP. In Vitro Blood-Brain Barrier Models for Nanomedicine: Particle-Specific Effects and Methodological Drawbacks. ACS APPLIED BIO MATERIALS 2019; 2:3279-3289. [PMID: 35030770 DOI: 10.1021/acsabm.9b00305] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Predicting the therapeutic efficacy of a nanocarrier, in a rapid and cost-effective way, is pivotal for the drug delivery to the central nervous system (CNS). In this context, in vitro testing platforms, like the transwell systems, offer numerous advantages to study the passage through the blood-brain barrier (BBB), such as overcoming ethical and methodological issues of in vivo models. However, the use of different transwell filters and nanocarriers with various physical-chemical features makes it difficult to assess the nanocarrier efficacy and achieve data reproducibility. In this work, we performed a systematic study to elucidate the role of the most widely used transwell filters in affecting the passage of nanocarriers, as a function of filter pore size and density. In particular, the transport of carboxyl- and amine-modified 100 nm polystyrene nanoparticles (NPs), chosen as model nanocarriers, was quantified and compared to the behavior of Lucifer yellow (LY), a molecular marker of paracellular transport. Results indicate that the filter type affects the growth and formation of the confluent endothelial barrier, as well as the transport of NPs. Interestingly, the in situ dispersion of NPs was found to play a key role in governing their passage through the filters, both in absence and in presence of the cellular barrier. By framing the underlying nanobiointeractions, we found that particle-specific effects modulated cellular uptake and barrier intracellular distribution, eventually governing transcytosis through their interplay with "size exclusion effects" by the porous filters. This study highlights the importance of a careful evaluation of the physical-chemical profile of the tested nanocarrier along with filter parameters for a correct methodological approach to test BBB permeability in nanomedicine.
Collapse
Affiliation(s)
- Leopoldo Sitia
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia (IIT), via Morego 30, Genova 16163, Italy.,Department of Biomedical and Clinical Sciences "L. Sacco″, Università Degli Studi di Milano, via G. B. Grassi 74, Milano 20157, Italy
| | - Tiziano Catelani
- Electron Microscopy Facility, Istituto Italiano di Tecnologia, via Morego 30, Genova 16163, Italy.,Piattaforma Interdipartimentale di Microscopia, Università Degli Studi di Milano-Bicocca, Piazza della Scienza 2, Milano 20126, Italy
| | - Daniela Guarnieri
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia (IIT), via Morego 30, Genova 16163, Italy.,Dipartimento di Chimica e Biologia "A. Zambelli", Università di Salerno, via Giovanni Paolo II 132, Fisciano, Salerno I-84084, Italy
| | - Pier Paolo Pompa
- Nanobiointeractions & Nanodiagnostics, Istituto Italiano di Tecnologia (IIT), via Morego 30, Genova 16163, Italy
| |
Collapse
|