1
|
Liu Chung Ming C, Patil R, Refaat A, Lal S, Wang X, Gentile C. Acetylcholine-loaded nanoparticles protect against doxorubicin-induced toxicity in in vitrocardiac spheroids. Biofabrication 2025; 17:025023. [PMID: 39965540 DOI: 10.1088/1758-5090/adb7c2] [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: 10/09/2024] [Accepted: 02/18/2025] [Indexed: 02/20/2025]
Abstract
Doxorubicin (DOX) is widely used in chemotherapy, yet it significantly contributes to heart failure-associated death. Acetylcholine (ACh) is cardioprotective by enhancing heart rate variability and reducing mitochondrial dysfunction and inflammation. Nonetheless, the protective role of ACh in countering DOX-induced cardiotoxicity (DIC) remains underexplored as current approaches to increasing ACh levels are invasive and unsafe for patients. In this study, we explore the protective effects of ACh against DIC through three distinct ACh administration strategies: (i) freely-suspended 100µM ACh; (ii) ACh-producing cholinergic neurons (CNs); or (iii) ACh-loaded nanoparticles (ACh-NPs). These are tested inin vitrocardiac spheroids (CSs), which have previously been shown to approximate the complex DIC. We assess ACh's protective effects by measuring the toxicity ratio (cell death/viability), contractile activity, gene expression changes via qPCR and nitric oxide (NO) signaling. Our findings show that ACh effectively attenuates DOX-induced cell death and contractile dysfunction. ACh also counteracts the DOX-induced downregulation of genes controlling myocardial fibrosis, endothelial and cardiomyocyte dysfunction, and autonomic dysregulation. ACh cardioprotection against DOX is dependent on NO signaling in endothelial cells but not in cardiac myocytes or fibroblasts. Altogether, this study shows for the first time that elevating ACh levels showed a promising therapeutic approach for preventing DIC.
Collapse
Affiliation(s)
- Clara Liu Chung Ming
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
- Cardiovascular Regeneration Group, Heart Research Institute, Newtown, NSW 2042, Australia
| | - Runali Patil
- IIT-Bombay Monash Research Academy, IIT Bombay, Powai, Mumbai, Maharashtra 400076, India
- Department of Medicine, Monash University, Melbourne, VIC 3800, Australia
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Ahmed Refaat
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC 3052, Australia
| | - Sean Lal
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW 2050, Australia
| | - Xiaowei Wang
- Department of Medicine, Monash University, Melbourne, VIC 3800, Australia
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC 3004, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Carmine Gentile
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Sydney, NSW, Australia
- Cardiovascular Regeneration Group, Heart Research Institute, Newtown, NSW 2042, Australia
| |
Collapse
|
2
|
Mazahir F, Alam MI, Yadav AK. Development of nanomedicines for the treatment of Alzheimer's disease: Raison d'être, strategies, challenges and regulatory aspects. Ageing Res Rev 2024; 98:102318. [PMID: 38705362 DOI: 10.1016/j.arr.2024.102318] [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: 02/14/2024] [Revised: 04/04/2024] [Accepted: 04/27/2024] [Indexed: 05/07/2024]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder characterized by progressive loss of memory. Presently, AD is challenging to treat with current drug therapy as their delivery to the brain is restricted by the presence of the blood-brain barrier. Nanomedicines, due to their size, high surface volume ratio, and ease of tailoring drug release characteristics, showed their potential to treat AD. The nanotechnology-based formulations for brain targeting are expected to enter the market in the near future. So, regulatory frameworks are required to ensure the quality, safety, and effectiveness of the nanomedicines to treat AD. In this review, we discuss different strategies, in-vitro blood-brain permeation models, in-vivo permeation assessment, and regulatory aspects for the development of nanomedicine to treat AD.
Collapse
Affiliation(s)
- Farhan Mazahir
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Raebareli, India
| | - Md Imtiyaz Alam
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Raebareli, India
| | - Awesh Kumar Yadav
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Raebareli, India.
| |
Collapse
|
3
|
Su W, Xu W, Liu E, Su W, Polyakov NE. Improving the Treatment Effect of Carotenoids on Alzheimer's Disease through Various Nano-Delivery Systems. Int J Mol Sci 2023; 24:ijms24087652. [PMID: 37108814 PMCID: PMC10142927 DOI: 10.3390/ijms24087652] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/10/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023] Open
Abstract
Natural bioactive compounds have recently emerged as a current strategy for Alzheimer's disease treatment. Carotenoids, including astaxanthin, lycopene, lutein, fucoxanthin, crocin and others are natural pigments and antioxidants, and can be used to treat a variety of diseases, including Alzheimer's disease. However, carotenoids, as oil-soluble substances with additional unsaturated groups, suffer from low solubility, poor stability and poor bioavailability. Therefore, the preparation of various nano-drug delivery systems from carotenoids is a current measure to achieve efficient application of carotenoids. Different carotenoid delivery systems can improve the solubility, stability, permeability and bioavailability of carotenoids to a certain extent to achieve Alzheimer's disease efficacy. This review summarizes recent data on different carotenoid nano-drug delivery systems for the treatment of Alzheimer's disease, including polymer, lipid, inorganic and hybrid nano-drug delivery systems. These drug delivery systems have been shown to have a beneficial therapeutic effect on Alzheimer's disease to a certain extent.
Collapse
Affiliation(s)
- Wenjing Su
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wenhao Xu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Enshuo Liu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Weike Su
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Nikolay E Polyakov
- Institute of Solid State Chemistry and Mechanochemistry, 630128 Novosibirsk, Russia
- Institute of Chemical Kinetics and Combustion, 630090 Novosibirsk, Russia
| |
Collapse
|
4
|
Shala AL, Arduino I, Salihu MB, Denora N. Quercetin and Its Nano-Formulations for Brain Tumor Therapy—Current Developments and Future Perspectives for Paediatric Studies. Pharmaceutics 2023; 15:pharmaceutics15030963. [PMID: 36986827 PMCID: PMC10057501 DOI: 10.3390/pharmaceutics15030963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 03/10/2023] [Accepted: 03/14/2023] [Indexed: 03/19/2023] Open
Abstract
The development of efficient treatments for tumors affecting the central nervous system (CNS) remains an open challenge. Particularly, gliomas are the most malignant and lethal form of brain tumors in adults, causing death in patients just over 6 months after diagnosis without treatment. The current treatment protocol consists of surgery, followed using synthetic drugs and radiation. However, the efficacy of these protocols is associated with side effects, poor prognosis and with a median survival of fewer than two years. Recently, many studies were focused on applying plant-derived products to manage various diseases, including brain cancers. Quercetin is a bioactive compound derived from various fruits and vegetables (asparagus, apples, berries, cherries, onions and red leaf lettuce). Numerous in vivo and in vitro studies highlighted that quercetin through multitargeted molecular mechanisms (apoptosis, necrosis, anti-proliferative activity and suppression of tumor invasion and migration) effectively reduces the progression of tumor cells. This review aims to summarize current developments and recent advances of quercetin’s anticancer potential in brain tumors. Since all reported studies demonstrating the anti-cancer potential of quercetin were conducted using adult models, it is suggested to expand further research in the field of paediatrics. This could offer new perspectives on brain cancer treatment for paediatric patients.
Collapse
Affiliation(s)
- Aida Loshaj Shala
- Department of Drug Analysis and Pharmaceutical Technology, Faculty of Medicine, University of Prishtina, 10000 Prishtina, Kosovo
| | - Ilaria Arduino
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari “Aldo Moro”, Orabona St. 4, 70125 Bari, Italy
| | - Mimoza Basholli Salihu
- Department of Drug Analysis and Pharmaceutical Technology, Faculty of Medicine, University of Prishtina, 10000 Prishtina, Kosovo
| | - Nunzio Denora
- Department of Pharmacy—Pharmaceutical Sciences, University of Bari “Aldo Moro”, Orabona St. 4, 70125 Bari, Italy
- Correspondence:
| |
Collapse
|
5
|
Angolkar M, Paramshetti S, Halagali P, Jain V, Patil AB, Somanna P. Nanotechnological advancements in the brain tumor therapy: a novel approach. Ther Deliv 2023; 13:531-557. [PMID: 36802944 DOI: 10.4155/tde-2022-0035] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Nanotechnological advancements over the past few years have led to the development of newer treatment strategies in brain cancer therapy which leads to the establishment of nano oncology. Nanostructures with high specificity, are best suitable to penetrate the blood-brain barrier (BBB). Their desired physicochemical properties, such as small sizes, shape, higher surface area to volume ratio, distinctive structural features, and the possibility to attach various substances on their surface transform them into potential transport carriers able to cross various cellular and tissue barriers, including the BBB. The review emphasizes nanotechnology-based treatment strategies for the exploration of brain tumors and highlights the current progress of different nanomaterials for the effective delivery of drugs for brain tumor therapy.
Collapse
Affiliation(s)
- Mohit Angolkar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Sharanya Paramshetti
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Praveen Halagali
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Amit B Patil
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Preethi Somanna
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| |
Collapse
|
6
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
7
|
Gratpain V, Mwema A, Labrak Y, Muccioli GG, van Pesch V, des Rieux A. Extracellular vesicles for the treatment of central nervous system diseases. Adv Drug Deliv Rev 2021; 174:535-552. [PMID: 33991589 DOI: 10.1016/j.addr.2021.05.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/14/2021] [Accepted: 05/11/2021] [Indexed: 02/07/2023]
Abstract
The interest in extracellular vesicles (EVs) increased during the last decade. It is now established that these vesicles play a role in the pathogenesis of central nervous system diseases (CNS), which explains why they are studied as biomarkers in these pathologies. On the other hand, EVs can also present therapeutic properties, often similar to their parent cells, as observed with mesenchymal stem cell-derived EVs. They can then be used as therapeutics, alone or combined with a bioactive molecule, for the treatment of CNS diseases, as they can cross the blood-brain barrier more easily than synthetic nanomedicines and are less immunogenic. A few clinical trials are currently on-going but there are still challenges to overcome for further clinical translation such as the scale-up of the production, the lack of standardization for isolation and characterization methods and the low encapsulation efficiency.
Collapse
Affiliation(s)
- Viridiane Gratpain
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium
| | - Ariane Mwema
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium; Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium
| | - Yasmine Labrak
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium; Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium
| | - Giulio G Muccioli
- Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium
| | - Vincent van Pesch
- Institute of Neuroscience, Neurochemistry Unit, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium; Cliniques Universitaires Saint-Luc, 1200 Brussels, Belgium
| | - Anne des Rieux
- Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Université Catholique de Louvain, UCLouvain, 1200 Brussels, Belgium.
| |
Collapse
|
8
|
You Q, Sabel BA. Nanoparticles as a tool to deliver drugs to the retina and brain: an update. Neural Regen Res 2021; 16:283-284. [PMID: 32859776 PMCID: PMC7896206 DOI: 10.4103/1673-5374.290886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Qing You
- Institute of Medical Psychology, Otto-von-Guericke University of Magdeburg, Medical Faculty, Magdeburg, Germany
| | - Bernhard A Sabel
- Institute of Medical Psychology, Otto-von-Guericke University of Magdeburg, Medical Faculty, Magdeburg, Germany
| |
Collapse
|
9
|
Hartl N, Adams F, Merkel OM. From adsorption to covalent bonding: Apolipoprotein E functionalization of polymeric nanoparticles for drug delivery across the blood-brain barrier. ADVANCED THERAPEUTICS 2021; 4:2000092. [PMID: 33542947 PMCID: PMC7116687 DOI: 10.1002/adtp.202000092] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Indexed: 12/17/2022]
Abstract
The blood-brain barrier (BBB) is composed of brain endothelial cells, pericytes, and astrocytes, which build a tight cellular barrier. Therapeutic (macro)molecules are not able to transit through the BBB in their free form. This limitation is bypassed by apolipoprotein E (ApoE)-functionalized polymeric nanoparticles (NPs) that are able to transport drugs (e.g. dalargin, loperamide, doxorubicin, nerve growth factor) across the BBB via low density lipoprotein (LDL) receptor mediated transcytosis. Coating with polysorbate 80 or poloxamer 188 facilitates ApoE adsorption onto polymeric NPs enabling recognition by LDL receptors of brain endothelial cells. This effect is even enhanced when NPs are directly coated with ApoE without surfactant anchor. Similarly, covalent coupling of ApoE to NPs that bear reactive groups on their surface leads to significantly improved brain uptake while avoiding the use of surfactants. Several in vitro BBB models using brain endothelial cells or co-cultures with astrocytes/pericytes/glioma cells are described which provide first insights regarding the ability of a drug delivery system to cross this barrier. In vivo models are employed to simulate central nervous system-relevant diseases such as Alzheimer's or Parkinson's disease and cerebral cancer.
Collapse
Affiliation(s)
| | | | - Olivia M. Merkel
- Pharmaceutical Technology and Biopharmaceutics, Department Pharmacy, Ludwig-Maximilians-University, Butenandtstr. 5-13, 81377 Munich, Germany
| |
Collapse
|
10
|
Wünsch A, Mulac D, Langer K. Lipoprotein imitating nanoparticles: Lecithin coating binds ApoE and mediates non-lysosomal uptake leading to transcytosis over the blood-brain barrier. Int J Pharm 2020; 589:119821. [DOI: 10.1016/j.ijpharm.2020.119821] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/14/2020] [Accepted: 08/24/2020] [Indexed: 12/29/2022]
|
11
|
Abstract
![]()
Nanocarriers
(NCs) are promising tools to improve drug delivery
across the blood–brain barrier (BBB) for more effective treatment
of brain disorders, although there is a scarcity of clinical translation
of brain-directed NCs. In order to drive the development of brain-oriented
NCs toward clinical success, it is essential to understand the prerequisites
for nanodelivery to be successful in brain treatment. In this Perspective,
we present how pharmacokinetic/pharmacodynamic (PK/PD), formulation
and nanotoxicity factors impact the therapeutic success of brain-specific
nanodelivery. Properties including high loading efficiency, slow in vivo drug release, long systemic circulation, an increase
in unbound brain-to-plasma concentration/exposure ratio (Kp,uu,brain), high drug potency, and minimal nanotoxicity
are prerequisites that should preferably be combined to maximize the
therapeutic potential of a brain-targeted NC. The PK of brain-directed
NCs needs to be evaluated in a more therapeutically relevant manner,
focusing on the released, unbound drug. It is more crucial to increase
the Kp,uu,brain than to improve the ability
of the NC to cross the BBB in its intact form. Brain-targeted NCs,
which are mostly developed for treating brain tumors, including metastases,
should aim to enhance drug delivery not just to tumor regions with
disrupted BBB, but equally important to regions with intact BBB where
the drugs themselves have problems reaching. This article provides
critical insights into how a brain-targeted nanoformulation needs
to be designed and optimized to achieve therapeutic success in the
brain.
Collapse
Affiliation(s)
- Yang Hu
- Translational PKPD Research Group, Department of Pharmacy, Faculty of Pharmacy, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Margareta Hammarlund-Udenaes
- Translational PKPD Research Group, Department of Pharmacy, Faculty of Pharmacy, Uppsala University, SE-751 23 Uppsala, Sweden
| |
Collapse
|
12
|
Abasi S, Bhat A, Guiseppi‐Elie A. Electrode Selection for Electrostimulation and TEER Using ECSARA. ELECTROANAL 2020. [DOI: 10.1002/elan.202060313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Sara Abasi
- Center for Bioelectronics Biosensors and Biochips (C3B®) Department of Biomedical Engineering and Department of Electrical and Computer Engineering Texas A&M University College Station TX 77843 USA
| | - Ankita Bhat
- Center for Bioelectronics Biosensors and Biochips (C3B®) Department of Biomedical Engineering and Department of Electrical and Computer Engineering Texas A&M University College Station TX 77843 USA
| | - Anthony Guiseppi‐Elie
- Center for Bioelectronics Biosensors and Biochips (C3B®) Department of Biomedical Engineering and Department of Electrical and Computer Engineering Texas A&M University College Station TX 77843 USA
- Houston Methodist Institute for Academic Medicine and Houston Methodist Research Institute 6670 Bertner Ave. Houston TX 77030 USA
- Department of Electrical and Computer Engineering Texas A&M University College Station TX 77843 USA
- ABTECH Scientific, Inc. Biotechnology Research Park 800 East Leigh Street Richmond VA 23219 USA
| |
Collapse
|
13
|
You Q, Sokolov M, Grigartzik L, Hintz W, van Wachem BGM, Henrich-Noack P, Sabel BA. How Nanoparticle Physicochemical Parameters Affect Drug Delivery to Cells in the Retina via Systemic Interactions. Mol Pharm 2019; 16:5068-5075. [PMID: 31609624 DOI: 10.1021/acs.molpharmaceut.9b01046] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Minor changes in the composition of poloxamer 188-modified, DEAE-dextran-stabilized (PDD) polybutylcyanoacrylate (PBCA) nanoparticles (NPs), by altering the physicochemical parameters (such as size or surface charge), can substantially influence their delivery kinetics across the blood-retina barrier (BRB) in vivo. We now investigated the physicochemical mechanisms underlying these different behaviors of NP variations at biological barriers and their influence on the cellular and body distribution. Retinal whole mounts from rats injected in vivo with fluorescent PBCA NPs were processed for retina imaging ex vivo to obtain a detailed distribution of NPs with cellular resolution in retinal tissue. In line with previous in vivo imaging results, NPs with a larger size and medium surface charge accumulated more readily in brain tissue, and they could be more easily detected in retinal ganglion cells (RGCs), demonstrating the potential of these NPs for drug delivery into neurons. The biodistribution of the NPs revealed a higher accumulation of small-sized NPs in peripheral organs, which may reduce the passage of these particles into brain tissue via a "steal effect" mechanism. Thus, systemic interactions significantly determine the potential of NPs to deliver markers or drugs to the central nervous system (CNS). In this way, minor changes of NPs' physicochemical parameters can significantly impact their rate of brain/body biodistribution.
Collapse
Affiliation(s)
- Qing You
- Institute of Medical Psychology , Otto-von-Guericke University , Magdeburg 39120 , Germany
| | - Maxim Sokolov
- Institute of Medical Psychology , Otto-von-Guericke University , Magdeburg 39120 , Germany
| | - Lisa Grigartzik
- Institute of Medical Psychology , Otto-von-Guericke University , Magdeburg 39120 , Germany
| | - Werner Hintz
- Institute of Process Engineering , Otto-von-Guericke University , Magdeburg 39106 , Germany
| | - Berend G M van Wachem
- Institute of Process Engineering , Otto-von-Guericke University , Magdeburg 39106 , Germany
| | - Petra Henrich-Noack
- Institute of Medical Psychology , Otto-von-Guericke University , Magdeburg 39120 , Germany.,Clinic of Neurology with Institute of Translational Neurology , University Clinic Münster , Münster 48149 , Germany
| | - Bernhard A Sabel
- Institute of Medical Psychology , Otto-von-Guericke University , Magdeburg 39120 , Germany.,InEye Hospital , Chengdu University of TCM , Chengdu 610084 , PR China
| |
Collapse
|
14
|
Zottel A, Videtič Paska A, Jovčevska I. Nanotechnology Meets Oncology: Nanomaterials in Brain Cancer Research, Diagnosis and Therapy. MATERIALS (BASEL, SWITZERLAND) 2019; 12:1588. [PMID: 31096609 PMCID: PMC6567262 DOI: 10.3390/ma12101588] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 05/08/2019] [Accepted: 05/13/2019] [Indexed: 01/08/2023]
Abstract
Advances in technology of the past decades led to development of new nanometer scale diagnosis and treatment approaches in cancer medicine leading to establishment of nanooncology. Inorganic and organic nanomaterials have been shown to improve bioimaging techniques and targeted drug delivery systems. Their favorable physico-chemical characteristics, like small sizes, large surface area compared to volume, specific structural characteristics, and possibility to attach different molecules on their surface transform them into excellent transport vehicles able to cross cell and/or tissue barriers, including the blood-brain barrier. The latter is one of the greatest challenges in diagnosis and treatment of brain cancers. Application of nanomaterials can prolong the circulation time of the drugs and contrasting agents in the brain, posing an excellent opportunity for advancing the treatment of the most aggressive form of the brain cancer-glioblastomas. However, possible unwanted side-effects and toxicity issues must be considered before final clinical translation of nanoparticles.
Collapse
Affiliation(s)
- Alja Zottel
- Medical Center for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
| | - Alja Videtič Paska
- Medical Center for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
| | - Ivana Jovčevska
- Medical Center for Molecular Biology, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia.
| |
Collapse
|
15
|
Comparison between Polybutylcyanoacrylate Nanoparticles with Either Surface-Adsorbed or Encapsulated Brain-Derived Neurotrophic Factor on the Neural Differentiation of iPSCs. Int J Mol Sci 2019; 20:ijms20010182. [PMID: 30621332 PMCID: PMC6337453 DOI: 10.3390/ijms20010182] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/22/2018] [Accepted: 12/31/2018] [Indexed: 01/18/2023] Open
Abstract
The brain-derived neurotrophic factor (BDNF) is vital in the neural differentiation of neural stem/progenitor cells, and together may have therapeutic potential for neural regeneration. In this study, a multiplexed polybutylcyanoacrylate nanoparticle (PBCA NP) delivery platform was constructed, incorporating either surface-adsorbed or encapsulated BDNF for the induction of neural differentiation in induced pleuripotent stem cells (iPSCs), where tween 80 (T80) and superparamagnetic iron oxide (SPIO) were added for central nervous system (CNS) targeting and magnetic resonance (MR) image tracking, respectively. Both methods by which the BDNF was carried resulted in loading efficiencies greater than 95%. The nanoparticle-mediated delivery of BDNF resulted in neural differentiation of iPSCs detected on immunofluorescence staining as early as 7 days, with enhanced differentiation efficiency by 1.3-fold compared to the control on flow cytometry; the delivery system of surface-adsorbed BDNF gave rise to cells that had the best neural development than the encapsulated formulation. T80-coating disrupted the in vitro blood–brain barrier model with a corresponding 1.5- to two-fold increase in permeability. SPIO-loaded PBCA NPs exhibited a concentration-dependent, rapid decay in signal intensity on the phantom MR experiment. This study demonstrates the versatility of the PBCA NP, and the surface-adsorption of BDNF is the preferred method of delivery for the differentiation of iPSCs.
Collapse
|
16
|
Galla HJ. Monocultures of primary porcine brain capillary endothelial cells: Still a functional in vitro model for the blood-brain-barrier. J Control Release 2018; 285:172-177. [PMID: 30005905 DOI: 10.1016/j.jconrel.2018.07.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/26/2018] [Accepted: 07/09/2018] [Indexed: 12/21/2022]
Abstract
The main obstacle for the treatment of brain diseases is the restriction of the passage of pharmaceuticals across the blood-brain barrier. Endothelial cells line up the cerebral micro vessels and prevent the uncontrolled transfer of polar substances by intercellular tight junctions. In addition to this physical barrier, active transporters of the multi-drug-resistance prevent the passage of hydrophobic substances by refluxing them back to the blood stream. This paper reviews the development and selected applications of an in vitro porcine brain derived primary cell culture system established in the authors lab that closely resembles the BBB in vivo and could thus be used to study beyond other applications drug delivery to the brain. An essential technique to control the intactness or destruction of the barrier, the impedance spectroscopy, will be introduced. It will be shown that nanoparticles can cross the blood brain barrier by two mechanisms: opening the tight junctions and thus allowing parallel import of substances into the brain as well as receptor mediated endocytosis using brain specific target molecules. However cytotoxic effects have to be considered as well which beside standard cytotoxicity assays could be also determined by impedance technology. Moreover it will be shown that enzymes e.g. for enzyme replacement therapy could be transferred across the barrier by proper tuning or chemical modification of the enzyme. Since this review is based on a conference presentation it will mainly focus on applications of the monoculture system developed in the authors lab which under given culture conditions is useful due to its easy availability, robustness, good reproducibility and also due to its simplicity. Improvements of this model made by other groups will be acknowledged but not discussed here in detail.
Collapse
Affiliation(s)
- Hans-Joachim Galla
- Institute for Biochemistry, Westfälische Wilhems Universität,Münster, Wilhelm Klemm Str. 2, 48149 Münster, Germany.
| |
Collapse
|
17
|
Lohren H, Bornhorst J, Fitkau R, Pohl G, Galla HJ, Schwerdtle T. Effects on and transfer across the blood-brain barrier in vitro-Comparison of organic and inorganic mercury species. BMC Pharmacol Toxicol 2016; 17:63. [PMID: 27978854 PMCID: PMC5159962 DOI: 10.1186/s40360-016-0106-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 11/19/2016] [Indexed: 01/22/2023] Open
Abstract
Background Transport of methylmercury (MeHg) across the blood-brain barrier towards the brain side is well discussed in literature, while ethylmercury (EtHg) and inorganic mercury are not adequately characterized regarding their entry into the brain. Studies investigating a possible efflux out of the brain are not described to our knowledge. Methods This study compares, for the first time, effects of organic methylmercury chloride (MeHgCl), EtHg-containing thiomersal and inorganic Hg chloride (HgCl2) on as well as their transfer across a primary porcine in vitro model of the blood-brain barrier. Results With respect to the barrier integrity, the barrier model exhibited a much higher sensitivity towards HgCl2 following basolateral incubation (brain-facing side) as compared to apical application (blood-facing side). These HgCl2 induced effects on the barrier integrity after brain side incubation are comparable to that of the organic species, although MeHgCl and thiomersal exerted much higher cytotoxic effects in the barrier building cells. Hg transfer rates following exposure to organic species in both directions argue for diffusion as transfer mechanism. Inorganic Hg application surprisingly resulted in a Hg transfer out of the brain-facing compartment. Conclusions In case of MeHgCl and thiomersal incubation, mercury crossed the barrier in both directions, with a slight accumulation in the basolateral, brain-facing compartment, after simultaneous incubation in both compartments. For HgCl2, our data provide first evidence that the blood-brain barrier transfers mercury out of the brain.
Collapse
Affiliation(s)
- Hanna Lohren
- Department of Food Chemistry, Institute of Nutritional Science, Univeristy of Potsdam, Potsdam, Germany
| | - Julia Bornhorst
- Department of Food Chemistry, Institute of Nutritional Science, Univeristy of Potsdam, Potsdam, Germany
| | - Romy Fitkau
- Department of Food Chemistry, Institute of Nutritional Science, Univeristy of Potsdam, Potsdam, Germany
| | - Gabriele Pohl
- Department of Food Chemistry, Institute of Nutritional Science, Univeristy of Potsdam, Potsdam, Germany
| | | | - Tanja Schwerdtle
- Department of Food Chemistry, Institute of Nutritional Science, Univeristy of Potsdam, Potsdam, Germany.
| |
Collapse
|
18
|
He X, Zhu Y, Wang M, Jing G, Zhu R, Wang S. Antidepressant effects of curcumin and HU-211 coencapsulated solid lipid nanoparticles against corticosterone-induced cellular and animal models of major depression. Int J Nanomedicine 2016; 11:4975-4990. [PMID: 27757031 PMCID: PMC5055126 DOI: 10.2147/ijn.s109088] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Major depression is a complex neuropsychiatric disorder with few treatment approaches. The use of nontargeted antidepressants induced many side effects with their low efficacy. A more precise targeting strategy is to develop nanotechnology-based drug delivery systems; hence, we employed solid lipid nanoparticles (SLNs) to encapsulate HU-211 and curcumin (Cur). The antidepressant effects of the dual-drug nanoparticles (Cur/SLNs-HU-211) for major depression treatment were investigated in corticosterone-induced cellular and animal models of major depression. Cur/SLNs-HU-211 can effectively protect PC12 cells from corticosterone-induced apoptosis and can release more dopamine, which may be associated with the higher uptake of Cur/SLNs-HU-211 shown by cellular uptake behavior analysis. Additionally, Cur/SLNs-HU-211 significantly reduced the immobility time in forced swim test, enhanced fall latency in rotarod test, and improved the level of dopamine in mice blood. Cur/SLNs-HU-211 can deliver more Cur to the brain and thus produce a significant increase in neurotransmitters level in brain tissue, especially in the hippocampus and striatum. The results of Western blot and immunofluorescence revealed that Cur/SLNs-HU-211 can significantly enhance the expression of CB1, p-MEK1, and p-ERK1/2. Our study suggests that Cur/SLNs-HU-211 may have great potential for major depression treatment.
Collapse
Affiliation(s)
- Xiaolie He
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Yanjing Zhu
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Mei Wang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Guoxin Jing
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Rongrong Zhu
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| | - Shilong Wang
- Research Center for Translational Medicine at East Hospital, School of Life Science and Technology, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
19
|
MicroRNA-320a Strengthens Intestinal Barrier Function and Follows the Course of Experimental Colitis. Inflamm Bowel Dis 2016; 22:2341-55. [PMID: 27607334 DOI: 10.1097/mib.0000000000000917] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Inflammatory bowel disease is a chronic-remittent disorder with the risk of disabling complications due to uncontrolled inflammation. Accurate biomarkers are needed to noninvasively monitor the disease course to tailor therapy. We evaluated the potential of the specific microRNA (miR)-320a to monitor disease activity in experimental colitis or patients with Crohn's disease and investigated its functional role in intestinal epithelial barrier formation. METHODS The impact of miR-320a on intestinal barrier function was tested in vitro in T84 epithelial cells by transepithelial resistance measurement and quantitative real-time polymerase chain reaction analysis on inflammatory and microbial stimulation. Experimental colitis was studied in dextran sodium sulfate colitis, T-cell transfer colitis, and IL-10 mice. Disease course was monitored by body weight measurement, colonoscopy, and histological examination. MiR-320a expression during inflammation was assessed in T84 cells, murine blood, and colonic tissue and in peripheral blood from patients with Crohn's disease with active or quiescent disease. RESULTS MiR-320a transfection of T84 cells reinforced barrier integrity reflected by increased transepithelial resistance (P < 0.01) and inhibited barrier-destructive enteropathogenic Escherichia coli effects resulting in increased tight junction protein JAM-A expression (P = 0.02) and decrease of barrier integrity-destabilizing miR-320a target PPP2R5B (P < 0.001). Tumor necrosis factor-α and interleukin-1β stimulation increased a miR-320a epxression in T84 cells. MiR-320a level was increased in blood samples from colitic mice and patients with Crohn's disease showing a strong correlation with disease activity (r = 0.67). CONCLUSIONS MiR-320a strengthens intestinal barrier function in vitro and has the potential to monitor disease activity of colitic mice. Future studies are needed to further evaluate the potential of miR-320a in patients with inflammatory bowel disease.
Collapse
|
20
|
Neves AR, Queiroz JF, Weksler B, Romero IA, Couraud PO, Reis S. Solid lipid nanoparticles as a vehicle for brain-targeted drug delivery: two new strategies of functionalization with apolipoprotein E. NANOTECHNOLOGY 2015; 26:495103. [PMID: 26574295 DOI: 10.1088/0957-4484/26/49/495103] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Nanotechnology can be an important tool to improve the permeability of some drugs for the blood-brain barrier. In this work we created a new system to enter the brain by functionalizing solid lipid nanoparticles with apolipoprotein E, aiming to enhance their binding to low-density lipoprotein receptors on the blood-brain barrier endothelial cells. Solid lipid nanoparticles were successfully functionalized with apolipoprotein E using two distinct strategies that took advantage of the strong interaction between biotin and avidin. Transmission electron microscopy images revealed spherical nanoparticles, and dynamic light scattering gave a Z-average under 200 nm, a polydispersity index below 0.2, and a zeta potential between -10 mV and -15 mV. The functionalization of solid lipid nanoparticles with apolipoprotein E was demonstrated by infrared spectroscopy and fluorimetric assays. In vitro cytotoxic effects were evaluated by MTT and LDH assays in the human cerebral microvascular endothelial cells (hCMEC/D3) cell line, a human blood-brain barrier model, and revealed no toxicity up to 1.5 mg ml(-1) over 4 h of incubation. The brain permeability was evaluated in transwell devices with hCMEC/D3 monolayers, and a 1.5-fold increment in barrier transit was verified for functionalized nanoparticles when compared with non-functionalized ones. The results suggested that these novel apolipoprotein E-functionalized nanoparticles resulted in dynamic stable systems capable of being used for an improved and specialized brain delivery of drugs through the blood-brain barrier.
Collapse
Affiliation(s)
- Ana Rute Neves
- REQUIMTE, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | | | | | | | | | | |
Collapse
|
21
|
Anand P, O’Neil A, Lin E, Douglas T, Holford M. Tailored delivery of analgesic ziconotide across a blood brain barrier model using viral nanocontainers. Sci Rep 2015; 5:12497. [PMID: 26234920 PMCID: PMC4522602 DOI: 10.1038/srep12497] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/11/2015] [Indexed: 02/07/2023] Open
Abstract
The blood brain barrier (BBB) is often an insurmountable obstacle for a large number of candidate drugs, including peptides, antibiotics, and chemotherapeutic agents. Devising an adroit delivery method to cross the BBB is essential to unlocking widespread application of peptide therapeutics. Presented here is an engineered nanocontainer for delivering peptidic drugs across the BBB encapsulating the analgesic marine snail peptide ziconotide (Prialt®). We developed a bi-functional viral nanocontainer based on the Salmonella typhimurium bacteriophage P22 capsid, genetically incorporating ziconotide in the interior cavity, and chemically attaching cell penetrating HIV-Tat peptide on the exterior of the capsid. Virus like particles (VLPs) of P22 containing ziconotide were successfully transported in several BBB models of rat and human brain microvascular endothelial cells (BMVEC) using a recyclable noncytotoxic endocytic pathway. This work demonstrates proof in principle for developing a possible alternative to intrathecal injection of ziconotide using a tunable VLP drug delivery nanocontainer to cross the BBB.
Collapse
Affiliation(s)
- Prachi Anand
- Hunter College-CUNY, Belfer Research Building, 413 E, 69th Street, New York, NY-10021 (USA)
- The American Museum of Natural History, Central Park West 79th Street, New York, NY-10024 (USA)
| | - Alison O’Neil
- Indiana University, 800 E. Kirkwood Ave., Bloomington, IN-47405 (USA)
| | - Emily Lin
- Hunter College-CUNY, Belfer Research Building, 413 E, 69th Street, New York, NY-10021 (USA)
| | - Trevor Douglas
- Indiana University, 800 E. Kirkwood Ave., Bloomington, IN-47405 (USA)
| | - Mandë Holford
- Hunter College-CUNY, Belfer Research Building, 413 E, 69th Street, New York, NY-10021 (USA)
- The American Museum of Natural History, Central Park West 79th Street, New York, NY-10024 (USA)
| |
Collapse
|
22
|
Md S, Mustafa G, Baboota S, Ali J. Nanoneurotherapeutics approach intended for direct nose to brain delivery. Drug Dev Ind Pharm 2015; 41:1922-34. [PMID: 26057769 DOI: 10.3109/03639045.2015.1052081] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CONTEXT Brain disorders remain the world's leading cause of disability, and account for more hospitalizations and prolonged care than almost all other diseases combined. The majority of drugs, proteins and peptides do not readily permeate into brain due to the presence of the blood-brain barrier (BBB), thus impeding treatment of these conditions. OBJECTIVE Attention has turned to developing novel and effective delivery systems to provide good bioavailability in the brain. METHODS Intranasal administration is a non-invasive method of drug delivery that may bypass the BBB, allowing therapeutic substances direct access to the brain. However, intranasal administration produces quite low drug concentrations in the brain due limited nasal mucosal permeability and the harsh nasal cavity environment. Pre-clinical studies using encapsulation of drugs in nanoparticulate systems improved the nose to brain targeting and bioavailability in brain. However, the toxic effects of nanoparticles on brain function are unknown. RESULT AND CONCLUSION This review highlights the understanding of several brain diseases and the important pathophysiological mechanisms involved. The review discusses the role of nanotherapeutics in treating brain disorders via nose to brain delivery, the mechanisms of drug absorption across nasal mucosa to the brain, strategies to overcome the blood brain barrier, nanoformulation strategies for enhanced brain targeting via nasal route and neurotoxicity issues of nanoparticles.
Collapse
Affiliation(s)
- Shadab Md
- a Department of Pharmaceutical Technology , School of Pharmacy, International Medical University (IMU) , Kuala Lumpur , Malaysia
| | - Gulam Mustafa
- b College of Pharmacy, Al-Dawadmi Campus, Shaqra University , Riyadh , Kingdom of Saudi Arabia , and.,c Faculty of Pharmacy, Department of Pharmaceutics , Jamia Hamdard, New Delhi , India
| | - Sanjula Baboota
- c Faculty of Pharmacy, Department of Pharmaceutics , Jamia Hamdard, New Delhi , India
| | - Javed Ali
- c Faculty of Pharmacy, Department of Pharmaceutics , Jamia Hamdard, New Delhi , India
| |
Collapse
|
23
|
Nanoparticle-mediated growth factor delivery systems: A new way to treat Alzheimer's disease. J Control Release 2015; 206:187-205. [DOI: 10.1016/j.jconrel.2015.03.024] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/19/2015] [Accepted: 03/20/2015] [Indexed: 01/03/2023]
|
24
|
Gao S, Xu Y, Asghar S, Chen M, Zou L, Eltayeb S, Huo M, Ping Q, Xiao Y. Polybutylcyanoacrylate nanocarriers as promising targeted drug delivery systems. J Drug Target 2015; 23:481-96. [DOI: 10.3109/1061186x.2015.1020426] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
25
|
Stangenberg R, Wu Y, Hedrich J, Kurzbach D, Wehner D, Weidinger G, Kuan SL, Jansen MI, Jelezko F, Luhmann HJ, Hinderberger D, Weil T, Müllen K. A polyphenylene dendrimer drug transporter with precisely positioned amphiphilic surface patches. Adv Healthc Mater 2015; 4:377-84. [PMID: 25182694 DOI: 10.1002/adhm.201400291] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Revised: 07/19/2014] [Indexed: 01/14/2023]
Abstract
The design and synthesis of a polyphenylene dendrimer (PPD 3) with discrete binding sites for lipophilic guest molecules and characteristic surface patterns is presented. Its semi-rigidity in combination with a precise positioning of hydrophilic and hydrophobic groups at the periphery yields a refined architecture with lipophilic binding pockets that accommodate defined numbers of biologically relevant guest molecules such as fatty acids or the drug doxorubicin. The size, architecture, and surface textures allow to even penetrate brain endothelial cells that are a major component of the extremely tight blood-brain barrier. In addition, low to no toxicity is observed in in vivo studies using zebrafish embryos. The unique PPD scaffold allows the precise placement of functional groups in a given environment and offers a universal platform for designing drug transporters that closely mimic many features of proteins.
Collapse
Affiliation(s)
- René Stangenberg
- Max Planck Institute for Polymer Research; Ackermannweg 10 55128 Mainz Germany
| | - Yuzhou Wu
- Institute of Organic Chemistry III; Ulm University; Albert-Einstein-Allee 11 89081 Ulm Germany
| | - Jana Hedrich
- Institute for Physiology; Johannes Gutenberg University Mainz; Duesbergweg 6 55128 Mainz Germany
| | - Dennis Kurzbach
- Max Planck Institute for Polymer Research; Ackermannweg 10 55128 Mainz Germany
| | - Daniel Wehner
- Institute for Biochemistry and Molecular Biology; Ulm University; Albert-Einstein-Allee 11 89081 Ulm Germany
| | - Gilbert Weidinger
- Institute for Biochemistry and Molecular Biology; Ulm University; Albert-Einstein-Allee 11 89081 Ulm Germany
| | - Seah Ling Kuan
- Institute of Organic Chemistry III; Ulm University; Albert-Einstein-Allee 11 89081 Ulm Germany
| | - Malin Insa Jansen
- Institute for Physiology; Johannes Gutenberg University Mainz; Duesbergweg 6 55128 Mainz Germany
| | - Fedor Jelezko
- Institute for Quantum Optics; Ulm University; Albert-Einstein-Allee 11 89081 Ulm Germany
| | - Heiko J. Luhmann
- Institute for Physiology; Johannes Gutenberg University Mainz; Duesbergweg 6 55128 Mainz Germany
| | - Dariush Hinderberger
- Max Planck Institute for Polymer Research; Ackermannweg 10 55128 Mainz Germany
- Institute of Chemistry; Martin-Luther-Universität; Halle-Wittenberg Von-Danckelmann-Platz 4 06120 Halle (Saale) Germany
| | - Tanja Weil
- Institute of Organic Chemistry III; Ulm University; Albert-Einstein-Allee 11 89081 Ulm Germany
| | - Klaus Müllen
- Max Planck Institute for Polymer Research; Ackermannweg 10 55128 Mainz Germany
| |
Collapse
|
26
|
Kolter M, Ott M, Hauer C, Reimold I, Fricker G. Nanotoxicity of poly(n-butylcyano-acrylate) nanoparticles at the blood-brain barrier, in human whole blood and in vivo. J Control Release 2014; 197:165-79. [PMID: 25445700 DOI: 10.1016/j.jconrel.2014.11.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 10/30/2014] [Accepted: 11/03/2014] [Indexed: 12/24/2022]
Abstract
Therapy of diseases of the central nervous system is a major challenge since drugs have to overcome the blood-brain barrier (BBB). A powerful strategy to enhance cerebral drug concentration is administration of drug-loaded poly(n-butylcyano-acrylate) (PBCA) nanoparticles coated with polysorbate 80 (PS80). This study evaluates the toxicity of PBCA-nanoparticles at the BBB, representing the target organ, the inflammatory response in human whole blood, as the site of administration and in a rat model in vivo. PBCA-nanoparticles were prepared by a mini-emulsion method and characterized concerning size, surface charge, shape and PS80-adsorption. The influence on metabolic activity, cell viability and integrity of the BBB was analyzed in an in vitro model of the BBB. In ex vivo experiments in human whole blood the release of 12 inflammatory cytokines was investigated. In addition, the inflammatory response was studied in vivo in rats and complemented with the analysis of different organ toxicity parameters. PBCA-nanoparticles showed time- and concentration-dependent effects on metabolic activity, cell viability and BBB integrity. No cell death or loss of metabolic activity was observed for nanoparticle-concentrations ≤500μg/ml up to 3h of treatment. Within 12 tested inflammatory cytokines, only interleukin-8 displayed a significant release after nanoparticle exposure in human blood. No severe inflammatory processes or organ damages were identified in rats in vivo. Thus, PBCA-nanoparticles are a promising drug delivery system to overcome the BBB since they showed hardly any cytotoxic or inflammatory effect at therapeutic concentrations and incubation times.
Collapse
Affiliation(s)
- Marise Kolter
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany.
| | - Melanie Ott
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany
| | - Christian Hauer
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany
| | - Isolde Reimold
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
27
|
Tajes M, Ramos-Fernández E, Weng-Jiang X, Bosch-Morató M, Guivernau B, Eraso-Pichot A, Salvador B, Fernàndez-Busquets X, Roquer J, Muñoz FJ. The blood-brain barrier: structure, function and therapeutic approaches to cross it. Mol Membr Biol 2014; 31:152-67. [PMID: 25046533 DOI: 10.3109/09687688.2014.937468] [Citation(s) in RCA: 179] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The blood-brain barrier (BBB) is constituted by a specialized vascular endothelium that interacts directly with astrocytes, neurons and pericytes. It protects the brain from the molecules of the systemic circulation but it has to be overcome for the proper treatment of brain cancer, psychiatric disorders or neurodegenerative diseases, which are dramatically increasing as the population ages. In the present work we have revised the current knowledge on the cellular structure of the BBB and the different procedures utilized currently and those proposed to cross it. Chemical modifications of the drugs, such as increasing their lipophilicity, turn them more prone to be internalized in the brain. Other mechanisms are the use of molecular tools to bind the drugs such as small immunoglobulins, liposomes or nanoparticles that will act as Trojan Horses favoring the drug delivery in brain. This fusion of the classical pharmacology with nanotechnology has opened a wide field to many different approaches with promising results to hypothesize that BBB will not be a major problem for the new generation of neuroactive drugs. The present review provides an overview of all state-of-the-art of the BBB structure and function, as well as of the classic strategies and these appeared in recent years to deliver drugs into the brain for the treatment of Central Nervous System (CNS) diseases.
Collapse
Affiliation(s)
- Marta Tajes
- Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF) , Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Cupaioli FA, Zucca FA, Boraschi D, Zecca L. Engineered nanoparticles. How brain friendly is this new guest? Prog Neurobiol 2014; 119-120:20-38. [PMID: 24820405 DOI: 10.1016/j.pneurobio.2014.05.002] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 04/24/2014] [Accepted: 05/02/2014] [Indexed: 12/19/2022]
Abstract
In the last 30 years, the use of engineered nanoparticles (NPs) has progressively increased in many industrial and medical applications. In therapy, NPs may allow more effective cellular and subcellular targeting of drugs. In diagnostic applications, quantum dots are exploited for their optical characteristics, while superparamagnetic iron oxides NPs are used in magnetic resonance imaging. NPs are used in semiconductors, packaging, textiles, solar cells, batteries and plastic materials. Despite the great progress in nanotechnologies, comparatively little is known to date on the effects that exposure to NPs may have on the human body, in general and specifically on the brain. NPs can enter the human body through skin, digestive tract, airways and blood and they may cross the blood-brain barrier to reach the central nervous system. In addition to the paucity of studies describing NP effects on brain function, some of them also suffer of insufficient NPs characterization, inadequate standardization of conditions and lack of contaminant evaluation, so that results from different studies can hardly be compared. It has been shown in vitro and in vivo in rodents that NPs can impair dopaminergic and serotoninergic systems. Changes of neuronal morphology and neuronal death were reported in mice treated with NPs. NPs can also affect the respiratory chain of mitochondria and Bax protein levels, thereby causing apoptosis. Changes in expression of genes involved in redox pathways in mouse brain regions were described. NPs can induce autophagy, and accumulate in lysosomes impairing their degradation capacity. Cytoskeleton and vesicle trafficking may also be affected. NPs treated animals showed neuroinflammation with microglia activation, which could induce neurodegeneration. Considering the available data, it is important to design adequate models and experimental systems to evaluate in a reliable and controlled fashion the effects of NPs on the brain, and generate data representative of effects on the human brain, thereby useful for developing robust and valid nanosafety standards.
Collapse
Affiliation(s)
- Francesca A Cupaioli
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Fabio A Zucca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Diana Boraschi
- Institute of Biomedical Technologies, National Research Council of Italy, Unit of Pisa, Pisa, Italy
| | - Luigi Zecca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy.
| |
Collapse
|
29
|
Voigt N, Henrich-Noack P, Kockentiedt S, Hintz W, Tomas J, Sabel BA. Surfactants, not size or zeta-potential influence blood–brain barrier passage of polymeric nanoparticles. Eur J Pharm Biopharm 2014; 87:19-29. [DOI: 10.1016/j.ejpb.2014.02.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 01/16/2014] [Accepted: 02/24/2014] [Indexed: 10/25/2022]
|
30
|
Purushothaman M, Gopala Krishnan PS, Nayak SK. Effect of Isoalkyl Lactates as Pendant Group on Poly(acrylic acid). JOURNAL OF MACROMOLECULAR SCIENCE PART A-PURE AND APPLIED CHEMISTRY 2014. [DOI: 10.1080/10601325.2014.906251] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
31
|
Bicker J, Alves G, Fortuna A, Falcão A. Blood-brain barrier models and their relevance for a successful development of CNS drug delivery systems: a review. Eur J Pharm Biopharm 2014; 87:409-32. [PMID: 24686194 DOI: 10.1016/j.ejpb.2014.03.012] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2013] [Revised: 03/13/2014] [Accepted: 03/20/2014] [Indexed: 02/05/2023]
Abstract
During the research and development of new drugs directed at the central nervous system, there is a considerable attrition rate caused by their hampered access to the brain by the blood-brain barrier. Throughout the years, several in vitro models have been developed in an attempt to mimic critical functionalities of the blood-brain barrier and reliably predict the permeability of drug candidates. However, the current challenge lies in developing a model that retains fundamental blood-brain barrier characteristics and simultaneously remains compatible with the high throughput demands of pharmaceutical industries. This review firstly describes the roles of all elements of the neurovascular unit and their influence on drug brain penetration. In vitro models, including non-cell based and cell-based models, and in vivo models are herein presented, with a particular emphasis on their methodological aspects. Lastly, their contribution to the improvement of brain drug delivery strategies and drug transport across the blood-brain barrier is also discussed.
Collapse
Affiliation(s)
- Joana Bicker
- Laboratory of Pharmacology, University of Coimbra, Coimbra, Portugal; CNC - Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Gilberto Alves
- CNC - Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.
| | - Ana Fortuna
- Laboratory of Pharmacology, University of Coimbra, Coimbra, Portugal; CNC - Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Amílcar Falcão
- Laboratory of Pharmacology, University of Coimbra, Coimbra, Portugal; CNC - Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
32
|
Geldenhuys W, Wehrung D, Groshev A, Hirani A, Sutariya V. Brain-targeted delivery of doxorubicin using glutathione-coated nanoparticles for brain cancers. Pharm Dev Technol 2014; 20:497-506. [DOI: 10.3109/10837450.2014.892130] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
33
|
Strategies to overcome the barrier: use of nanoparticles as carriers and modulators of barrier properties. Cell Tissue Res 2014; 355:717-26. [DOI: 10.1007/s00441-014-1819-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 01/16/2014] [Indexed: 12/14/2022]
|
34
|
Ding H, Sagar V, Agudelo M, Pilakka-Kanthikeel S, Atluri VSR, Raymond A, Thangavel S, Nair MP. Enhanced blood-brain barrier transmigration using a novel transferrin embedded fluorescent magneto-liposome nanoformulation. NANOTECHNOLOGY 2014; 25:055101. [PMID: 24406534 PMCID: PMC4520325 DOI: 10.1088/0957-4484/25/5/055101] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The blood-brain barrier (BBB) is considered as the primary impediment barrier for most drugs. Delivering therapeutic agents to the brain is still a big challenge to date. In our study, a dual mechanism, receptor mediation combined with external non-invasive magnetic force, was incorporated into ferrous magnet-based liposomes for BBB transmigration enhancement. The homogenous magnetic nanoparticles (MNPs), with a size of ∼10 nm, were synthesized and confirmed by TEM and XRD respectively. The classical magnetism assay showed the presence of the characteristic superparamagnetic property. These MNPs encapsulated in PEGylated fluorescent liposomes as magneto-liposomes (MLs) showed mono-dispersion, ∼130 ± 10 nm diameter, by dynamic laser scattering (DLS) using the lipid-extrusion technique. Remarkably, a magnetite encapsulation efficiency of nearly 60% was achieved. Moreover, the luminescence and hydrodynamic size of the MLs was stable for over two months at 4 ° C. Additionally, the integrity of the ML structure remained unaffected through 120 rounds of circulation mimicking human blood fluid. After biocompatibility confirmation by cytotoxicity evaluation, these fluorescent MLs were further embedded with transferrin and applied to an in vitro BBB transmigration study in the presence or absence of external magnetic force. Comparing with magnetic force- or transferrin receptor-mediated transportation alone, their synergy resulted in 50-100% increased transmigration without affecting the BBB integrity. Consequently, confocal microscopy and iron concentration in BBB-composed cells further confirmed the higher cellular uptake of ML particles due to the synergic effect. Thus, our multifunctional liposomal magnetic nanocarriers possess great potential in particle transmigration across the BBB and may have a bright future in drug delivery to the brain.
Collapse
Affiliation(s)
- Hong Ding
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
- Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| | - Vidya Sagar
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
- Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| | - Marisela Agudelo
- Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| | - Sudheesh Pilakka-Kanthikeel
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
- Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| | - Venkata Subba Rao Atluri
- Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| | - Andrea Raymond
- Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| | - Samikkannu Thangavel
- Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
| | - Madhavan P. Nair
- Center for Personalized Nanomedicine, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
- Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, Florida 33199, USA
- Corresponding Authors: Madhavan P. Nair. Tel: 305-348-1493, Fax: 305-348-1109.
| |
Collapse
|
35
|
Chaturvedi M, Molino Y, Sreedhar B, Khrestchatisky M, Kaczmarek L. Tissue inhibitor of matrix metalloproteinases-1 loaded poly(lactic-co-glycolic acid) nanoparticles for delivery across the blood-brain barrier. Int J Nanomedicine 2014; 9:575-88. [PMID: 24531257 PMCID: PMC3901738 DOI: 10.2147/ijn.s54750] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Aim The aim of this study was to develop poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) for delivery of a protein – tissue inhibitor of matrix metalloproteinases 1 (TIMP-1) – across the blood–brain barrier (BBB) to inhibit deleterious matrix metalloproteinases (MMPs). Materials and methods The NPs were formulated by multiple-emulsion solvent-evaporation, and for enhancing BBB penetration, they were coated with polysorbate 80 (Ps80). We compared Ps80-coated and uncoated NPs for their toxicity, binding, and BBB penetration on primary rat brain capillary endothelial cell cultures and the rat brain endothelial 4 cell line. These studies were followed by in vivo studies for brain delivery of these NPs. Results Results showed that neither Ps80-coated nor uncoated NPs caused significant opening of the BBB, and essentially they were nontoxic. NPs without Ps80 coating had more binding to endothelial cells compared to Ps80-coated NPs. Penetration studies showed that TIMP-1 NPs + Ps80 had 11.21%±1.35% penetration, whereas TIMP-1 alone and TIMP-1 NPs without Ps80 coating did not cross the endothelial monolayer. In vivo studies indicated BBB penetration of intravenously injected TIMP-1 NPs + Ps80. Conclusion The study demonstrated that Ps80 coating of NPs does not cause significant toxic effects to endothelial cells and that it can be used to enhance the delivery of protein across endothelial cell barriers, both in vitro and in vivo.
Collapse
Affiliation(s)
| | | | - Bojja Sreedhar
- Indian Institute of Chemical Technology, Hyderabad, India
| | | | | |
Collapse
|
36
|
Gaillard PJ, Visser CC, de Boer M, Appeldoorn CCM, Rip J. Blood-to-Brain Drug Delivery Using Nanocarriers. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/978-1-4614-9105-7_15] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
37
|
Paolinelli R, Corada M, Ferrarini L, Devraj K, Artus C, Czupalla CJ, Rudini N, Maddaluno L, Papa E, Engelhardt B, Couraud PO, Liebner S, Dejana E. Wnt activation of immortalized brain endothelial cells as a tool for generating a standardized model of the blood brain barrier in vitro. PLoS One 2013; 8:e70233. [PMID: 23940549 PMCID: PMC3734070 DOI: 10.1371/journal.pone.0070233] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 06/18/2013] [Indexed: 02/02/2023] Open
Abstract
Reproducing the characteristics and the functional responses of the blood–brain barrier (BBB) in vitro represents an important task for the research community, and would be a critical biotechnological breakthrough. Pharmaceutical and biotechnology industries provide strong demand for inexpensive and easy-to-handle in vitro BBB models to screen novel drug candidates. Recently, it was shown that canonical Wnt signaling is responsible for the induction of the BBB properties in the neonatal brain microvasculature in vivo. In the present study, following on from earlier observations, we have developed a novel model of the BBB in vitro that may be suitable for large scale screening assays. This model is based on immortalized endothelial cell lines derived from murine and human brain, with no need for co-culture with astrocytes. To maintain the BBB endothelial cell properties, the cell lines are cultured in the presence of Wnt3a or drugs that stabilize β-catenin, or they are infected with a transcriptionally active form of β-catenin. Upon these treatments, the cell lines maintain expression of BBB-specific markers, which results in elevated transendothelial electrical resistance and reduced cell permeability. Importantly, these properties are retained for several passages in culture, and they can be reproduced and maintained in different laboratories over time. We conclude that the brain-derived endothelial cell lines that we have investigated gain their specialized characteristics upon activation of the canonical Wnt pathway. This model may be thus suitable to test the BBB permeability to chemicals or large molecular weight proteins, transmigration of inflammatory cells, treatments with cytokines, and genetic manipulation.
Collapse
Affiliation(s)
| | - Monica Corada
- IFOM-FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Luca Ferrarini
- IFOM-FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Kavi Devraj
- Institute of Neurology (Edinger Institute), Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Cédric Artus
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Cathrin J. Czupalla
- Institute of Neurology (Edinger Institute), Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Noemi Rudini
- IFOM-FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Luigi Maddaluno
- IFOM-FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Eleanna Papa
- IFOM-FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | | | - Pierre Olivier Couraud
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Stefan Liebner
- Institute of Neurology (Edinger Institute), Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Elisabetta Dejana
- IFOM-FIRC Institute of Molecular Oncology Foundation, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
- * E-mail:
| |
Collapse
|
38
|
Inamura A, Adachi Y, Inoue T, He Y, Tokuda N, Nawata T, Shirao S, Nomura S, Fujii M, Ikeda E, Owada Y, Suzuki M. Cooling treatment transiently increases the permeability of brain capillary endothelial cells through translocation of claudin-5. Neurochem Res 2013; 38:1641-7. [PMID: 23653089 DOI: 10.1007/s11064-013-1066-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 04/25/2013] [Accepted: 04/30/2013] [Indexed: 01/24/2023]
Abstract
The blood-brain-barrier (BBB) is formed by different cell types, of which brain microvascular endothelial cells are major structural constituents. The goal of this study was to examine the effects of cooling on the permeability of the BBB with reference to tight junction formation of brain microendothelial cells. The sensorimotor cortex above the dura mater in adult male Wistar rats was focally cooled to a temperature of 5 °C for 1 h, then immunostaining for immunoglobulin G (IgG) was performed to evaluate the permeability of the BBB. Permeability produced by cooling was also evaluated in cultured murine brain endothelial cells (bEnd3) based on measurement of trans-epithelial electric resistance (TEER). Immunocytochemistry and Western blotting of proteins associated with tight junctions in bEnd3 were performed to determine protein distribution before and after cooling. After focal cooling of the rat brain cortex, diffuse immunostaining for IgG was observed primarily around the small vasculature and in the extracellular spaces of parenchyma of the cortex. In cultured bEnd3, TEER significantly decreased during cooling (15 °C) and recovered to normal levels after rewarming to 37 °C. Immunocytochemistry and Western blotting showed that claudin-5, a critical regulatory protein for tight junctions, was translocated from the membrane to the cytoplasm after cooling in cultured bEnd3 cells. These results suggest that focal brain cooling may open the BBB transiently through an effect on tight junctions of brain microendothelial cells, and that therapeutically this approach may allow control of BBB function and drug delivery through the BBB.
Collapse
Affiliation(s)
- Akinori Inamura
- Department of Neurosurgery, Graduate School of Medicine, Yamaguchi University, Yamaguchi, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Influence of T-2 and HT-2 toxin on the blood-brain barrier in vitro: new experimental hints for neurotoxic effects. PLoS One 2013; 8:e60484. [PMID: 23544145 PMCID: PMC3609806 DOI: 10.1371/journal.pone.0060484] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 02/27/2013] [Indexed: 12/23/2022] Open
Abstract
The trichothecene mycotoxin T-2 toxin is a common contaminant of food and feed and is also present in processed cereal derived products. Cytotoxic effects of T-2 toxin and its main metabolite HT-2 toxin are already well described with apoptosis being a major mechanism of action. However, effects on the central nervous system were until now only reported rarely. In this study we investigated the effects of T-2 and HT-2 toxin on the blood-brain barrier (BBB) in vitro. Besides strong cytotoxic effects on the BBB as determined by the CCK-8 assay, impairment of the barrier function starting at low nanomolar concentrations were observed for T-2 toxin. HT-2 toxin, however, caused barrier disruption at higher concentrations compared to T-2 toxin. Further, the influence on the tight junction protein occludin was studied and permeability of both toxins across the BBB was detected when applied from the apical (blood) or the basolateral (brain) side respectively. These results clearly indicate the ability of both toxins to enter the brain via the BBB.
Collapse
|
40
|
Nagpal K, Singh SK, Mishra DN. Drug targeting to brain: a systematic approach to study the factors, parameters and approaches for prediction of permeability of drugs across BBB. Expert Opin Drug Deliv 2013; 10:927-55. [DOI: 10.1517/17425247.2013.762354] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
41
|
Benson K, Cramer S, Galla HJ. Impedance-based cell monitoring: barrier properties and beyond. Fluids Barriers CNS 2013; 10:5. [PMID: 23305242 PMCID: PMC3560213 DOI: 10.1186/2045-8118-10-5] [Citation(s) in RCA: 255] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 12/19/2012] [Indexed: 12/02/2022] Open
Abstract
In multicellular organisms epithelial and endothelial cells form selective permeable interfaces between tissue compartments of different chemical compositions. Tight junctions which connect adjacent cells, control the passage of molecules across the barrier and, in addition, facilitate active transport processes. The cellular barriers are not static but can be deliberately modulated by exposure to specific external stimuli. In vitro models representing the essential absorption barriers of the body are nowadays available, thus allowing investigation of the parameters that control permeability as well as transport processes across those barriers. Independent of the origin of the barrier forming cells, techniques are needed to quantify their barrier integrity. One simple assay is to measure the permeability for given hydrophilic substrates possessing different molecular weights like sucrose or dextrans. However, this technique is time-consuming and labor-intensive. Moreover, radioactive or fluorescently-labeled substrates are needed to allow easy analytical detection. Finally, if transport processes are investigated, the standard permeant may interfere with the transport process under investigation or might even alter the barrier integrity by itself. Thus, independent, non-invasive techniques are needed to quantify the barrier integrity continuously during the experiment. Such techniques are available and are mainly based on the measurement of the transendothelial or transepithelial electrical resistance (TEER) of barrier forming cells grown on porous membranes. Simple devices using two sets of electrodes (so-called Voltohmeters) are widely used. In addition, an easy-to-use physical technique called impedance spectroscopy allows the continuous analysis of both the TEER and the electrical capacitance giving additional information about the barrier properties of cells grown on permeable membranes. This technique is useful as a quality control for barrier forming cells. Another impedance-based approach requires cells to be grown directly on solid, micro-structured electrodes. Here, we will discuss the physical background of the different techniques; advantages, disadvantages, and applications will be scrutinized. The aim is to give the reader a comprehensive understanding concerning the range and limits of the application, mainly focusing on endothelial cells.
Collapse
Affiliation(s)
- Kathrin Benson
- Institut für Biochemie, Westfälische Wilhelms-Universität Münster, Wilhelm Klemm Straße 2, Münster, 48149, Germany
| | - Sandra Cramer
- Institut für Biochemie, Westfälische Wilhelms-Universität Münster, Wilhelm Klemm Straße 2, Münster, 48149, Germany
| | - Hans-Joachim Galla
- Institut für Biochemie, Westfälische Wilhelms-Universität Münster, Wilhelm Klemm Straße 2, Münster, 48149, Germany
| |
Collapse
|
42
|
Freese C, Unger RE, Deller RC, Gibson MI, Brochhausen C, Klok HA, Kirkpatrick CJ. Uptake of poly(2-hydroxypropylmethacrylamide)-coated gold nanoparticles in microvascular endothelial cells and transport across the blood–brain barrier. Biomater Sci 2013; 1:824-833. [DOI: 10.1039/c3bm60050e] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
43
|
Hemmelmann M, Metz VV, Koynov K, Blank K, Postina R, Zentel R. Amphiphilic HPMA–LMA copolymers increase the transport of Rhodamine 123 across a BBB model without harming its barrier integrity. J Control Release 2012; 163:170-7. [DOI: 10.1016/j.jconrel.2012.08.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 08/29/2012] [Accepted: 08/31/2012] [Indexed: 01/27/2023]
|
44
|
Orthmann A, Fichtner I, Zeisig R. Improving the transport of chemotherapeutic drugs across the blood-brain barrier. Expert Rev Clin Pharmacol 2012; 4:477-90. [PMID: 22114857 DOI: 10.1586/ecp.11.26] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The successful treatment of brain tumors or metastases in the brain is still hampered by the very efficient blood-brain barrier, which prevents the cerebral accumulation of a pharmacologically sufficient amount of a drug. Beside the possibility of disintegrating the functionality of this effective working barrier, a nanocarrier-mediated transport is presently an interesting and promising method to increase the drug concentration in the brain. Nanocarriers are small vesicles (<200 nm) and can be prepared by polymerization, resulting in nanoparticles, or by producing superficial lipid structures to incorporate the drug. In this context, liposomes are of importance owing to their ability to adapt their properties to the pharmacological requirements. In this article, we will give an overview of current possibilities of enhancing anticancer drug transport across the blood-brain barrier, based on its structure and functionality. Special consideration will be given to recent liposomal approaches that use active targeting for receptor-mediated transport across this physiological barrier.
Collapse
Affiliation(s)
- Andrea Orthmann
- Max Delbrück Center for Molecular Medicine, Experimental Pharmacology, Robert-Rössle-Str. 10, 13122 Berlin, German
| | | | | |
Collapse
|
45
|
Gonçalves AS, Macedo AS, Souto EB. Therapeutic nanosystems for oncology nanomedicine. Clin Transl Oncol 2012; 14:883-90. [DOI: 10.1007/s12094-012-0912-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 06/15/2012] [Indexed: 10/28/2022]
|
46
|
Liu Y, Lu W. Recent advances in brain tumor-targeted nano-drug delivery systems. Expert Opin Drug Deliv 2012; 9:671-86. [DOI: 10.1517/17425247.2012.682726] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
47
|
Biddlestone-Thorpe L, Marchi N, Guo K, Ghosh C, Janigro D, Valerie K, Yang H. Nanomaterial-mediated CNS delivery of diagnostic and therapeutic agents. Adv Drug Deliv Rev 2012; 64:605-13. [PMID: 22178615 DOI: 10.1016/j.addr.2011.11.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Revised: 11/21/2011] [Accepted: 11/22/2011] [Indexed: 12/15/2022]
Abstract
Research into the diagnosis and treatment of central nervous system (CNS) diseases has been enhanced by rapid advances in nanotechnology and an expansion in the library of nanostructured carriers. This review discusses the latest applications of nanomaterials in the CNS with an emphasis on brain tumors. Novel administration routes and transport mechanisms for nanomaterial-mediated CNS delivery of diagnostic and therapeutic agents to bypass or cross the blood brain barrier (BBB) are also discussed. These include temporary disruption of the BBB, use of impregnated polymers (polymer wafers), convection-enhanced delivery (CED), and intranasal delivery. Moreover, an in vitro BBB model capable of mimicking geometrical, cellular and rheological features of the human cerebrovasculature has been developed. This is a useful tool that can be used for screening CNS nanoparticles or therapeutics prior to in vivo and clinical investigation. A discussion of this novel model is included.
Collapse
|
48
|
Wong HL, Wu XY, Bendayan R. Nanotechnological advances for the delivery of CNS therapeutics. Adv Drug Deliv Rev 2012; 64:686-700. [PMID: 22100125 DOI: 10.1016/j.addr.2011.10.007] [Citation(s) in RCA: 299] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Accepted: 10/27/2011] [Indexed: 12/18/2022]
Abstract
Effective non-invasive treatment of neurological diseases is often limited by the poor access of therapeutic agents into the central nervous system (CNS). The majority of drugs and biotechnological agents do not readily permeate into brain parenchyma due to the presence of two anatomical and biochemical dynamic barriers: the blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB). Therefore, one of the most significant challenges facing CNS drug development is the availability of effective brain targeting technology. Recent advances in nanotechnology have provided promising solutions to this challenge. Several nanocarriers ranging from the more established systems, e.g. polymeric nanoparticles, solid lipid nanoparticles, liposomes, micelles to the newer systems, e.g. dendrimers, nanogels, nanoemulsions and nanosuspensions have been studied for the delivery of CNS therapeutics. Many of these nanomedicines can be effectively transported across various in vitro and in vivo BBB models by endocytosis and/or transcytosis, and demonstrated early preclinical success for the management of CNS conditions such as brain tumors, HIV encephalopathy, Alzheimer's disease and acute ischemic stroke. Future development of CNS nanomedicines need to focus on increasing their drug-trafficking performance and specificity for brain tissue using novel targeting moieties, improving their BBB permeability and reducing their neurotoxicity.
Collapse
|
49
|
Qiao R, Jia Q, Hüwel S, Xia R, Liu T, Gao F, Galla HJ, Gao M. Receptor-mediated delivery of magnetic nanoparticles across the blood-brain barrier. ACS NANO 2012; 6:3304-10. [PMID: 22443607 DOI: 10.1021/nn300240p] [Citation(s) in RCA: 220] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
A brain delivery probe was prepared by covalently conjugating lactoferrin (Lf) to a poly(ethylene glycol) (PEG)-coated Fe(3)O(4) nanoparticle in order to facilitate the transport of the nanoparticles across the blood-brain barrier (BBB) by receptor-mediated transcytosis via the Lf receptor present on cerebral endothelial cells. The efficacy of the Fe(3)O(4)-Lf conjugate to cross the BBB was evaluated in vitro using a cell culture model for the blood-brain barrier as well as in vivo in SD rats. For an in vitro experiment, a well-established porcine BBB model was used based on the primary culture of cerebral capillary endothelial cells grown on filter supports, thus allowing one to follow the transfer of nanoparticles from the apical (blood) to the basolateral (brain) side. For in vivo experiments, SD rats were used as animal model to detect the passage of the nanoparticles through the BBB by MRI techniques. The results of both in vitro and in vivo experiments revealed that the Fe(3)O(4)-Lf probe exhibited an enhanced ability to cross the BBB in comparison to the PEG-coated Fe(3)O(4) nanoparticles and further suggested that the Lf-receptor-mediated transcytosis was an effective measure for delivering the nanoparticles across the BBB.
Collapse
Affiliation(s)
- Ruirui Qiao
- Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Qiaojuan Jia
- Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Sabine Hüwel
- Institute for Biochemistry, Westfälische Wilhelms-Universität, Wilhelm Klemm Straße 2, 48149 Münster, Germany
| | - Rui Xia
- Molecular Imaging Laboratory, Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ting Liu
- Molecular Imaging Laboratory, Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fabao Gao
- Molecular Imaging Laboratory, Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hans-Joachim Galla
- Institute for Biochemistry, Westfälische Wilhelms-Universität, Wilhelm Klemm Straße 2, 48149 Münster, Germany
| | - Mingyuan Gao
- Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| |
Collapse
|
50
|
Costantino L, Boraschi D. Is there a clinical future for polymeric nanoparticles as brain-targeting drug delivery agents? Drug Discov Today 2012; 17:367-78. [DOI: 10.1016/j.drudis.2011.10.028] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2011] [Revised: 10/06/2011] [Accepted: 10/31/2011] [Indexed: 01/07/2023]
|