1
|
Razavi ZS, Razavi FS, Alizadeh SS. Inorganic nanoparticles and blood-brain barrier modulation: Advancing targeted neurological therapies. Eur J Med Chem 2025; 287:117357. [PMID: 39947054 DOI: 10.1016/j.ejmech.2025.117357] [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: 12/09/2024] [Revised: 01/19/2025] [Accepted: 01/31/2025] [Indexed: 02/24/2025]
Abstract
The blood-brain barrier (BBB) is a protective barrier that complicates the treatment of neurological disorders. Pharmaceutical compounds encounter significant challenges in crossing the central nervous system (CNS). Nanoparticles (NPs) are promising candidates for treating neurological conditions as they help facilitate drug delivery. This review explores the diverse characteristics and mechanisms of inorganic NPs (INPs), including metal-based, ferric-oxide, and carbon-based nanoparticles, which facilitate their passage through the BBB. Emphasis is placed on the physicochemical properties of NPs such as size, shape, surface charge, and surface modifications and their role in enhancing drug delivery efficacy, reducing immune clearance, and improving BBB permeability. Specific synthesis approaches are demonstrated, with an emphasis on the influence of each one on NP property, biological activity and the capability of an NP for its intended application. As for the advances in the field, the review emphasizes those characterized the NP formulation and surface chemistry that conquered the BBB and tested the need for its alteration. Current findings indicate that NP therapy can in the future enable effective targeting of specific brain disorders and eventually evolve this drug delivery system, which would allow for lower doses with less side effects.
Collapse
Affiliation(s)
- Zahra Sadat Razavi
- Physiology Research Center, Iran University Medical Sciences, Tehran, Iran; Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran.
| | - Fateme Sadat Razavi
- Department of Mechanical Engineering, K. N. Toosi University of Technology, Tehran, Iran
| | | |
Collapse
|
2
|
Nayak U, Halagali P, Panchal KN, Tippavajhala VK, Mudgal J, Radhakrishnan R, Manikkath J. Nanoparticles in CNS Therapeutics: Pioneering Drug Delivery Advancements. Curr Pharm Des 2025; 31:443-460. [PMID: 39318210 DOI: 10.2174/0113816128328722240828184410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/19/2024] [Accepted: 07/29/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION The incidence of Central Nervous System (CNS) disorders, including Parkinson's disease, Alzheimer's disease, stroke, and malignancies, has risen significantly in recent decades, contributing to millions of deaths annually. Efficacious treatment of these disorders requires medicines targeting the brain. The Blood-brain Barrier (BBB) poses a formidable challenge to effective drug delivery to the brain, hindering progress in CNS therapeutics. This review explores the latest developments in nanoparticulate carriers, highlighting their potential to overcome BBB limitations. OBJECTIVE This study aimed to evaluate and summarise the critical factors and pathways in the nanoparticle- based CNS targeted drug delivery. METHODS An extensive literature search was conducted, comprising the initial development of nanoparticle- based CNS-targeted drug delivery approaches to the latest advancements using various online search tools. RESULTS The properties of nanoparticles, such as type of nanoparticles, size, shape, surface charge, hydrophobicity, and surface functionalisation, along with properties of the BBB during normal and pathological conditions and their impact on the delivery of nanoparticles across the BBB, are identified and discussed here. CONCLUSION Important properties and pathways that determine the penetration of nanoparticles across the CNS are reviewed in this article, along with recent advances in the field.
Collapse
Affiliation(s)
- Usha Nayak
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Praveen Halagali
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Khushi N Panchal
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Vamshi Krishna Tippavajhala
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
- Academic Unit of Oral and Maxillofacial Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield S102TA, UK
| | - Jyothsna Manikkath
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| |
Collapse
|
3
|
He X, Peng Y, Huang S, Xiao Z, Li G, Zuo Z, Zhang L, Shuai X, Zheng H, Hu X. Blood Brain Barrier-Crossing Delivery of Felodipine Nanodrug Ameliorates Anxiety-Like Behavior and Cognitive Impairment in Alzheimer's Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401731. [PMID: 38981028 PMCID: PMC11425895 DOI: 10.1002/advs.202401731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/29/2024] [Indexed: 07/11/2024]
Abstract
Alzheimer's disease (AD) is the most common age-related neurodegenerative disorder leading to cognitive decline. Excessive cytosolic calcium (Ca2+) accumulation plays a critical role in the pathogenesis of AD since it activates the NOD-like receptor family, pyrin domain containing 3 (NLRP3), switches the endoplasmic reticulum (ER) unfolded protein response (UPR) toward proapoptotic signaling and promotes Aβ seeding. Herein, a liposomal nanodrug (felodipine@LND) is developed incorporating a calcium channel antagonist felodipine for Alzheimer's disease treatment through a low-intensity pulse ultrasound (LIPUS) irradiation-assisted blood brain barrier (BBB)-crossing drug delivery. The multifunctional felodipine@LND is effectively delivered to diseased brain through applying a LIPUS irradiation to the skull, which resulted in a series of positive effects against AD. Markedly, the nanodrug treatment switched the ER UPR toward antioxidant signaling, prevented the surface translocation of ER calreticulin (CALR) in microglia, and inhibited the NLRP3 activation and Aβ seeding. In addition, it promoted the degradation of damaged mitochondria via mitophagy, thereby inhibiting the neuronal apoptosis. Therefore, the anxiety-like behavior and cognitive impairment of 5xFAD mice with AD is significantly ameliorated, which manifested the potential of LIPUS - assisted BBB-crossing delivery of felodipine@LND to serve as a paradigm for AD therapy based on the well-recognized clinically available felodipine.
Collapse
Affiliation(s)
- Xiaofei He
- Department of Rehabilitation MedicineThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhouGuangdong510630China
| | - Yuan Peng
- Department of Rehabilitation MedicineGuangzhou First People's HospitalGuangzhou510180China
| | - Sicong Huang
- School of Materials Science and Engineering Sun Yat‐sen UniversityGuangzhou510275China
| | - Zecong Xiao
- Nanomedicine Research CenterThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Ge Li
- Guangdong Provincial Key Laboratory of Laboratory AnimalsGuangdong Laboratory Animals Monitoring Institute11 Fengxin RoadGuangzhouGuangdong510663China
| | - Zejie Zuo
- Department of Rehabilitation MedicineThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhouGuangdong510630China
| | - Liying Zhang
- Department of Rehabilitation MedicineThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhouGuangdong510630China
| | - Xintao Shuai
- Nanomedicine Research CenterThe Third Affiliated Hospital of Sun Yat‐sen UniversityGuangzhou510630China
| | - Haiqing Zheng
- Department of Rehabilitation MedicineThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhouGuangdong510630China
| | - Xiquan Hu
- Department of Rehabilitation MedicineThe Third Affiliated HospitalSun Yat‐sen University600 Tianhe RoadGuangzhouGuangdong510630China
| |
Collapse
|
4
|
Hu Y, Wang X, Niu Y, He K, Tang M. Application of quantum dots in brain diseases and their neurotoxic mechanism. NANOSCALE ADVANCES 2024; 6:3733-3746. [PMID: 39050959 PMCID: PMC11265591 DOI: 10.1039/d4na00028e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 06/01/2024] [Indexed: 07/27/2024]
Abstract
The early-stage diagnosis and therapy of brain diseases pose a persistent challenge in the field of biomedicine. Quantum dots (QDs), nano-luminescent materials known for their small size and fluorescence imaging capabilities, present promising capabilities for diagnosing, monitoring, and treating brain diseases. Although some investigations about QDs have been conducted in clinical trials, the concerns about the toxicity of QDs have continued. In addition, the lack of effective toxicity evaluation methods and systems and the difference between in vivo and in vitro toxicity evaluation hinder QDs application. The primary objective of this paper is to introduce the neurotoxic effects and mechanisms attributable to QDs. First, we elucidate the utilization of QDs in brain disorders. Second, we sketch out three pathways through which QDs traverse into brain tissue. Ultimately, expound upon the adverse consequences of QDs on the brain and the mechanism of neurotoxicity in depth. Finally, we provide a comprehensive summary and outlook on the potential development of quantum dots in neurotoxicity and the difficulties to be overcome.
Collapse
Affiliation(s)
- Yuanyuan Hu
- Key Laboratory of Environmental Medicine & Engineering, Ministry of Education, School of Public Health, Southeast University Nanjing Jiangsu 210009 China
| | - Xiaoli Wang
- Key Laboratory of Environmental Medicine & Engineering, Ministry of Education, School of Public Health, Southeast University Nanjing Jiangsu 210009 China
| | - Yiru Niu
- Key Laboratory of Environmental Medicine & Engineering, Ministry of Education, School of Public Health, Southeast University Nanjing Jiangsu 210009 China
| | - Keyu He
- Blood Transfusion Department, Clinical Laboratory, Zhongda Hospital, Southeast University Nanjing Jiangsu 210009 China
| | - Meng Tang
- Key Laboratory of Environmental Medicine & Engineering, Ministry of Education, School of Public Health, Southeast University Nanjing Jiangsu 210009 China
| |
Collapse
|
5
|
Wehn AC, Krestel E, Harapan BN, Klymchenko A, Plesnila N, Khalin I. To see or not to see: In vivo nanocarrier detection methods in the brain and their challenges. J Control Release 2024; 371:216-236. [PMID: 38810705 DOI: 10.1016/j.jconrel.2024.05.044] [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: 02/16/2024] [Revised: 05/18/2024] [Accepted: 05/23/2024] [Indexed: 05/31/2024]
Abstract
Nanoparticles have a great potential to significantly improve the delivery of therapeutics to the brain and may also be equipped with properties to investigate brain function. The brain, being a highly complex organ shielded by selective barriers, requires its own specialized detection system. However, a significant hurdle to achieve these goals is still the identification of individual nanoparticles within the brain with sufficient cellular, subcellular, and temporal resolution. This review aims to provide a comprehensive summary of the current knowledge on detection systems for tracking nanoparticles across the blood-brain barrier and within the brain. We discuss commonly employed in vivo and ex vivo nanoparticle identification and quantification methods, as well as various imaging modalities able to detect nanoparticles in the brain. Advantages and weaknesses of these modalities as well as the biological factors that must be considered when interpreting results obtained through nanotechnologies are summarized. Finally, we critically evaluate the prevailing limitations of existing technologies and explore potential solutions.
Collapse
Affiliation(s)
- Antonia Clarissa Wehn
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Department of Neurosurgery, University of Munich Medical Center, Marchioninistraße 17, 81377 Munich, Germany.
| | - Eva Krestel
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany.
| | - Biyan Nathanael Harapan
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Department of Neurosurgery, University of Munich Medical Center, Marchioninistraße 17, 81377 Munich, Germany.
| | - Andrey Klymchenko
- Laboratoire de Biophotonique et Pharmacologie, CNRS UMR 7213, Université de Strasbourg, 74 route du Rhin - CS 60024, 67401 Illkirch Cedex, France.
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Munich Cluster of Systems Neurology (SyNergy), Feodor-Lynen-Straße 17, 81377 Munich, Germany.
| | - Igor Khalin
- Institute for Stroke and Dementia Research (ISD), Munich University Hospital, Feodor-Lynen-Straße 17, 81377, Germany; Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institute Blood and Brain @ Caen-Normandie (BB@C), 14 074 Bd Henri Becquerel, 14000 Caen, France.
| |
Collapse
|
6
|
Guan S, Tang M. Exposure of quantum dots in the nervous system: Central nervous system risks and the blood-brain barrier interface. J Appl Toxicol 2024; 44:936-952. [PMID: 38062852 DOI: 10.1002/jat.4568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 07/21/2024]
Abstract
Quantum dots currently possess significant importance in the field of biomedical science. Upon introduction into the body, quantum dots exhibit a tendency to accumulate in diverse tissues including the central nervous system (CNS). Consequently, it becomes imperative to devote specific attention to their potential toxic effects. Moreover, the preservation of optimal CNS function relies heavily on blood-brain barrier (BBB) integrity, thereby necessitating its prioritization in neurotoxicological investigations. A more comprehensive understanding of the BBB and CNS characteristics, along with the underlying mechanisms that may contribute to neurotoxicity, will greatly aid researchers in the development of effective design strategies. This article offers an in-depth look at the methods used to reduce the harmful effects of quantum dots on the nervous system, alongside the progression of effective treatments for brain-related conditions. The focal point of this discussion is the BBB and its intricate association with the CNS and neurotoxicology. The discourse commences by recent advancements in the medical application of quantum dots are examined. Subsequently, elucidating the mechanisms through which quantum dots infiltrate the human body and traverse into the brain. Additionally, the discourse delves into the factors that facilitate the passage of quantum dots across the BBB, primarily encompassing the physicochemical properties of quantum dots and the BBB's inherent capacity for self-permeability alteration. Furthermore, a concluding summary is presented, emphasizing existing research deficiencies and identifying promising avenues for further investigation within this field.
Collapse
Affiliation(s)
- Shujing Guan
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Meng Tang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| |
Collapse
|
7
|
Khan I, Gril B, Paranjape AN, Robinson CM, Difilippantonio S, Biernat W, Bieńkowski M, Pęksa R, Duchnowska R, Jassem J, Brastianos PK, Metellus P, Bialecki E, Woodroofe CC, Wu H, Swenson RE, Steeg PS. Comparison of Three Transcytotic Pathways for Distribution to Brain Metastases of Breast Cancer. Mol Cancer Ther 2023; 22:646-658. [PMID: 36912773 PMCID: PMC10164055 DOI: 10.1158/1535-7163.mct-22-0815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023]
Abstract
Advances in drug treatments for brain metastases of breast cancer have improved progression-free survival but new, more efficacious strategies are needed. Most chemotherapeutic drugs infiltrate brain metastases by moving between brain capillary endothelial cells, paracellular distribution, resulting in heterogeneous distribution, lower than that of systemic metastases. Herein, we tested three well-known transcytotic pathways through brain capillary endothelial cells as potential avenues for drug access: transferrin receptor (TfR) peptide, low-density lipoprotein receptor 1 (LRP1) peptide, albumin. Each was far-red labeled, injected into two hematogenous models of brain metastases, circulated for two different times, and their uptake quantified in metastases and uninvolved (nonmetastatic) brain. Surprisingly, all three pathways demonstrated distinct distribution patterns in vivo. Two were suboptimal: TfR distributed to uninvolved brain but poorly in metastases, while LRP1 was poorly distributed. Albumin distributed to virtually all metastases in both model systems, significantly greater than in uninvolved brain (P < 0.0001). Further experiments revealed that albumin entered both macrometastases and micrometastases, the targets of treatment and prevention translational strategies. Albumin uptake into brain metastases was not correlated with the uptake of a paracellular probe (biocytin). We identified a novel mechanism of albumin endocytosis through the endothelia of brain metastases consistent with clathrin-independent endocytosis (CIE), involving the neonatal Fc receptor, galectin-3, and glycosphingolipids. Components of the CIE process were found on metastatic endothelial cells in human craniotomies. The data suggest a reconsideration of albumin as a translational mechanism for improved drug delivery to brain metastases and possibly other central nervous system (CNS) cancers.In conclusion, drug therapy for brain metastasis needs improvement. We surveyed three transcytotic pathways as potential delivery systems in brain-tropic models and found that albumin has optimal properties. Albumin used a novel endocytic mechanism.
Collapse
Affiliation(s)
- Imran Khan
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Brunilde Gril
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Anurag N. Paranjape
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Christina M. Robinson
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD
| | - Simone Difilippantonio
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research (FNLCR), Frederick, MD
| | | | | | - Rafał Pęksa
- Department of Pathology, Medical University of Gdańsk, Poland
| | - Renata Duchnowska
- Department of Oncology, Military Institute of Medicine, Warsaw, Poland
| | - Jacek Jassem
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Poland
| | - Priscilla K. Brastianos
- Division of Neuro-Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Philippe Metellus
- Ramsay Général de Santé, Hôpital Privé Clairval, Département de Neurochirurgie and Aix-Marseille Université, Institut de Neurophysiopathologie – UMR 7051, Marseille, France
| | - Emilie Bialecki
- Ramsay Général de Santé, Hôpital Privé Clairval, Département de Neurochirurgie and Aix-Marseille Université, Institut de Neurophysiopathologie – UMR 7051, Marseille, France
| | - Carolyn C. Woodroofe
- Chemistry and Synthesis Center, National Heart, Lung and Blood Institute, NIH, Bethesda, MD
| | - Haitao Wu
- Chemistry and Synthesis Center, National Heart, Lung and Blood Institute, NIH, Bethesda, MD
| | - Rolf E. Swenson
- Chemistry and Synthesis Center, National Heart, Lung and Blood Institute, NIH, Bethesda, MD
| | - Patricia S. Steeg
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
8
|
Lee JH, Chapman DV, Saltzman WM. Nanoparticle Targeting with Antibodies in the Central Nervous System. BME FRONTIERS 2023; 4:0012. [PMID: 37849659 PMCID: PMC10085254 DOI: 10.34133/bmef.0012] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/19/2023] [Indexed: 10/19/2023] Open
Abstract
Treatments for disease in the central nervous system (CNS) are limited because of difficulties in agent penetration through the blood-brain barrier, achieving optimal dosing, and mitigating off-target effects. The prospect of precision medicine in CNS treatment suggests an opportunity for therapeutic nanotechnology, which offers tunability and adaptability to address specific diseases as well as targetability when combined with antibodies (Abs). Here, we review the strategies to attach Abs to nanoparticles (NPs), including conventional approaches of chemisorption and physisorption as well as attempts to combine irreversible Ab immobilization with controlled orientation. We also summarize trends that have been observed through studies of systemically delivered Ab-NP conjugates in animals. Finally, we discuss the future outlook for Ab-NPs to deliver therapeutics into the CNS.
Collapse
Affiliation(s)
| | | | - W. Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| |
Collapse
|
9
|
Voss K, Sewell AE, Krystofiak ES, Gibson-Corley KN, Young AC, Basham JH, Sugiura A, Arner EN, Beavers WN, Kunkle DE, Dickson ME, Needle GA, Skaar EP, Rathmell WK, Ormseth MJ, Major AS, Rathmell JC. Elevated transferrin receptor impairs T cell metabolism and function in systemic lupus erythematosus. Sci Immunol 2023; 8:eabq0178. [PMID: 36638190 PMCID: PMC9936798 DOI: 10.1126/sciimmunol.abq0178] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 12/15/2022] [Indexed: 01/15/2023]
Abstract
T cells in systemic lupus erythematosus (SLE) exhibit multiple metabolic abnormalities. Excess iron can impair mitochondria and may contribute to SLE. To gain insights into this potential role of iron in SLE, we performed a CRISPR screen of iron handling genes on T cells. Transferrin receptor (CD71) was identified as differentially critical for TH1 and inhibitory for induced regulatory T cells (iTregs). Activated T cells induced CD71 and iron uptake, which was exaggerated in SLE-prone T cells. Cell surface CD71 was enhanced in SLE-prone T cells by increased endosomal recycling. Blocking CD71 reduced intracellular iron and mTORC1 signaling, which inhibited TH1 and TH17 cells yet enhanced iTregs. In vivo treatment reduced kidney pathology and increased CD4 T cell production of IL-10 in SLE-prone mice. Disease severity correlated with CD71 expression on TH17 cells from patients with SLE, and blocking CD71 in vitro enhanced IL-10 secretion. T cell iron uptake via CD71 thus contributes to T cell dysfunction and can be targeted to limit SLE-associated pathology.
Collapse
Affiliation(s)
- Kelsey Voss
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Allison E. Sewell
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Evan S. Krystofiak
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| | - Katherine N. Gibson-Corley
- Division of Comparative Medicine, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Arissa C. Young
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jacob H. Basham
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ayaka Sugiura
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Emily N. Arner
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - William N. Beavers
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Dillon E. Kunkle
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Megan E. Dickson
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gabriel A. Needle
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric P. Skaar
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Chemical Biology, Vanderbilt University, Nashville, TN, USA
| | - W. Kimryn Rathmell
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michelle J. Ormseth
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, TN, USA
| | - Amy S. Major
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, TN, USA
| | - Jeffrey C. Rathmell
- Division of Molecular Pathogenesis, Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
10
|
Molecular Imaging of Ultrasound-Mediated Blood-Brain Barrier Disruption in a Mouse Orthotopic Glioblastoma Model. Pharmaceutics 2022; 14:pharmaceutics14102227. [PMID: 36297663 PMCID: PMC9610067 DOI: 10.3390/pharmaceutics14102227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/17/2022] Open
Abstract
Glioblastoma (GBM) is an aggressive and malignant primary brain tumor. The blood-brain barrier (BBB) limits the therapeutic options available to tackle this incurable tumor. Transient disruption of the BBB by focused ultrasound (FUS) is a promising and safe approach to increase the brain and tumor concentration of drugs administered systemically. Non-invasive, sensitive, and reliable imaging approaches are required to better understand the impact of FUS on the BBB and brain microenvironment. In this study, nuclear imaging (SPECT/CT and PET/CT) was used to quantify neuroinflammation 48 h post-FUS and estimate the influence of FUS on BBB opening and tumor growth in vivo. BBB disruptions were performed on healthy and GBM-bearing mice (U-87 MG xenograft orthotopic model). The BBB recovery kinetics were followed and quantified by [99mTc]Tc-DTPA SPECT/CT imaging at 0.5 h, 3 h and 24 h post-FUS. The absence of neuroinflammation was confirmed by [18F]FDG PET/CT imaging 48 h post-FUS. The presence of the tumor and its growth were evaluated by [68Ga]Ga-RGD2 PET/CT imaging and post-mortem histological analysis, showing that tumor growth was not influenced by FUS. In conclusion, molecular imaging can be used to evaluate the time frame for systemic treatment combined with transient BBB opening and to test its efficacy over time.
Collapse
|
11
|
Quantum dots: The cutting-edge nanotheranostics in brain cancer management. J Control Release 2022; 350:698-715. [PMID: 36057397 DOI: 10.1016/j.jconrel.2022.08.047] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 12/14/2022]
Abstract
Quantum dots (QDs) are semiconductor nanocrystals possessing unique optoelectrical properties in that they can emit light energy of specific tunable wavelengths when excited by photons. They are gaining attention nowadays owing to their all-around ability to allow high-quality bio-imaging along with targeted drug delivery. The most lethal central nervous system (CNS) disorders are brain cancers or malignant brain tumors. CNS is guarded by the blood-brain barrier which poses a selective blockade toward drug delivery into the brain. QDs have displayed strong potential to deliver therapeutic agents into the brain successfully. Their bio-imaging capability due to photoluminescence and specific targeting ability through the attachment of ligand biomolecules make them preferable clinical tools for coming times. Biocompatible QDs are emerging as nanotheranostic tools to identify/diagnose and selectively kill cancer cells. The current review focuses on QDs and associated nanoformulations as potential futuristic clinical aids in the continuous battle against brain cancer.
Collapse
|
12
|
Pandey N, Anastasiadis P, Carney CP, Kanvinde PP, Woodworth GF, Winkles JA, Kim AJ. Nanotherapeutic treatment of the invasive glioblastoma tumor microenvironment. Adv Drug Deliv Rev 2022; 188:114415. [PMID: 35787387 PMCID: PMC10947564 DOI: 10.1016/j.addr.2022.114415] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/20/2022] [Accepted: 06/26/2022] [Indexed: 12/11/2022]
Abstract
Glioblastoma (GBM) is the most common malignant adult brain cancer with no curative treatment strategy. A significant hurdle in GBM treatment is effective therapeutic delivery to the brain-invading tumor cells that remain following surgery within functioning brain regions. Developing therapies that can either directly target these brain-invading tumor cells or act on other cell types and molecular processes supporting tumor cell invasion and recurrence are essential steps in advancing new treatments in the clinic. This review highlights some of the drug delivery strategies and nanotherapeutic technologies that are designed to target brain-invading GBM cells or non-neoplastic, invasion-supporting cells residing within the GBM tumor microenvironment.
Collapse
Affiliation(s)
- Nikhil Pandey
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Pavlos Anastasiadis
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Christine P Carney
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Pranjali P Kanvinde
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Graeme F Woodworth
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, MD, 20742, United States
| | - Jeffrey A Winkles
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, MD 21201, United States.
| | - Anthony J Kim
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, United States; Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, United States; Fischell Department of Bioengineering, A. James Clarke School of Engineering, University of Maryland, College Park, MD, 20742, United States.
| |
Collapse
|
13
|
Ghouri MD, Saleem J, Ren J, Liu J, Umer A, Cai R, Chen C. Nanomaterials‐Mediated Structural and Physiological Modulation of Blood Brain Barrier for Therapeutic Purposes. ADVANCED MATERIALS INTERFACES 2022; 9. [DOI: 10.1002/admi.202101391] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Indexed: 01/06/2025]
Abstract
AbstractBlood brain barrier (BBB) protects homeostasis and sensitive environment of brain from several toxic substances coming from the systemic circulation. This barrier along with those substances also prevents therapeutic chemicals to reach brain tissues for several brain diseases. BBB consists of a number of cell types and junctions that help maintain its intricate structure and physiology. To open BBB for therapeutic purposes, researchers are keen to explore the use of nanomaterials as therapeutic agents. Nanomaterials have unique physio‐chemical properties such as, increased surface area to mass ratio, superior adsorption capacity, and a wide variety of functionalization possibilities in contrast to bulk materials, making them sought‐after for research pertaining to brain delivery of therapeutic substances. Both organic and inorganic nanomaterials have been researched in this regard with numerous interesting functionalizations, and their toxicity and distribution profiles have been well assessed. Different pathways taken up by nanomaterials to cross BBB like adsorptive‐mediated transcytosis, inhibition of active efflux pumps, receptor‐mediated transport, and cell‐mediated endocytosis have also been investigated. This review summarizes the structural and physiological properties and the modulation techniques of BBB for delivery of adsorbed/functionalized nano delivery platforms and imaging nanomaterials across.
Collapse
Affiliation(s)
- Muhammad Daniyal Ghouri
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Chinese Academy of Sciences (CAS) Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Jabran Saleem
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Chinese Academy of Sciences (CAS) Beijing 100190 China
| | - Jiayu Ren
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Chinese Academy of Sciences (CAS) Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Jiaming Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Chinese Academy of Sciences (CAS) Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Arsalan Umer
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Chinese Academy of Sciences (CAS) Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Rong Cai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Chinese Academy of Sciences (CAS) Beijing 100190 China
- GBA National Institute for Nanotechnology Innovation Guangdong 510700 China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology of China Chinese Academy of Sciences (CAS) Beijing 100190 China
- University of Chinese Academy of Sciences Beijing 100049 China
- GBA National Institute for Nanotechnology Innovation Guangdong 510700 China
| |
Collapse
|
14
|
Som Chaudhury S, Sinha K, Das Mukhopadhyay C. Intranasal route: The green corridor for Alzheimer's disease therapeutics. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
15
|
Alotaibi BS, Buabeid M, Ibrahim NA, Kharaba ZJ, Ijaz M, Noreen S, Murtaza G. Potential of Nanocarrier-Based Drug Delivery Systems for Brain Targeting: A Current Review of Literature. Int J Nanomedicine 2021; 16:7517-7533. [PMID: 34795481 PMCID: PMC8593899 DOI: 10.2147/ijn.s333657] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/18/2021] [Indexed: 12/18/2022] Open
Abstract
The advent of nanotechnologies such as nanocarriers and nanotherapeutics has changed the treatment strategy and developed a more efficacious novel drug delivery system. Various drug delivery systems are focused on drug-targeting of brain cells. However, the manifestation of the brain barrier is the main hurdle for the effective delivery of chemotherapeutics, ultimately causing treatment failure of various drugs. To solve this problem, various nanocarrier-based drug delivery system has been developed for brain targeting. This review outlines nanocarrier-based composites for different brain diseases and highlights nanocarriers for drug targeting towards brain cells. It also summarizes the latest developments in nanocarrier-based delivery systems containing liposomal systems, dendrimers, polymeric micelles, polymeric nanocarriers, quantum dots (QDs), and gold nanoparticles. Besides, the optimal properties of nanocarriers and therapeutic implications for brain targeting have been extensively studied. Finally, the potential applications and research opportunities for nanocarriers in brain targeting are discussed.
Collapse
Affiliation(s)
- Badriyah Shadid Alotaibi
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Manal Buabeid
- Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
- Department of Clinical Sciences, Ajman University, Ajman, 346, United Arab Emirates
| | - Nihal Abdalla Ibrahim
- Medical and Bio-allied Health Sciences Research Centre, Ajman University, Ajman, United Arab Emirates
- Department of Clinical Sciences, Ajman University, Ajman, 346, United Arab Emirates
| | - Zelal Jaber Kharaba
- Department of Clinical Sciences, College of Pharmacy, Al-Ain University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Munazza Ijaz
- Institute of Molecular Biology and Biotechnology, the University of Lahore, Lahore, Pakistan
| | - Sobia Noreen
- Department of Pharmaceutics, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Ghulam Murtaza
- Department of Pharmacy, COMSATS University Islamabad, Lahore Campus, Lahore, 54000, Pakistan
| |
Collapse
|
16
|
Villalva MD, Agarwal V, Ulanova M, Sachdev PS, Braidy N. Quantum dots as a theranostic approach in Alzheimer's disease: a systematic review. Nanomedicine (Lond) 2021; 16:1595-1611. [PMID: 34180261 DOI: 10.2217/nnm-2021-0104] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Aim: Quantum dots (QDs) are nanoparticles that have an emerging application as theranostic agents in several neurodegenerative diseases. The advantage of QDs as nanomedicine is due to their unique optical properties that provide high sensitivity, stability and selectivity at a nanoscale range. Objective: To offer renewed insight into current QD research and elucidate its promising application in Alzheimer's disease (AD) diagnosis and therapy. Methods: A comprehensive literature search was conducted in PubMed and Google Scholar databases that included the following search terms: 'quantum dots', 'blood-brain barrier', 'cytotoxicity', 'toxicity' and 'Alzheimer's disease'; PRISMA guidelines were adhered to. Results: Thirty-four publications were selected to evaluate the ability of QDs to cross the blood-brain barrier, potential toxicity and current AD diagnostic and therapeutic applications. Conclusion: QD's unique optical properties and versatility to conjugate to various biomolecules, while maintaining a nanoscale size, render them a promising theranostic tool in AD.
Collapse
Affiliation(s)
- Maria D Villalva
- Centre for Healthy Brain Aging, School of Psychiatry, University of New South Wales (UNSW), Sydney, Australia
| | - Vipul Agarwal
- Cluster for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney, Australia
| | - Marina Ulanova
- Centre for Healthy Brain Aging, School of Psychiatry, University of New South Wales (UNSW), Sydney, Australia
| | - Perminder S Sachdev
- Centre for Healthy Brain Aging, School of Psychiatry, University of New South Wales (UNSW), Sydney, Australia.,Neuropsychiatric Institute, Euroa Centre, Prince of Wales Hospital, Sydney, Australia
| | - Nady Braidy
- Centre for Healthy Brain Aging, School of Psychiatry, University of New South Wales (UNSW), Sydney, Australia
| |
Collapse
|
17
|
Delivery of Therapeutic Agents to the Central Nervous System and the Promise of Extracellular Vesicles. Pharmaceutics 2021; 13:pharmaceutics13040492. [PMID: 33916841 PMCID: PMC8067091 DOI: 10.3390/pharmaceutics13040492] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 12/17/2022] Open
Abstract
The central nervous system (CNS) is surrounded by the blood–brain barrier (BBB), a semipermeable border of endothelial cells that prevents pathogens, solutes and most molecules from non-selectively crossing into the CNS. Thus, the BBB acts to protect the CNS from potentially deleterious insults. Unfortunately, the BBB also frequently presents a significant barrier to therapies, impeding passage of drugs and biologicals to target cells within the CNS. This review provides an overview of different approaches to deliver therapeutics across the BBB, with an emphasis in extracellular vesicles as delivery vehicles to the CNS.
Collapse
|
18
|
D'Souza A, Dave KM, Stetler RA, S. Manickam D. Targeting the blood-brain barrier for the delivery of stroke therapies. Adv Drug Deliv Rev 2021; 171:332-351. [PMID: 33497734 DOI: 10.1016/j.addr.2021.01.015] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 02/06/2023]
Abstract
A variety of neuroprotectants have shown promise in treating ischemic stroke, yet their delivery to the brain remains a challenge. The endothelial cells lining the blood-brain barrier (BBB) are emerging as a dynamic factor in the response to neurological injury and disease, and the endothelial-neuronal matrix coupling is fundamentally neuroprotective. In this review, we discuss approaches that target the endothelium for drug delivery both across the BBB and to the BBB as a viable strategy to facilitate neuroprotective effects, using the example of brain-derived neurotrophic factor (BDNF). We highlight the advances in cell-derived extracellular vesicles (EVs) used for CNS targeting and drug delivery. We also discuss the potential of engineered EVs as a potent strategy to deliver BDNF or other drug candidates to the ischemic brain, particularly when coupled with internal components like mitochondria that may increase cellular energetics in injured endothelial cells.
Collapse
|
19
|
Vinod C, Jena S. Nano-Neurotheranostics: Impact of Nanoparticles on Neural Dysfunctions and Strategies to Reduce Toxicity for Improved Efficacy. Front Pharmacol 2021; 12:612692. [PMID: 33841144 PMCID: PMC8033012 DOI: 10.3389/fphar.2021.612692] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
Nanotheranostics is one of the emerging research areas in the field of nanobiotechnology offering exciting promises for diagnosis, bio-separation, imaging mechanisms, hyperthermia, phototherapy, chemotherapy, drug delivery, gene delivery, among other uses. The major criteria for any nanotheranostic-materials is 1) to interact with proteins and cells without meddling with their basic activities, 2) to maintain their physical properties after surface modifications and 3) must be nontoxic. One of the challenging targets for nanotheranostics is the nervous system with major hindrances from the neurovascular units, the functional units of blood-brain barrier. As blood-brain barrier is crucial for protecting the CNS from toxins and metabolic fluctuations, most of the synthetic nanomaterials cannot pass through this barrier making it difficult for diagnosing or targeting the cells. Biodegradable nanoparticles show a promising role in this aspect. Certain neural pathologies have compromised barrier creating a path for most of the nanoparticles to enter into the cells. However, such carriers may pose a risk of side effects to non-neural tissues and their toxicity needs to be elucidated at preclinical levels. This article reviews about the different types of nanotheranostic strategies applied in nervous dysfunctions. Further, the side effects of these carriers are reviewed and appropriate methods to test the toxicity of such nano-carriers are suggested to improve the effectiveness of nano-carrier based diagnosis and treatments.
Collapse
Affiliation(s)
- Chiluka Vinod
- Department of Biological Sciences, School of Applied Sciences, KIIT University, Bhubaneswar, India
| | - Srikanta Jena
- Department of Zoology, School of Life Sciences, Ravenshaw University, Cuttack, India
| |
Collapse
|
20
|
Leclerc M, Dudonné S, Calon F. Can Natural Products Exert Neuroprotection without Crossing the Blood-Brain Barrier? Int J Mol Sci 2021; 22:ijms22073356. [PMID: 33805947 PMCID: PMC8037419 DOI: 10.3390/ijms22073356] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 12/17/2022] Open
Abstract
The scope of evidence on the neuroprotective impact of natural products has been greatly extended in recent years. However, a key question that remains to be answered is whether natural products act directly on targets located in the central nervous system (CNS), or whether they act indirectly through other mechanisms in the periphery. While molecules utilized for brain diseases are typically bestowed with a capacity to cross the blood–brain barrier, it has been recently uncovered that peripheral metabolism impacts brain functions, including cognition. The gut–microbiota–brain axis is receiving increasing attention as another indirect pathway for orally administered compounds to act on the CNS. In this review, we will briefly explore these possibilities focusing on two classes of natural products: omega-3 polyunsaturated fatty acids (n-3 PUFAs) from marine sources and polyphenols from plants. The former will be used as an example of a natural product with relatively high brain bioavailability but with tightly regulated transport and metabolism, and the latter as an example of natural compounds with low brain bioavailability, yet with a growing amount of preclinical and clinical evidence of efficacy. In conclusion, it is proposed that bioavailability data should be sought early in the development of natural products to help identifying relevant mechanisms and potential impact on prevalent CNS disorders, such as Alzheimer’s disease.
Collapse
Affiliation(s)
- Manon Leclerc
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada;
- Axe Neurosciences, Centre de Recherche du CHU de Québec–Université Laval, Québec, QC G1V 4G2, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, QC G1V 0A6, Canada;
- OptiNutriBrain-Laboratoire International Associé (NutriNeuro France-INAF Canada), Québec, QC G1V 0A6, Canada
| | - Stéphanie Dudonné
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, QC G1V 0A6, Canada;
- OptiNutriBrain-Laboratoire International Associé (NutriNeuro France-INAF Canada), Québec, QC G1V 0A6, Canada
| | - Frédéric Calon
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada;
- Axe Neurosciences, Centre de Recherche du CHU de Québec–Université Laval, Québec, QC G1V 4G2, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Québec, QC G1V 0A6, Canada;
- OptiNutriBrain-Laboratoire International Associé (NutriNeuro France-INAF Canada), Québec, QC G1V 0A6, Canada
- Correspondence: ; Tel.: +1-(418)-525-4444 (ext. 48697); Fax: +1-(418)-654-2761
| |
Collapse
|
21
|
Hempel C, Kapishnikov S, Perez-Berna AJ, Werner S, Guttmann P, Pereiro E, Qvortrup K, Andresen TL. The need to freeze-Dehydration during specimen preparation for electron microscopy collapses the endothelial glycocalyx regardless of fixation method. Microcirculation 2020; 27:e12643. [PMID: 32542908 DOI: 10.1111/micc.12643] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/25/2020] [Accepted: 06/06/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVE The endothelial glycocalyx covers the luminal surface of the endothelium and plays key roles in vascular function. Despite its biological importance, ideal visualization techniques are lacking. The current study aimed to improve the preservation and subsequent imaging quality of the endothelial glycocalyx. METHODS In mice, the endothelial glycocalyx was contrasted with a mixture of lanthanum and dysprosium (LaDy). Standard chemical fixation was compared with high-pressure frozen specimens processed with freeze substitution. Also, isolated brain microvessels and cultured endothelial cells were high-pressure frozen and by transmission soft x-rays, imaged under cryogenic conditions. RESULTS The endothelial glycocalyx was in some tissues significantly more voluminous from chemically fixed specimens compared with high-pressure frozen specimens. LaDy labeling introduced excessive absorption contrast, which impeded glycocalyx measurements in isolated brain microvessels when using transmission soft x-rays. In non-contrasted vessels, the glycocalyx was not resolved. LaDy-contrasted, cultured brain endothelial cells allowed to assess glycocalyx volume in vitro. CONCLUSIONS Both chemical and cryogenic fixation followed by dehydration lead to substantial collapse of the glycocalyx. Cryogenic fixation without freeze substitution could be a way forward although transmission soft x-ray tomography based solely on amplitude contrast seems unsuitable.
Collapse
Affiliation(s)
- Casper Hempel
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark.,Centre for Medical Parasitology, Department for Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark.,Department for Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Sergey Kapishnikov
- Department X-Ray microscopy, Helmholtz-Zentrum Berlin für Materialien und Energie GmbH, Berlin, Germany.,Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark
| | | | - Stephan Werner
- Department X-Ray microscopy, Helmholtz-Zentrum Berlin für Materialien und Energie GmbH, Berlin, Germany
| | - Peter Guttmann
- Department X-Ray microscopy, Helmholtz-Zentrum Berlin für Materialien und Energie GmbH, Berlin, Germany
| | - Eva Pereiro
- MISTRAL Beamline-Experiments Division, ALBA Synchrotron Light Source, Barcelona, Spain
| | - Klaus Qvortrup
- Core Facility for Integrated Microscopy (CFIM), University of Copenhagen, Copenhagen, Denmark
| | - Thomas Lars Andresen
- Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| |
Collapse
|
22
|
Yang AC, Stevens MY, Chen MB, Lee DP, Stähli D, Gate D, Contrepois K, Chen W, Iram T, Zhang L, Vest RT, Chaney A, Lehallier B, Olsson N, du Bois H, Hsieh R, Cropper HC, Berdnik D, Li L, Wang EY, Traber GM, Bertozzi CR, Luo J, Snyder MP, Elias JE, Quake SR, James ML, Wyss-Coray T. Physiological blood-brain transport is impaired with age by a shift in transcytosis. Nature 2020; 583:425-430. [PMID: 32612231 DOI: 10.1038/s41586-020-2453-z] [Citation(s) in RCA: 297] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 05/21/2020] [Indexed: 12/31/2022]
Abstract
The vascular interface of the brain, known as the blood-brain barrier (BBB), is understood to maintain brain function in part via its low transcellular permeability1-3. Yet, recent studies have demonstrated that brain ageing is sensitive to circulatory proteins4,5. Thus, it is unclear whether permeability to individually injected exogenous tracers-as is standard in BBB studies-fully represents blood-to-brain transport. Here we label hundreds of proteins constituting the mouse blood plasma proteome, and upon their systemic administration, study the BBB with its physiological ligand. We find that plasma proteins readily permeate the healthy brain parenchyma, with transport maintained by BBB-specific transcriptional programmes. Unlike IgG antibody, plasma protein uptake diminishes in the aged brain, driven by an age-related shift in transport from ligand-specific receptor-mediated to non-specific caveolar transcytosis. This age-related shift occurs alongside a specific loss of pericyte coverage. Pharmacological inhibition of the age-upregulated phosphatase ALPL, a predicted negative regulator of transport, enhances brain uptake of therapeutically relevant transferrin, transferrin receptor antibody and plasma. These findings reveal the extent of physiological protein transcytosis to the healthy brain, a mechanism of widespread BBB dysfunction with age and a strategy for enhanced drug delivery.
Collapse
Affiliation(s)
- Andrew C Yang
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA.,ChEM-H, Stanford University, Stanford, CA, USA.,Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Marc Y Stevens
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle B Chen
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Davis P Lee
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel Stähli
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - David Gate
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Winnie Chen
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Tal Iram
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Ryan T Vest
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Department of Chemical Engineering, Stanford, CA, USA
| | - Aisling Chaney
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Benoit Lehallier
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Niclas Olsson
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA, USA.,Calico Life Sciences LLC, South San Francisco, CA, USA
| | - Haley du Bois
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Ryan Hsieh
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Haley C Cropper
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniela Berdnik
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Lulin Li
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Elizabeth Y Wang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Gavin M Traber
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Carolyn R Bertozzi
- ChEM-H, Stanford University, Stanford, CA, USA.,Department of Chemistry, Stanford University, Stanford, CA, USA.,Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Jian Luo
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Veterans Administration Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Stephen R Quake
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA.,Chan Zuckerberg Biohub, Stanford, CA, USA
| | - Michelle L James
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.,Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA.,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Tony Wyss-Coray
- ChEM-H, Stanford University, Stanford, CA, USA. .,Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Chemistry, Stanford University, Stanford, CA, USA. .,Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA. .,Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
23
|
Filali S, Pirot F, Miossec P. Biological Applications and Toxicity Minimization of Semiconductor Quantum Dots. Trends Biotechnol 2020; 38:163-177. [DOI: 10.1016/j.tibtech.2019.07.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/27/2019] [Accepted: 07/30/2019] [Indexed: 12/18/2022]
|
24
|
Targeting and imaging of monocyte-derived macrophages in rat's injured artery following local delivery of liposomal quantum dots. J Control Release 2020; 318:145-157. [DOI: 10.1016/j.jconrel.2019.12.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 11/26/2019] [Accepted: 12/08/2019] [Indexed: 12/27/2022]
|
25
|
Analyzing Nanotheraputics-Based Approaches for the Management of Psychotic Disorders. J Pharm Sci 2019; 108:3757-3768. [DOI: 10.1016/j.xphs.2019.08.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/07/2019] [Accepted: 08/29/2019] [Indexed: 12/12/2022]
|
26
|
Johnsen KB, Burkhart A, Thomsen LB, Andresen TL, Moos T. Targeting the transferrin receptor for brain drug delivery. Prog Neurobiol 2019; 181:101665. [DOI: 10.1016/j.pneurobio.2019.101665] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/10/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023]
|
27
|
Zhang M, Bishop BP, Thompson NL, Hildahl K, Dang B, Mironchuk O, Chen N, Aoki R, Holmberg VC, Nance E. Quantum Dot Cellular Uptake and Toxicity in the Developing Brain: Implications for Use as Imaging Probes. NANOSCALE ADVANCES 2019; 1:3424-3442. [PMID: 31867563 PMCID: PMC6924642 DOI: 10.1039/c9na00334g] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/27/2019] [Indexed: 05/30/2023]
Abstract
Nanometer-sized luminescent semiconductor quantum dots (QDs) have been utilized as imaging and therapeutic agents in a variety of disease settings, including diseases of the central nervous system. QDs have several advantages over traditional fluorescent probes including their small size (5-10 nm), tunable excitation and emission spectra, tailorable surface functionality, efficient photoluminescence, and robust photostability, which are ideal characteristics for in vivo imaging. Although QDs are promising imaging agents in brain-related applications, no systematic evaluation of QD behavior in brain-relevant conditions has yet been done. Therefore, we sought to investigate QD colloidal stability, cellular uptake, and toxicity in vitro, ex vivo, and in vivo in the brain environment. We found that QD behavior is highly dependent on surface functionality and that treatment of cultured organotypic whole hemisphere (OWH) slices with QDs results in dose-dependent toxicity and metallothionein increase, but no subsequent mRNA expression level changes in inflammatory cytokines or other oxidative stress. QDs coated with poly(ethylene glycol) (PEG) were protected from aggregation in neurophysiologically relevant fluids and in tissue, allowing for greater penetration. Importantly, QD behavior differed in cultured slices as compared to monolayer cell cultures, and behavior in cultured slices aligned more closely with that seen in vivo. Irrespective of surface chemistry and brain-relevant platform, non-aggregated QDs were primarily internalized by microglia in a region-dependent manner both in slices and in vivo upon systemic administration. This knowledge will help guide further engineering of candidate QD-based imaging probes for neurological application.
Collapse
Affiliation(s)
- Mengying Zhang
- Molecular Engineering & Sciences Institute, University of WashingtonSeattleWA 98195-1652USA
| | - Brittany P. Bishop
- Department of Chemical Engineering, University of WashingtonSeattleWA 98195-1750USA
| | - Nicole L. Thompson
- Department of Chemical Engineering, University of WashingtonSeattleWA 98195-1750USA
| | - Kate Hildahl
- Department of Chemical Engineering, University of WashingtonSeattleWA 98195-1750USA
| | - Binh Dang
- Department of Chemical Engineering, University of WashingtonSeattleWA 98195-1750USA
| | - Olesya Mironchuk
- Department of Bioengineering, University of WashingtonSeattleWA 98195-5061USA
| | - Nina Chen
- Department of Biology, University of WashingtonSeattleWA 98195-1800USA
| | - Reyn Aoki
- Department of Chemical Engineering, University of WashingtonSeattleWA 98195-1750USA
| | - Vincent C. Holmberg
- Molecular Engineering & Sciences Institute, University of WashingtonSeattleWA 98195-1652USA
- Department of Chemical Engineering, University of WashingtonSeattleWA 98195-1750USA
- Clean Energy Institute, University of WashingtonSeattleWA 98195-1653USA
| | - Elizabeth Nance
- Molecular Engineering & Sciences Institute, University of WashingtonSeattleWA 98195-1652USA
- Department of Chemical Engineering, University of WashingtonSeattleWA 98195-1750USA
- Center on Human Development and Disability, University of WashingtonSeattleWA 98195-7920USA
- Department of Radiology, University of WashingtonSeattleWA 98195-7117USA
| |
Collapse
|
28
|
Samal J, Rebelo AL, Pandit A. A window into the brain: Tools to assess pre-clinical efficacy of biomaterials-based therapies on central nervous system disorders. Adv Drug Deliv Rev 2019; 148:68-145. [PMID: 30710594 DOI: 10.1016/j.addr.2019.01.012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 01/04/2019] [Accepted: 01/28/2019] [Indexed: 12/13/2022]
Abstract
Therapeutic conveyance into the brain is a cardinal requirement for treatment of diverse central nervous system (CNS) disorders and associated pathophysiology. Effectual shielding of the brain by the blood-brain barrier (BBB) sieves out major proportion of therapeutics with the exception of small lipophilic molecules. Various nano-delivery systems (NDS) provide an effective solution around this obstacle owing to their small size and targeting properties. To date, these systems have been used for several pre-clinical disease models including glioma, neurodegenerative diseases and psychotic disorders. An efficacy screen for these systems involves a test battery designed to probe into the multiple facets of therapeutic delivery. Despite their wide application in redressing various disease targets, the efficacy evaluation strategies for all can be broadly grouped into four modalities, namely: histological, bio-imaging, molecular and behavioural. This review presents a comprehensive insight into all of these modalities along with their strengths and weaknesses as well as perspectives on an ideal design for a panel of tests to screen brain nano-delivery systems.
Collapse
Affiliation(s)
- Juhi Samal
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Ana Lucia Rebelo
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, Centre for Research in Medical Devices, National University of Ireland Galway, Galway, Ireland.
| |
Collapse
|
29
|
Bourassa P, Alata W, Tremblay C, Paris-Robidas S, Calon F. Transferrin Receptor-Mediated Uptake at the Blood-Brain Barrier Is Not Impaired by Alzheimer's Disease Neuropathology. Mol Pharm 2019; 16:583-594. [PMID: 30609376 DOI: 10.1021/acs.molpharmaceut.8b00870] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The transferrin receptor (TfR) is highly expressed by brain capillary endothelial cells (BCECs) forming the blood-brain barrier (BBB) and is therefore considered as a potential target for brain drug delivery. Monoclonal antibodies binding to the TfR, such as clone Ri7, have been shown to internalize into BCECs in vivo. However, since Alzheimer's disease (AD) is accompanied by a BBB dysfunction, it raises concerns about whether TfR-mediated transport becomes inefficient during the progression of the disease. Measurements of TfR levels using Western blot analysis in whole homogenates from human post-mortem parietal cortex and hippocampus did not reveal any significant difference between individuals with or without a neuropathological diagnosis of AD (respectively, n = 19 and 22 for the parietal cortex and n = 12 and 14 for hippocampus). Similarly, TfR concentrations in isolated human brain microvessels from parietal cortex were similar between controls and AD cases. TfR levels in isolated murine brain microvessels were not significantly different between groups of 12- and 18-month-old NonTg and 3xTg-AD mice, the latter modeling Aβ and τ neuropathologies. In situ brain perfusion assays were then conducted to measure the brain uptake and internalization of fluorolabeled Ri7 in BCECs upon binding. Consistently, TfR-mediated uptake in BCECs was similar between 3xTg-AD mice and nontransgenic controls (∼0.3 μL·g-1·s-1) at 12, 18, and 22 months of age. Fluorescence microscopy analysis following intravenous administration of fluorolabeled Ri7 highlighted that the signal from the antibody was widely distributed throughout the cerebral vasculature but not in neurons or astrocytes. Overall, our data suggest that both TfR protein levels and TfR-dependent internalization mechanisms are preserved in the presence of Aβ and τ neuropathologies, supporting the potential of TfR as a vector target for drug delivery into BCECs in AD.
Collapse
Affiliation(s)
- Philippe Bourassa
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Wael Alata
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Cyntia Tremblay
- Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Sarah Paris-Robidas
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| | - Frédéric Calon
- Faculté de Pharmacie , Université Laval , Québec , QC G1V 0A6 , Canada.,Axe Neurosciences , Centre de Recherche du CHU de Québec-Université Laval , Québec , QC G1V 4G2 , Canada
| |
Collapse
|
30
|
Pulgar VM. Transcytosis to Cross the Blood Brain Barrier, New Advancements and Challenges. Front Neurosci 2019; 12:1019. [PMID: 30686985 PMCID: PMC6337067 DOI: 10.3389/fnins.2018.01019] [Citation(s) in RCA: 292] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 12/18/2018] [Indexed: 01/21/2023] Open
Abstract
The blood brain barrier (BBB) presents a formidable challenge to the delivery of drugs into the brain. Several strategies aim to overcome this obstacle and promote efficient and specific crossing through BBB of therapeutically relevant agents. One of those strategies uses the physiological process of receptor-mediated transcytosis (RMT) to transport cargo through the brain endothelial cells toward brain parenchyma. Recent developments in our understanding of intracellular trafficking and receptor binding as well as in protein engineering and nanotechnology have potentiated the opportunities for treatment of CNS diseases using RMT. In this mini-review, the current understanding of BBB structure is discussed, and recent findings exemplifying critical advances in RMT-mediated brain drug delivery are briefly presented.
Collapse
Affiliation(s)
- Victor M Pulgar
- Department of Pharmaceutical Sciences, Campbell University, Buies Creek, NC, United States.,Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
31
|
Johnsen KB, Bak M, Kempen PJ, Melander F, Burkhart A, Thomsen MS, Nielsen MS, Moos T, Andresen TL. Antibody affinity and valency impact brain uptake of transferrin receptor-targeted gold nanoparticles. Theranostics 2018; 8:3416-3436. [PMID: 29930740 PMCID: PMC6010987 DOI: 10.7150/thno.25228] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/17/2018] [Indexed: 11/15/2022] Open
Abstract
Rationale: The ability to treat invalidating neurological diseases is impeded by the presence of the blood-brain barrier (BBB), which inhibits the transport of most blood-borne substances into the brain parenchyma. Targeting the transferrin receptor (TfR) on the surface of brain capillaries has been a popular strategy to give a preferential accumulation of drugs or nanomedicines, but several aspects of this targeting strategy remain elusive. Here we report that TfR-targeted gold nanoparticles (AuNPs) can accumulate in brain capillaries and further transport across the BBB to enter the brain parenchyma. Methods: We characterized our targeting strategy both in vitro using primary models of the BBB and in vivo using quantitative measurements of gold accumulation by inductively-coupled plasma-mass spectrometry together with morphological assessments using light microscopy after silver enhancement and transmission electron microscopy with energy-dispersive X-ray spectroscopy. Results: We find that the uptake capacity is significantly modulated by the affinity and valency of the AuNP-conjugated antibodies. Specifically, antibodies with high and low affinities mediate a low and intermediate uptake of AuNPs into the brain, respectively, whereas a monovalent (bi-specific) antibody improves the uptake capacity remarkably. Conclusion: Our findings indicate that monovalent ligands may be beneficial for obtaining transcytosis of TfR-targeted nanomedicines across the BBB, which is relevant for future design of nanomedicines for brain drug delivery.
Collapse
|
32
|
Kumar A, Tan A, Wong J, Spagnoli JC, Lam J, Blevins BD, G N, Thorne L, Ashkan K, Xie J, Liu H. Nanotechnology for Neuroscience: Promising Approaches for Diagnostics, Therapeutics and Brain Activity Mapping. ADVANCED FUNCTIONAL MATERIALS 2017; 27:1700489. [PMID: 30853878 PMCID: PMC6404766 DOI: 10.1002/adfm.201700489] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Unlocking the secrets of the brain is a task fraught with complexity and challenge - not least due to the intricacy of the circuits involved. With advancements in the scale and precision of scientific technologies, we are increasingly equipped to explore how these components interact to produce a vast range of outputs that constitute function and disease. Here, an insight is offered into key areas in which the marriage of neuroscience and nanotechnology has revolutionized the industry. The evolution of ever more sophisticated nanomaterials culminates in network-operant functionalized agents. In turn, these materials contribute to novel diagnostic and therapeutic strategies, including drug delivery, neuroprotection, neural regeneration, neuroimaging and neurosurgery. Further, the entrance of nanotechnology into future research arenas including optogenetics, molecular/ion sensing and monitoring, and piezoelectric effects is discussed. Finally, considerations in nanoneurotoxicity, the main barrier to clinical translation, are reviewed, and direction for future perspectives is provided.
Collapse
Affiliation(s)
- Anil Kumar
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China
| | - Aaron Tan
- UCL Medical School, University College London (UCL), London, United Kingdom
| | - Joanna Wong
- Imperial College School of Medicine, Imperial College London,London, United Kingdom
| | - Jonathan Clayton Spagnoli
- Department of Chemistry, Bio-Imaging Research Center, University of Georgia, Athens, Georgia 30602, United States
| | - James Lam
- UCL Medical School, University College London (UCL), London, United Kingdom
| | - Brianna Diane Blevins
- Department of Chemistry, Bio-Imaging Research Center, University of Georgia, Athens, Georgia 30602, United States
| | - Natasha G
- UCL Medical School, University College London (UCL), London, United Kingdom
| | - Lewis Thorne
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, Queen Square, London, United Kingdom
| | - Keyoumars Ashkan
- Department of Neurosurgery, King's College Hospital NHS Foundation Trust, King's College London, London, United Kingdom
| | - Jin Xie
- Department of Chemistry, Bio-Imaging Research Center, University of Georgia, Athens, Georgia 30602, United States
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan 250100, China
| |
Collapse
|
33
|
Johnsen KB, Burkhart A, Melander F, Kempen PJ, Vejlebo JB, Siupka P, Nielsen MS, Andresen TL, Moos T. Targeting transferrin receptors at the blood-brain barrier improves the uptake of immunoliposomes and subsequent cargo transport into the brain parenchyma. Sci Rep 2017; 7:10396. [PMID: 28871203 PMCID: PMC5583399 DOI: 10.1038/s41598-017-11220-1] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 08/16/2017] [Indexed: 01/23/2023] Open
Abstract
Drug delivery to the brain is hampered by the presence of the blood-brain barrier, which excludes most molecules from freely diffusing into the brain, and tightly regulates the active transport mechanisms that ensure sufficient delivery of nutrients to the brain parenchyma. Harnessing the possibility of delivering neuroactive drugs by way of receptors already present on the brain endothelium has been of interest for many years. The transferrin receptor is of special interest since its expression is limited to the endothelium of the brain as opposed to peripheral endothelium. Here, we investigate the possibility of delivering immunoliposomes and their encapsulated cargo to the brain via targeting of the transferrin receptor. We find that transferrin receptor-targeting increases the association between the immunoliposomes and primary endothelial cells in vitro, but that this does not correlate with increased cargo transcytosis. Furthermore, we show that the transferrin receptor-targeted immunoliposomes accumulate along the microvessels of the brains of rats, but find no evidence for transcytosis of the immunoliposome. Conversely, the increased accumulation correlated both with increased cargo uptake in the brain endothelium and subsequent cargo transport into the brain. These findings suggest that transferrin receptor-targeting is a relevant strategy of increasing drug exposure to the brain.
Collapse
Affiliation(s)
- Kasper Bendix Johnsen
- Laboratory for Neurobiology, Biomedicine, Institute of Health Science and Technology, Aalborg University, Aalborg, Denmark
- Center for Nanomedicine and Theranostics, Department of Micro- and Nanotechnology, Technical University of Denmark, Lyngby, Denmark
| | - Annette Burkhart
- Laboratory for Neurobiology, Biomedicine, Institute of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Fredrik Melander
- Center for Nanomedicine and Theranostics, Department of Micro- and Nanotechnology, Technical University of Denmark, Lyngby, Denmark
| | - Paul Joseph Kempen
- Center for Nanomedicine and Theranostics, Department of Micro- and Nanotechnology, Technical University of Denmark, Lyngby, Denmark
| | - Jonas Bruun Vejlebo
- Center for Nanomedicine and Theranostics, Department of Micro- and Nanotechnology, Technical University of Denmark, Lyngby, Denmark
| | - Piotr Siupka
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Thomas Lars Andresen
- Center for Nanomedicine and Theranostics, Department of Micro- and Nanotechnology, Technical University of Denmark, Lyngby, Denmark
| | - Torben Moos
- Laboratory for Neurobiology, Biomedicine, Institute of Health Science and Technology, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
34
|
Zhao Y, Zhang Y, Qin G, Cheng J, Zeng W, Liu S, Kong H, Wang X, Wang Q, Qu H. In vivo biodistribution and behavior of CdTe/ZnS quantum dots. Int J Nanomedicine 2017; 12:1927-1939. [PMID: 28331316 PMCID: PMC5352250 DOI: 10.2147/ijn.s121075] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The unique features of quantum dots (QDs) make them desirable fluorescent tags for cell and developmental biology applications that require long-term, multitarget, and highly sensitive imaging. In this work, we imaged fluorescent cadmium telluride/zinc sulfide (CdTe/ZnS) QDs in organs, tissues, and cells, and analyzed the mechanism of their lymphatic uptake and cellular distribution. We observed that the fluorescent CdTe/ZnS QDs were internalized by lymph nodes in four cell lines from different tissue sources. We obtained the fluorescence intensity–QD concentrations curve by quantitative analysis. Our results demonstrate that cells containing QDs can complete mitosis normally and that distribution of QDs was uniform across cell types and involved the vesicular transport system, including the endoplasmic reticulum. This capacity for CdTe/ZnS QD targeting provides insights into the applicability and limitations of fluorescent QDs for imaging biological specimens.
Collapse
Affiliation(s)
- Yan Zhao
- School of Basic Medical Sciences
| | | | | | | | | | | | - Hui Kong
- School of Basic Medical Sciences
| | | | | | - Huihua Qu
- Center of Scientific Experiment, Beijing University of Chinese Medicine, Beijing, People's Republic of China
| |
Collapse
|
35
|
Abstract
CNS disorders are on the rise despite advancements in our understanding of their pathophysiological mechanisms. A major hurdle to the treatment of these disorders is the blood-brain barrier (BBB), which serves as an arduous janitor to protect the brain. Many drugs are being discovered for CNS disorders, which, however fail to enter the market because of their inability to cross the BBB. This is a pronounced challenge for the pharmaceutical fraternity. Hence, in addition to the discovery of novel entities and drug candidates, scientists are also developing new formulations of existing drugs for brain targeting. Several approaches have been investigated to allow therapeutics to cross the BBB. As the molecular structure of the BBB is better elucidated, several key approaches for brain targeting include physiological transport mechanisms such as adsorptive-mediated transcytosis, inhibition of active efflux pumps, receptor-mediated transport, cell-mediated endocytosis, and the use of peptide vectors. Drug-delivery approaches comprise delivery from microspheres, biodegradable wafers, and colloidal drug-carrier systems (e.g., liposomes, nanoparticles, nanogels, dendrimers, micelles, nanoemulsions, polymersomes, exosomes, and quantum dots). The current review discusses the latest advancements in these approaches, with a major focus on articles published in 2015 and 2016. In addition, we also cover the alternative delivery routes, such as intranasal and convection-enhanced diffusion methods, and disruption of the BBB for brain targeting.
Collapse
Affiliation(s)
- Mayur M Patel
- Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad, Gujarat, 382481, India.
| | - Bhoomika M Patel
- Institute of Pharmacy, Nirma University, SG Highway, Chharodi, Ahmedabad, Gujarat, 382481, India
| |
Collapse
|
36
|
Bouchoucha M, Béliveau É, Kleitz F, Calon F, Fortin MA. Antibody-conjugated mesoporous silica nanoparticles for brain microvessel endothelial cell targeting. J Mater Chem B 2017; 5:7721-7735. [DOI: 10.1039/c7tb01385j] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Size and bioconjugation effects of Ri7 antibody conjugated mesoporous silica nanoparticles (MSNs) influence in vitro and in vivo targeting ability to BMECs.
Collapse
Affiliation(s)
- Meryem Bouchoucha
- Department of Chemistry
- Université Laval
- Québec (QC)
- Canada
- Department of M.M.Materials Engineering
| | - Éric Béliveau
- Faculty of Pharmacy
- Université Laval
- Québec (QC)
- Canada
- Neurosciences Axis
| | - Freddy Kleitz
- Department of Chemistry
- Université Laval
- Québec (QC)
- Canada
- Centre de Recherche sur les Matériaux
| | - Frédéric Calon
- Faculty of Pharmacy
- Université Laval
- Québec (QC)
- Canada
- Neurosciences Axis
| | - Marc-André Fortin
- Department of M.M.Materials Engineering
- Université Laval
- Québec (QC)
- Canada
- Centre de Recherche du Centre Hospitalier Universitaire de Québec (CR-CHUQ)
| |
Collapse
|
37
|
Curtis C, Zhang M, Liao R, Wood T, Nance E. Systems-level thinking for nanoparticle-mediated therapeutic delivery to neurological diseases. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [PMID: 27562224 DOI: 10.1002/wnan.1422] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 06/27/2016] [Accepted: 07/17/2016] [Indexed: 12/27/2022]
Abstract
Neurological diseases account for 13% of the global burden of disease. As a result, treating these diseases costs $750 billion a year. Nanotechnology, which consists of small (~1-100 nm) but highly tailorable platforms, can provide significant opportunities for improving therapeutic delivery to the brain. Nanoparticles can increase drug solubility, overcome the blood-brain and brain penetration barriers, and provide timed release of a drug at a site of interest. Many researchers have successfully used nanotechnology to overcome individual barriers to therapeutic delivery to the brain, yet no platform has translated into a standard of care for any neurological disease. The challenge in translating nanotechnology platforms into clinical use for patients with neurological disease necessitates a new approach to: (1) collect information from the fields associated with understanding and treating brain diseases and (2) apply that information using scalable technologies in a clinically-relevant way. This approach requires systems-level thinking to integrate an understanding of biological barriers to therapeutic intervention in the brain with the engineering of nanoparticle material properties to overcome those barriers. To demonstrate how a systems perspective can tackle the challenge of treating neurological diseases using nanotechnology, this review will first present physiological barriers to drug delivery in the brain and common neurological disease hallmarks that influence these barriers. We will then analyze the design of nanotechnology platforms in preclinical in vivo efficacy studies for treatment of neurological disease, and map concepts for the interaction of nanoparticle physicochemical properties and pathophysiological hallmarks in the brain. WIREs Nanomed Nanobiotechnol 2017, 9:e1422. doi: 10.1002/wnan.1422 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Chad Curtis
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Mengying Zhang
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
| | - Rick Liao
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Thomas Wood
- Department of Physiology, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA.,Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA.,Department of Radiology, University of Washington, Seattle, WA, USA.,Center on Human Development and Disability, University of Washington, Seattle, WA, USA
| |
Collapse
|
38
|
Ricard C, Lamasse L, Jaouen A, Rougon G, Debarbieux F. Combination of an optical parametric oscillator and quantum-dots 655 to improve imaging depth of vasculature by intravital multicolor two-photon microscopy. BIOMEDICAL OPTICS EXPRESS 2016; 7:2362-2372. [PMID: 27375951 PMCID: PMC4918589 DOI: 10.1364/boe.7.002362] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 05/04/2016] [Accepted: 05/18/2016] [Indexed: 06/06/2023]
Abstract
Simultaneous imaging of different cell types and structures in the mouse central nervous system (CNS) by intravital two-photon microscopy requires the characterization of fluorophores and advances in approaches to visualize them. We describe the use of a two-photon infrared illumination generated by an optical parametric oscillator (OPO) on quantum-dots 655 (QD655) nanocrystals to improve resolution of the vasculature deeper in the mouse brain both in healthy and pathological conditions. Moreover, QD655 signal can be unmixed from the DsRed2, CFP, EGFP and EYFP fluorescent proteins, which enhances the panel of multi-parametric correlative investigations both in the cortex and the spinal cord.
Collapse
Affiliation(s)
- Clément Ricard
- Institut des Neurosciences de la Timone, Marseille, Aix-Marseille Université and CNRS UMR7289, France
- Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France
- Service de Pharmacie, Assistance Publique, Hôpitaux de Marseille, Marseille, France
- Centre de Recherche en Oncobiologie et Oncopharmacologie, INSERM UMR911 and Aix-Marseille Université, Marseille, France
- These authors contributed equally to this work
| | - Lisa Lamasse
- Institut des Neurosciences de la Timone, Marseille, Aix-Marseille Université and CNRS UMR7289, France
- Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France
- These authors contributed equally to this work
| | - Alexandre Jaouen
- Institut des Neurosciences de la Timone, Marseille, Aix-Marseille Université and CNRS UMR7289, France
- Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France
| | - Geneviève Rougon
- Institut des Neurosciences de la Timone, Marseille, Aix-Marseille Université and CNRS UMR7289, France
- Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France
| | - Franck Debarbieux
- Institut des Neurosciences de la Timone, Marseille, Aix-Marseille Université and CNRS UMR7289, France
- Centre Européen de Recherche en Imagerie Médicale, Aix-Marseille Université, Marseille, France
| |
Collapse
|