1
|
Jin Y, Du Q, Song M, Kang R, Zhou J, Zhang H, Ding Y. Amyloid-β-targeting immunotherapies for Alzheimer's disease. J Control Release 2024; 375:346-365. [PMID: 39271059 DOI: 10.1016/j.jconrel.2024.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/24/2024] [Accepted: 09/08/2024] [Indexed: 09/15/2024]
Abstract
Recent advances in clinical passive immunotherapy have provided compelling evidence that eliminating amyloid-β (Aβ) slows cognitive decline in Alzheimer's disease (AD). However, the modest benefits and side effects observed in clinical trials indicate that current immunotherapy therapy is not a panacea, highlighting the need for a deeper understanding of AD mechanisms and the significance of early intervention through optimized immunotherapy or immunoprevention. This review focuses on the centrality of Aβ pathology in AD and summarizes recent clinical progress in passive and active immunotherapies targeting Aβ, discussing their lessons and failures to inform future anti-Aβ biotherapeutics design. Various delivery strategies to optimize Aβ-targeting immunotherapies are outlined, highlighting their benefits and drawbacks in overcoming challenges such as poor stability and limited tissue accessibility of anti-Aβ biotherapeutics. Additionally, the perspectives and challenges of immunotherapy and immunoprevention targeting Aβ are concluded in the end, aiming to guide the development of next-generation anti-Aβ immunotherapeutic agents towards improved efficacy and safety.
Collapse
Affiliation(s)
- Yi Jin
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Qiaofei Du
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Mingjie Song
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Ruixin Kang
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Jianping Zhou
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Huaqing Zhang
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| | - Yang Ding
- Department of Pharmaceutics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
2
|
Aminyavari S, Afshari AR, Ahmadi SS, Kesharwani P, Sanati M, Sahebkar A. Unveiling the theranostic potential of SPIONs in Alzheimer's disease management. J Psychiatr Res 2024; 179:244-256. [PMID: 39321523 DOI: 10.1016/j.jpsychires.2024.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 08/22/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
Alzheimer's disease (AD) is a devastating kind of dementia that is becoming more common worldwide. Toxic amyloid-beta (Aβ) aggregates are the primary cause of AD onset and development. Superparamagnetic iron oxide nanoparticles (SPIONs) have received a lot of interest in AD therapy over the last decade because of their ability to redirect the Aβ fibrillation process and improve associated brain dysfunction. The potential diagnostic application of SPIONs in AD has dramatically increased this interest. Furthermore, surface-modified engineered SPIONs function as drug carriers to improve the efficacy of current therapies. Various preclinical and clinical studies on the role of SPIONs in AD pathology have produced encouraging results. However, due to their physicochemical properties (e.g., size, surface charge, and particle concentration) in the biological milieu, SPIONs may play the role of a preventive or accelerative agent in AD. Even though SPIONs are potential therapeutic and diagnostic options in AD, significant efforts are still needed to overcome the inconsistencies and safety concerns. This review evaluated the current understanding of how various SPIONs interact with AD models and explored the discrepancies in their efficacy and safety.
Collapse
Affiliation(s)
- Samaneh Aminyavari
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir R Afshari
- Department of Basic Sciences, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Seyed Sajad Ahmadi
- Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran; Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Lei T, Yang Z, Li H, Qin M, Gao H. Interactions between nanoparticles and pathological changes of vascular in Alzheimer's disease. Adv Drug Deliv Rev 2024; 207:115219. [PMID: 38401847 DOI: 10.1016/j.addr.2024.115219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 02/26/2024]
Abstract
Emerging evidence suggests that vascular pathological changes play a pivotal role in the pathogenesis of Alzheimer's disease (AD). The dysfunction of the cerebral vasculature occurs in the early course of AD, characterized by alterations in vascular morphology, diminished cerebral blood flow (CBF), impairment of the neurovascular unit (NVU), vasculature inflammation, and cerebral amyloid angiopathy. Vascular dysfunction not only facilitates the influx of neurotoxic substances into the brain, triggering inflammation and immune responses but also hampers the efflux of toxic proteins such as Aβ from the brain, thereby contributing to neurodegenerative changes in AD. Furthermore, these vascular changes significantly impact drug delivery and distribution within the brain. Therefore, developing targeted delivery systems or therapeutic strategies based on vascular alterations may potentially represent a novel breakthrough in AD treatment. This review comprehensively examines various aspects of vascular alterations in AD and outlines the current interactions between nanoparticles and pathological changes of vascular.
Collapse
Affiliation(s)
- Ting Lei
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zixiao Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hanmei Li
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Meng Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Mental Health Center and National Chengdu Center for Safety Evaluation of Drugs, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
4
|
Panghal A, Flora SJS. Nanotechnology in the diagnostic and therapy for Alzheimer's disease. Biochim Biophys Acta Gen Subj 2024; 1868:130559. [PMID: 38191034 DOI: 10.1016/j.bbagen.2024.130559] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/10/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder primarily characterized by β-amyloid plaque, intraneuronal tangles, significant neuronal loss and cognitive deficit. Treatment in the early stages of the disease is crucial for preventing or perhaps reversing the neurodegeneration in the AD cases. However, none of the current diagnostic procedures are capable of early diagnosis of AD. Further, the available treatments merely provide symptomatic alleviation in AD and do not address the underlying illness. Therefore, there is no permanent cure for AD currently. Better therapeutic outcomes need the optimum drug concentration in the central nervous system (CNS) by traversing blood-brain-barrier (BBB). Nanotechnology offers enormous promise to transform the treatment and diagnostics of neurodegenerative diseases. Nanotechnology based diagnostic tools, drug delivery systems and theragnostic are capable of highly sensitive molecular detection, effective drug targeting and their combination. Significant work has been done in this area over the last decade and prospective results have been obtained in AD therapy. This review explores the various applications of nanotechnology in addressing the varied facets of AD, ranging from early detection to therapeutic interventions. This review also looks at how nanotechnology can help with the development of disease-modifying medicines, such as the delivery of anti-amyloid, anti-tau, cholinesterase inhibitors, antioxidants and hormonal drugs. In conclusion, this paper discusses the role of nanotechnology in the early detection of AD, effective drug targeting to the CNS and theragnostic applications in the management of AD.
Collapse
Affiliation(s)
- Archna Panghal
- National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, Panjab 160012, India
| | - S J S Flora
- National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, Panjab 160012, India; Institute of Pharmaceutical Sciences, Era Medical University, Safarajganj, Lucknow 226003, U.P., India.
| |
Collapse
|
5
|
Stamenkovic S, Li Y, Waters J, Shih A. Deep Imaging to Dissect Microvascular Contributions to White Matter Degeneration in Rodent Models of Dementia. Stroke 2023; 54:1403-1415. [PMID: 37094035 PMCID: PMC10460612 DOI: 10.1161/strokeaha.122.037156] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
The increasing socio-economic burden of Alzheimer disease (AD) and AD-related dementias has created a pressing need to define targets for therapeutic intervention. Deficits in cerebral blood flow and neurovascular function have emerged as early contributors to disease progression. However, the cause, progression, and consequence of small vessel disease in AD/AD-related dementias remains poorly understood, making therapeutic targets difficult to pinpoint. Animal models that recapitulate features of AD/AD-related dementias may provide mechanistic insight because microvascular pathology can be studied as it develops in vivo. Recent advances in in vivo optical and ultrasound-based imaging of the rodent brain facilitate this goal by providing access to deeper brain structures, including white matter and hippocampus, which are more vulnerable to injury during cerebrovascular disease. Here, we highlight these novel imaging approaches and discuss their potential for improving our understanding of vascular contributions to AD/AD-related dementias.
Collapse
Affiliation(s)
- Stefan Stamenkovic
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Yuandong Li
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Andy Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
6
|
Ibrahim Fouad G, Mabrouk M, El-Sayed SAM, Rizk MZ, Beherei HH. Neurotherapeutic efficacy of loaded sulforaphane on iron oxide nanoparticles against cuprizone-induced neurotoxicity: role of MMP-9 and S100β. Toxicol Mech Methods 2023:1-17. [PMID: 36775846 DOI: 10.1080/15376516.2023.2177219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
Abstract
Cuprizone (CUP) induces neurotoxicity and demyelination in animal models by provoking the activation of glial cells and the generation of reactive oxygen species (ROS). Sulforaphane (SF) is a phytochemical that exhibits a neuroprotective potential. In this study, we investigated the neurotherapeutic and pro-remyelinating activities of SF and SF-loaded within iron oxide nanoparticles (IONP-SF) in CUP-exposed rats. Magnetite iron oxide nanoparticles (IONPs) were prepared using the hydrothermal method that was further loaded with SF (IONP-SF). The loading of SF within the magnetite nanoparticles was assessed using FTIR, TEM, DLS, Zetasizer, and XPS. For the in vivo investigations, adult male Wistar rats (n = 40) were administrated either on a regular diet or a diet with CUP (0.2%) for 5 weeks. The rats were divided into four groups: negative control, CUP-induced, CUP + SF, and CUP + IONP-SF. CUP-exposed brains exhibited a marked elevation in lipid peroxidation, along with a significant decrease in the activities of glutathione peroxidase (GPx), and catalase (CAT). In addition, CUP intoxication downregulated the expression of myelin basic protein (MBP) and myelin proteolipid protein (PLP), upregulated the expression of Matrix metallopeptidase-9 (MMP-9) and S100β, and increased caspase-3 immunoexpression, these results were supported histopathologically in the cerebral cortexes. Treatment of CUP-rats with either SF or IONP-SF demonstrated remyelinating and neurotherapeutic activities. We could conclude that IONP-SF was more effective than free SF in mitigating the CUP-induced downregulation of MBP, upregulation of S100β, and caspase-3 immunoexpression.
Collapse
Affiliation(s)
- Ghadha Ibrahim Fouad
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Cairo, Egypt
| | - Mostafa Mabrouk
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, Cairo, Egypt
| | - Sara A M El-Sayed
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, Cairo, Egypt
| | - Maha Z Rizk
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Cairo, Egypt
| | - Hanan H Beherei
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, Cairo, Egypt
| |
Collapse
|
7
|
Taliyan R, Kakoty V, Sarathlal KC, Kharavtekar SS, Karennanavar CR, Choudhary YK, Singhvi G, Riadi Y, Dubey SK, Kesharwani P. Nanocarrier mediated drug delivery as an impeccable therapeutic approach against Alzheimer's disease. J Control Release 2022; 343:528-550. [PMID: 35114208 DOI: 10.1016/j.jconrel.2022.01.044] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/22/2022] [Accepted: 01/25/2022] [Indexed: 12/14/2022]
Abstract
For the past several years, dementia, is one of the predominantly observed groups of symptoms in a geriatric population. Alzheimer's disease (AD) is a progressive memory related neurodegenerative disease, for which the current Food and drug administration approved therapeutics are only meant for a symptomatic management rather than targeting the root cause of AD. These therapeutics belong to two classes, Acetylcholine Esterase inhibitors and N-methyl D-aspartate antagonist. Furthermore, to facilitate neuroprotective action in AD, the drugs are majorly expected to reach the specific target area in the brain for the desired efficacy. Thus, there is a huge requirement for drug discovery and development for facilitating the entry of drugs more in brain to exert a specific action. The very first line of defense and the major limitation for the entry of drugs into the brain is the Blood Brain Barrier, followed by Blood-Cerebrospinal Fluid Barrier. More than a barrier, these mainly act as selectively permeable membranes, which allows entry of specific molecules into the brain. Furthermore, specific enzymes result in the degradation of xenobiotics. All these mechanisms pose as hurdles in the way of effective drug delivery in the brain. Thus, novel techniques need to be harbored for the facilitation of the delivery of such drugs into the brain. Nanocarriers are advantageous for facilitating the specific targeted drug treatment in AD. As nanomedicines are one of the novels and most useful approaches for AD, thus the present review mainly focuses on understanding the advanced use of nanocarriers for targeted drug delivery in the management of AD.
Collapse
Affiliation(s)
- Rajeev Taliyan
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, 333031, Rajasthan, India.
| | - Violina Kakoty
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, 333031, Rajasthan, India
| | - K C Sarathlal
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, 333031, Rajasthan, India
| | - Sanskruti Santosh Kharavtekar
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, 333031, Rajasthan, India
| | - Chandrashekar R Karennanavar
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, 333031, Rajasthan, India
| | | | - Gautam Singhvi
- Neuropsychopharmacology Division, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani Campus, 333031, Rajasthan, India
| | - Yassine Riadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Sunil Kumar Dubey
- Medical Research, R&D Healthcare Division, Emami Ltd, 13, BT Road, Belgharia, Kolkata 700056, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
8
|
Ni R. Magnetic Resonance Imaging in Animal Models of Alzheimer's Disease Amyloidosis. Int J Mol Sci 2021; 22:12768. [PMID: 34884573 PMCID: PMC8657987 DOI: 10.3390/ijms222312768] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Amyloid-beta (Aβ) plays an important role in the pathogenesis of Alzheimer's disease. Aberrant Aβ accumulation induces neuroinflammation, cerebrovascular alterations, and synaptic deficits, leading to cognitive impairment. Animal models recapitulating the Aβ pathology, such as transgenic, knock-in mouse and rat models, have facilitated the understanding of disease mechanisms and the development of therapeutics targeting Aβ. There is a rapid advance in high-field MRI in small animals. Versatile high-field magnetic resonance imaging (MRI) sequences, such as diffusion tensor imaging, arterial spin labeling, resting-state functional MRI, anatomical MRI, and MR spectroscopy, as well as contrast agents, have been developed for preclinical imaging in animal models. These tools have enabled high-resolution in vivo structural, functional, and molecular readouts with a whole-brain field of view. MRI has been used to visualize non-invasively the Aβ deposits, synaptic deficits, regional brain atrophy, impairment in white matter integrity, functional connectivity, and cerebrovascular and glymphatic system in animal models of Alzheimer's disease amyloidosis. Many of the readouts are translational toward clinical MRI applications in patients with Alzheimer's disease. In this review, we summarize the recent advances in MRI for visualizing the pathophysiology in amyloidosis animal models. We discuss the outstanding challenges in brain imaging using MRI in small animals and propose future outlook in visualizing Aβ-related alterations in the brains of animal models.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH Zurich & University of Zurich, 8093 Zurich, Switzerland;
- Institute for Regenerative Medicine, University of Zurich, 8952 Zurich, Switzerland
| |
Collapse
|
9
|
Niu F, Sharma A, Wang Z, Feng L, Muresanu DF, Sahib S, Tian ZR, Lafuente JV, Buzoianu AD, Castellani RJ, Nozari A, Menon PK, Patnaik R, Wiklund L, Sharma HS. Nanodelivery of oxiracetam enhances memory, functional recovery and induces neuroprotection following concussive head injury. PROGRESS IN BRAIN RESEARCH 2021; 265:139-230. [PMID: 34560921 DOI: 10.1016/bs.pbr.2021.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Military personnel are the most susceptible to concussive head injury (CHI) caused by explosion, blast or missile or blunt head trauma. Mild to moderate CHI could induce lifetime functional and cognitive disturbances causing significant decrease in quality of life. Severe CHI leads to instant death and lifetime paralysis. Thus, further exploration of novel therapeutic agents or new features of known pharmacological agents are needed to enhance quality of life of CHI victims. Previous reports from our laboratory showed that mild CHI induced by weight drop technique causing an impact of 0.224N results in profound progressive functional deficit, memory impairment and brain pathology from 5h after trauma that continued over several weeks of injury. In this investigation we report that TiO2 nanowired delivery of oxiracetam (50mg/kg, i.p.) daily for 5 days after CHI resulted in significant improvement of functional deficit on the 8th day. This was observed using Rota Rod treadmill, memory improvement assessed by the time spent in finding hidden platform under water. The motor function improvement is seen in oxiracetam treated CHI group by placing forepaw on an inclined mesh walking and foot print analysis for stride length and distance between hind feet. TiO2-nanowired oxiracetam also induced marked improvements in the cerebral blood flow, reduction in the BBB breakdown and edema formation as well as neuroprotection of neuronal, glial and myelin damages caused by CHI at light and electron microscopy on the 7th day after 5 days TiO2 oxiracetam treatment. Adverse biochemical events such as upregulation of CSF nitrite and nitrate, IL-6, TNF-a and p-Tau are also reduced significantly in oxiracetam treated CHI group. On the other hand post treatment of 100mg/kg dose of normal oxiracetam in identical conditions after CHI is needed to show slight but significant neuroprotection together with mild recovery of memory function and functional deficits on the 8th day. These observations are the first to point out that nanowired delivery of oxiracetam has superior neuroprotective ability in CHI. These results indicate a promising clinical future of TiO2 oxiracetam in treating CHI patients for better quality of life and neurorehabilitation, not reported earlier.
Collapse
Affiliation(s)
- Feng Niu
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, China
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Zhenguo Wang
- CSPC NBP Pharmaceutical Medicine, Shijiazhuang, China
| | - Lianyuan Feng
- Department of Neurology, Bethune International Peace Hospital, Shijiazhuang, China
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - Preeti K Menon
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
10
|
Anjum S, Ishaque S, Fatima H, Farooq W, Hano C, Abbasi BH, Anjum I. Emerging Applications of Nanotechnology in Healthcare Systems: Grand Challenges and Perspectives. Pharmaceuticals (Basel) 2021; 14:ph14080707. [PMID: 34451803 PMCID: PMC8401281 DOI: 10.3390/ph14080707] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/15/2021] [Accepted: 07/17/2021] [Indexed: 02/07/2023] Open
Abstract
Healthcare, as a basic human right, has often become the focus of the development of innovative technologies. Technological progress has significantly contributed to the provision of high-quality, on-time, acceptable, and affordable healthcare. Advancements in nanoscience have led to the emergence of a new generation of nanostructures. Each of them has a unique set of properties that account for their astonishing applications. Since its inception, nanotechnology has continuously affected healthcare and has exerted a tremendous influence on its transformation, contributing to better outcomes. In the last two decades, the world has seen nanotechnology taking steps towards its omnipresence and the process has been accelerated by extensive research in various healthcare sectors. The inclusion of nanotechnology and its allied nanocarriers/nanosystems in medicine is known as nanomedicine, a field that has brought about numerous benefits in disease prevention, diagnosis, and treatment. Various nanosystems have been found to be better candidates for theranostic purposes, in contrast to conventional ones. This review paper will shed light on medically significant nanosystems, as well as their applications and limitations in areas such as gene therapy, targeted drug delivery, and in the treatment of cancer and various genetic diseases. Although nanotechnology holds immense potential, it is yet to be exploited. More efforts need to be directed to overcome these limitations and make full use of its potential in order to revolutionize the healthcare sector in near future.
Collapse
Affiliation(s)
- Sumaira Anjum
- Department of Biotechnology, Kinnaird College for Women, Lahore 54000, Pakistan; (S.I.); (H.F.); (W.F.); (I.A.)
- Correspondence: ; Tel.: +92-300-6957038
| | - Sara Ishaque
- Department of Biotechnology, Kinnaird College for Women, Lahore 54000, Pakistan; (S.I.); (H.F.); (W.F.); (I.A.)
| | - Hijab Fatima
- Department of Biotechnology, Kinnaird College for Women, Lahore 54000, Pakistan; (S.I.); (H.F.); (W.F.); (I.A.)
| | - Wajiha Farooq
- Department of Biotechnology, Kinnaird College for Women, Lahore 54000, Pakistan; (S.I.); (H.F.); (W.F.); (I.A.)
| | - Christophe Hano
- Laboratoire de Biologie des Ligneux et des Grandes Cultures (LBLGC), INRAe USC1328, Université d’Orléans, 28000 Chartres, France;
| | - Bilal Haider Abbasi
- Department of Biotechnology, Quaid-i-Azam University, Islamabad 54000, Pakistan;
| | - Iram Anjum
- Department of Biotechnology, Kinnaird College for Women, Lahore 54000, Pakistan; (S.I.); (H.F.); (W.F.); (I.A.)
| |
Collapse
|
11
|
Noor N, Gani A, Gani A, Shah A, Ashraf ZU. Exploitation of polyphenols and proteins using nanoencapsulation for anti-viral and brain boosting properties - Evoking a synergistic strategy to combat COVID-19 pandemic. Int J Biol Macromol 2021; 180:375-384. [PMID: 33716131 PMCID: PMC7946821 DOI: 10.1016/j.ijbiomac.2021.03.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/31/2021] [Accepted: 03/04/2021] [Indexed: 12/16/2022]
Abstract
The world is currently under the threat of COVID pandemic and has focused every dimension of research in finding a cure to this novel disease. In this current situation, people are facing mental stress, agony, fear, depression and other associated symptoms which are taking a toll on their overall mental health. Nanoencapsulation of certain brain boosting polyphenols including quercetin, caffeine, cocoa flavanols and proteins like lectins can become new area of interest in the present scenario. Besides the brain boosting benefits, we have also highlighted the anti- viral activities of these compounds which we assume can play a possible role in combating COVID-19 given to their previous history of action against certain viruses. This review outlines the nanoencapsulation approaches of such synergistic compounds as a novel strategy to take the ongoing research a step ahead and also provides a new insight in bringing the role of nanotechnology in addressing the issues related to COVID pandemic.
Collapse
Affiliation(s)
- Nairah Noor
- Laboratory of Functional Food and Nutraceuticals, Department of Food Science and Technology, University of Kashmir, Srinagar 190006, India
| | - Adil Gani
- Laboratory of Functional Food and Nutraceuticals, Department of Food Science and Technology, University of Kashmir, Srinagar 190006, India; Department of Food Science, Rutgers University, 65 Dudley Road, New Brunswick, NJ 08901, United States.
| | - Asir Gani
- Laboratory of Functional Food and Nutraceuticals, Department of Food Science and Technology, University of Kashmir, Srinagar 190006, India
| | - Asima Shah
- Laboratory of Functional Food and Nutraceuticals, Department of Food Science and Technology, University of Kashmir, Srinagar 190006, India
| | - Zanoor Ul Ashraf
- Laboratory of Functional Food and Nutraceuticals, Department of Food Science and Technology, University of Kashmir, Srinagar 190006, India
| |
Collapse
|
12
|
Nguyen TT, Vo TK, Vo GV. Therapeutic Strategies and Nano-Drug Delivery Applications in Management of Aging Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1286:183-198. [PMID: 33725354 DOI: 10.1007/978-3-030-55035-6_13] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder in which the death of brain cells causes memory loss and cognitive decline. Existing drugs only suppress symptoms or delay further deterioration but do not address the cause of the disease. In spite of screening numerous drug candidates against various molecular targets of AD, only a few candidates, such as acetylcholinesterase inhibitors, are currently utilized as an effective clinical therapy. Currently, nano-based therapies can make a difference, providing new therapeutic options by helping drugs to cross the blood-brain barrier and enter the brain more effectively. The main aim of this review was to highlight advances in research on the development of nano-based therapeutics for improved treatment of AD.
Collapse
Affiliation(s)
- Thuy Trang Nguyen
- Faculty of Pharmacy, Ho Chi Minh City University of Technology (HUTECH), Ho Chi Minh City, Vietnam
| | - Tuong Kha Vo
- Vietnam Sports Hospital, Ministry of Culture, Sports and Tourism, Hanoi, Vietnam
| | - Giau Van Vo
- Department of Industrial and Environmental Engineering, Gachon University, Seongnam-si, South Korea. .,Department of Bionano Technology, Gachon University, Seongnam-si, South Korea. .,School of Medicine, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, Vietnam.
| |
Collapse
|
13
|
Zorkina Y, Abramova O, Ushakova V, Morozova A, Zubkov E, Valikhov M, Melnikov P, Majouga A, Chekhonin V. Nano Carrier Drug Delivery Systems for the Treatment of Neuropsychiatric Disorders: Advantages and Limitations. Molecules 2020; 25:E5294. [PMID: 33202839 PMCID: PMC7697162 DOI: 10.3390/molecules25225294] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022] Open
Abstract
Neuropsychiatric diseases are one of the main causes of disability, affecting millions of people. Various drugs are used for its treatment, although no effective therapy has been found yet. The blood brain barrier (BBB) significantly complicates drugs delivery to the target cells in the brain tissues. One of the problem-solving methods is the usage of nanocontainer systems. In this review we summarized the data about nanoparticles drug delivery systems and their application for the treatment of neuropsychiatric disorders. Firstly, we described and characterized types of nanocarriers: inorganic nanoparticles, polymeric and lipid nanocarriers, their advantages and disadvantages. We discussed ways to interact with nerve tissue and methods of BBB penetration. We provided a summary of nanotechnology-based pharmacotherapy of schizophrenia, bipolar disorder, depression, anxiety disorder and Alzheimer's disease, where development of nanocontainer drugs derives the most active. We described various experimental drugs for the treatment of Alzheimer's disease that include vector nanocontainers targeted on β-amyloid or tau-protein. Integrally, nanoparticles can substantially improve the drug delivery as its implication can increase BBB permeability, the pharmacodynamics and bioavailability of applied drugs. Thus, nanotechnology is anticipated to overcome the limitations of existing pharmacotherapy of psychiatric disorders and to effectively combine various treatment modalities in that direction.
Collapse
Affiliation(s)
- Yana Zorkina
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.A.); (V.U.); (A.M.); (E.Z.); (M.V.); (P.M.); (V.C.)
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Olga Abramova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.A.); (V.U.); (A.M.); (E.Z.); (M.V.); (P.M.); (V.C.)
| | - Valeriya Ushakova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.A.); (V.U.); (A.M.); (E.Z.); (M.V.); (P.M.); (V.C.)
- Department of Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Anna Morozova
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.A.); (V.U.); (A.M.); (E.Z.); (M.V.); (P.M.); (V.C.)
- Healthcare Department, Mental-Health Clinic No. 1 Named after N.A. Alexeev of Moscow, 117152 Moscow, Russia
| | - Eugene Zubkov
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.A.); (V.U.); (A.M.); (E.Z.); (M.V.); (P.M.); (V.C.)
| | - Marat Valikhov
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.A.); (V.U.); (A.M.); (E.Z.); (M.V.); (P.M.); (V.C.)
| | - Pavel Melnikov
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.A.); (V.U.); (A.M.); (E.Z.); (M.V.); (P.M.); (V.C.)
| | - Alexander Majouga
- D. Mendeleev University of Chemical Technology of Russia, 125047 Moscow, Russia;
| | - Vladimir Chekhonin
- Department Basic and Applied Neurobiology, V.P. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.A.); (V.U.); (A.M.); (E.Z.); (M.V.); (P.M.); (V.C.)
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
14
|
Fayazi R, Habibi-Rezaei M, Heiat M, Javadi-Zarnaghi F, Taheri RA. Glycated albumin precipitation using aptamer conjugated magnetic nanoparticles. Sci Rep 2020; 10:10716. [PMID: 32612182 PMCID: PMC7329883 DOI: 10.1038/s41598-020-67469-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Accepted: 06/02/2020] [Indexed: 01/10/2023] Open
Abstract
To develop a strategy for the elimination of prefibrillar amyloid aggregates, a three-step non-modified DNA aptamer conjugation on silica-coated magnetic nanoparticles was carried out to achieve aptamer conjugated on MNP (Ap-SiMNP). Prefibrillar amyloid aggregates are generated under a diabetic condition which are prominently participated in developing diabetic complications. The binding properties of candidate DNA aptamer against serum albumin prefibrillar amyloid aggregates (AA20) were verified using electrophoretic mobility shift assay (EMSA) and surface plasmon resonance spectroscopy (SPR) analysis. The chloro-functionalized silica-coated MNPs were synthesized then a nano-targeting structure as aptamer conjugated on MNP (Ap-SiMNP) was constructed. Finally, Ap-SiMNP was verified for specific binding efficiency and AA20 removal using an external magnetic field. The candidate aptamer showed a high binding capacity at EMSA and SPR analysis (KD = 3.4 × 10─9 M) and successfully used to construct Ap-SiMNP. Here, we show a proof of concept for an efficient bio-scavenger as Ap-SiMNP to provide a promising opportunity to consider as a possible strategy to overcome some diabetic complications through specific binding/removal of toxic AA20 species.
Collapse
Affiliation(s)
- R Fayazi
- School of Biology, University of Tehran, P.O.Box 14155-6455, Tehran, Iran
| | - M Habibi-Rezaei
- School of Biology, University of Tehran, P.O.Box 14155-6455, Tehran, Iran.
- Center of Excellence in Nano-Biomedicine, University of Tehran, Tehran, Iran.
| | - M Heiat
- Research Center for Gastroenterology and Liver Disease, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - F Javadi-Zarnaghi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - R A Taheri
- Nanobiotechnolology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Rabanel JM, Delbreil P, Banquy X, Brambilla D, Ramassamy C. Periphery-confined particulate systems for the management of neurodegenerative diseases and toxicity: Avoiding the blood-brain-barrier challenge. J Control Release 2020; 322:286-299. [PMID: 32243978 DOI: 10.1016/j.jconrel.2020.03.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 01/07/2023]
Abstract
The blood-brain barrier prevents passage of large and hydrophilic molecules, undermining efforts to deliver most active molecules, proteins and other macromolecules. To date, nanoparticle-assisted delivery has been extensively studied to overcome this challenge but with limited success. On the other hand, for certain brain therapeutic applications, periphery-confined particles could be of immediate therapeutic usefulness. The modulation of CNS dysfunctions from the peripheral compartment is a promising approach, as it does not involve invasive interventions. From recent studies, three main roles could be identified for periphery-confined particles: brain tissue detoxification via the "sink-effect"; a "circulating drug-reservoir" effect to improve drug delivery to brain tissues, and finally, brain vascular endothelium targeting to diagnose or heal vascular-related dysfunctions. These applications are much easier to implement as they do not involve complex therapeutic and targeting strategies and do not require crossing biological barriers. Micro/nano-devices required for such applications will likely be simpler to synthesize and will involve fewer complex materials. Moreover, peripheral particles are expected to be less prone to neurotoxicity and issues related to their diffusion in confined space.
Collapse
Affiliation(s)
- Jean-Michel Rabanel
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| | - Philippe Delbreil
- Faculty of Pharmacy, Université de Montréal, CP. 6128, succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Xavier Banquy
- Faculty of Pharmacy, Université de Montréal, CP. 6128, succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Davide Brambilla
- Faculty of Pharmacy, Université de Montréal, CP. 6128, succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Charles Ramassamy
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 boul. des Prairies, Laval, QC H7V 1B7, Canada
| |
Collapse
|
16
|
Cano A, Sánchez-López E, Ettcheto M, López-Machado A, Espina M, Souto EB, Galindo R, Camins A, García ML, Turowski P. Current advances in the development of novel polymeric nanoparticles for the treatment of neurodegenerative diseases. Nanomedicine (Lond) 2020; 15:1239-1261. [PMID: 32370600 DOI: 10.2217/nnm-2019-0443] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Effective intervention is essential to combat the coming epidemic of neurodegenerative (ND) diseases. Nanomedicine can overcome restrictions of CNS delivery imposed by the blood-brain barrier, and thus be instrumental in preclinical discovery and therapeutic intervention of ND diseases. Polymeric nanoparticles (PNPs) have shown great potential and versatility to encapsulate several compounds simultaneously in controlled drug-delivery systems and target them to the deepest brain regions. Here, we critically review recent advances in the development of drugs incorporated into PNPs and summarize the molecular changes and functional effects achieved in preclinical models of the most common ND disorders. We also briefly discuss the many challenges remaining to translate these findings and technological advances successfully to current clinical settings.
Collapse
Affiliation(s)
- Amanda Cano
- Department of Pharmacy, Pharmaceutical Technology & Physical Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain.,Institute of Nanoscience & Nanotechnology (IN2UB), Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Elena Sánchez-López
- Department of Pharmacy, Pharmaceutical Technology & Physical Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain.,Institute of Nanoscience & Nanotechnology (IN2UB), Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Miren Ettcheto
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacology, Toxicology & Therapeutic Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain.,Unit of Biochemistry & Pharmacology, Faculty of Medicine & Health Sciences, University of Rovira i Virgili, Reus (Tarragona), Spain
| | - Ana López-Machado
- Department of Pharmacy, Pharmaceutical Technology & Physical Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain.,Institute of Nanoscience & Nanotechnology (IN2UB), Barcelona, Spain
| | - Marta Espina
- Department of Pharmacy, Pharmaceutical Technology & Physical Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain.,Institute of Nanoscience & Nanotechnology (IN2UB), Barcelona, Spain
| | - Eliana B Souto
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.,CEB, Centre of Biological Engineering, University of Minho, Campus de Gualtar 4710-057, Braga, Portugal
| | - Ruth Galindo
- Department of Pharmacy, Pharmaceutical Technology & Physical Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain.,Institute of Nanoscience & Nanotechnology (IN2UB), Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Unit of Synthesis & Biomedical Applications of Peptides, Department of Biomedical Chemistry, Institute for Advanced Chemistry of Catalonia, Consejo Superior de Investigaciones Científicas (IQAC-CSIC), Barcelona, Spain
| | - Antonio Camins
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Department of Pharmacology, Toxicology & Therapeutic Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain
| | - Maria Luisa García
- Department of Pharmacy, Pharmaceutical Technology & Physical Chemistry, Faculty of Pharmacy & Food Sciences, University of Barcelona, Barcelona, Spain.,Institute of Nanoscience & Nanotechnology (IN2UB), Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Patric Turowski
- UCL Institute of Ophthalmology, University College of London, London, UK
| |
Collapse
|
17
|
Ke PC, Pilkington EH, Sun Y, Javed I, Kakinen A, Peng G, Ding F, Davis TP. Mitigation of Amyloidosis with Nanomaterials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901690. [PMID: 31183916 PMCID: PMC6904546 DOI: 10.1002/adma.201901690] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/15/2019] [Indexed: 05/19/2023]
Abstract
Amyloidosis is a biophysical phenomenon of protein aggregation with biological and pathogenic implications. Among the various strategies developed to date, nanomaterials and multifunctional nanocomposites possessing certain structural and physicochemical traits are promising candidates for mitigating amyloidosis in vitro and in vivo. The mechanisms underpinning protein aggregation and toxicity are introduced, and opportunities in materials science to drive this interdisciplinary field forward are highlighted. Advancement of this emerging frontier hinges on exploitation of protein self-assembly and interactions of amyloid proteins with nanoparticles, intracellular and extracellular proteins, chaperones, membranes, organelles, and biometals.
Collapse
Affiliation(s)
- Pu Chun Ke
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Emily H. Pilkington
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Yunxiang Sun
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
- Department of Physics, Ningbo University, Ningbo 315211, China
| | - Ibrahim Javed
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Aleksandr Kakinen
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Guotao Peng
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Thomas P. Davis
- ARC Center of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Qld 4072, Australia
| |
Collapse
|
18
|
Ulanova M, Poljak A, Wen W, Bongers A, Gloag L, Gooding J, Tilley R, Sachdev P, Braidy N. Nanoparticles as contrast agents for the diagnosis of Alzheimer’s disease: a systematic review. Nanomedicine (Lond) 2020; 15:725-743. [DOI: 10.2217/nnm-2019-0316] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nanoparticle (NP)-based magnetic contrast agents have opened the potential for MRI to be used for early diagnosis of Alzheimer’s disease (AD). This article aims to review the current progress of research in this field. A comprehensive literature search was performed based on PubMed, Medline, EMBASE, PsychINFO and Scopus databases using the following terms: ‘Alzheimer’s disease’ AND ‘nanoparticles’ AND ‘Magnetic Resonance Imaging.’ 33 studies were included that described the development and utility of various NPs for AD imaging, including their coating, functionalization, MRI relaxivity, toxicity and bioavailability. NPs show immense promise for neuroimaging, due to superior relaxivity and biocompatibility compared with currently available imaging agents. Consistent reporting is imperative for further progress in this field.
Collapse
Affiliation(s)
- Marina Ulanova
- Centre for Healthy Brain Ageing, School of Psychiatry, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Anne Poljak
- Centre for Healthy Brain Ageing, School of Psychiatry, The University of New South Wales, Sydney, NSW, 2052, Australia
- Mark Wainwright Analytical Centre, The University of New South Wales, Sydney, NSW, 2052, Australia
- School of Medical Sciences, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Wei Wen
- Centre for Healthy Brain Ageing, School of Psychiatry, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Andre Bongers
- Mark Wainwright Analytical Centre, The University of New South Wales, Sydney, NSW, 2052, Australia
- Biological Resources Imaging Laboratory, Mark Wainwright Analytical Centre, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Lucy Gloag
- School of Chemistry, The University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Justin Gooding
- School of Chemistry, The University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, The University of New South Wales, Sydney, NSW, 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Richard Tilley
- Mark Wainwright Analytical Centre, The University of New South Wales, Sydney, NSW, 2052, Australia
- School of Chemistry, The University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, The University of New South Wales, Sydney, NSW, 2052, Australia
| | - Perminder Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, The University of New South Wales, Sydney, NSW, 2052, Australia
- Neuropsychiatric Institute, Euroa Centre, Prince of Wales Hospital, Sydney, NSW, 2052, Australia
| | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, The University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
19
|
Amanzadeh E, Esmaeili A, Abadi REN, Kazemipour N, Pahlevanneshan Z, Beheshti S. Quercetin conjugated with superparamagnetic iron oxide nanoparticles improves learning and memory better than free quercetin via interacting with proteins involved in LTP. Sci Rep 2019; 9:6876. [PMID: 31053743 PMCID: PMC6499818 DOI: 10.1038/s41598-019-43345-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 04/23/2019] [Indexed: 01/20/2023] Open
Abstract
Biomedical application of quercetin (QT) as an effective flavonoid has limitations due to its low bioavailability. Superparamagnetic iron oxide nanoparticle (SPION) is a novel drug delivery system that enhances the bioavailability of quercetin. The effect of short time usage of quercetin on learning and memory function and its signaling pathways in the healthy rat is not well understood. The aim of this study was to investigate the effect of free quercetin and in conjugation with SPION on learning and memory in healthy rats and to find quercetin target proteins involved in learning and memory using Morris water maze (MWM) and computational methods respectively. Results of MWM show an improvement in learning and memory of rats treated with either quercetin or QT-SPION. Better learning and memory functions using QT-SPION reveal increased bioavailability of quercetin. Comparative molecular docking studies show the better binding affinity of quercetin to RSK2, MSK1, CytC, Cdc42, Apaf1, FADD, CRK proteins. Quercetin in comparison to specific inhibitors of each protein also demonstrates a better QT binding affinity. This suggests that quercetin binds to proteins leading to prevent neural cell apoptosis and improves learning and memory. Therefore, SPIONs could increase the bioavailability of quercetin and by this way improve learning and memory.
Collapse
Affiliation(s)
- Elnaz Amanzadeh
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Abolghasem Esmaeili
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran.
| | | | - Nasrin Kazemipour
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Zari Pahlevanneshan
- Department of Chemistry, Catalysis Division, University of Isfahan, Isfahan, Iran
| | - Siamak Beheshti
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| |
Collapse
|
20
|
Fernández-Cabada T, Ramos-Gómez M. A Novel Contrast Agent Based on Magnetic Nanoparticles for Cholesterol Detection as Alzheimer's Disease Biomarker. NANOSCALE RESEARCH LETTERS 2019; 14:36. [PMID: 30684043 PMCID: PMC6349267 DOI: 10.1186/s11671-019-2863-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/10/2019] [Indexed: 05/03/2023]
Abstract
BACKGROUND Considering the high incidence of Alzheimer's disease among the world population over the years, and the costs that the disease poses in sanitary and social terms to countries, it is necessary to develop non-invasive diagnostic tests that allow to detect early biomarkers of the disease. Within the early diagnosis methods, the development of contrast agents for magnetic resonance imaging becomes especially useful. Accumulating evidence suggests that cholesterol may play a role in the pathogenesis of Alzheimer's disease since abnormal deposits of cholesterol surrounding senile plaques have been described in animal transgenic models and patients with Alzheimer's disease. In vivo experiments have also shown that diet-induced hypercholesterolemia enhances intraneuronal accumulation of β-amyloid protein accompanied by microgliosis and accelerates β-amyloid deposition in brains. PRESENTATION OF THE HYPOTHESIS In the present study, we propose for the first time the synthesis of a new nanoconjugate composed of magnetic nanoparticles bound to an anti-cholesterol antibody, to detect the abnormal deposits of cholesterol observed in senile plaques in Alzheimer's disease by magnetic resonance imaging. The nanoplatform could also reveal the decrease of cholesterol observed in neuronal plasmatic membranes associated with this pathology. TESTING THE HYPOTHESIS Experimental design to test the hypothesis will be done first in vitro and then in ex vivo and in vivo studies in a second stage. IMPLICATIONS OF THE HYPOTHESIS The designed nanoplatform could therefore detect cholesterol deposits at the cerebral level. The detection of this biomarker in areas coinciding with senile plaque accumulations could provide early information on the onset and progression of Alzheimer's disease.
Collapse
Affiliation(s)
- Tamara Fernández-Cabada
- Centre for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Campus de Montegancedo, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Milagros Ramos-Gómez
- Centre for Biomedical Technology (CTB), Universidad Politécnica de Madrid, Campus de Montegancedo, Pozuelo de Alarcón, 28223 Madrid, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| |
Collapse
|
21
|
Karthivashan G, Ganesan P, Park SY, Kim JS, Choi DK. Therapeutic strategies and nano-drug delivery applications in management of ageing Alzheimer's disease. Drug Deliv 2018; 25:307-320. [PMID: 29350055 PMCID: PMC6058502 DOI: 10.1080/10717544.2018.1428243] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/11/2018] [Indexed: 01/21/2023] Open
Abstract
In recent years, the incidental rate of neurodegenerative disorders has increased proportionately with the aging population. Alzheimer's disease (AD) is one of the most commonly reported neurodegenerative disorders, and it is estimated to increase by roughly 30% among the aged population. In spite of screening numerous drug candidates against various molecular targets of AD, only a few candidates - such as acetylcholinesterase inhibitors are currently utilized as an effective clinical therapy. However, targeted drug delivery of these drugs to the central nervous system (CNS) exhibits several limitations including meager solubility, low bioavailability, and reduced efficiency due to the impediments of the blood-brain barrier (BBB). Current advances in nanotechnology present opportunities to overcome such limitations in delivering active drug candidates. Nanodrug delivery systems are promising in targeting several therapeutic moieties by easing the penetration of drug molecules across the CNS and improving their bioavailability. Recently, a wide range of nano-carriers, such as polymers, emulsions, lipo-carriers, solid lipid carriers, carbon nanotubes, metal based carriers etc., have been adapted to develop successful therapeutics with sustained release and improved efficacy. Here, we discuss few recently updated nano-drug delivery applications that have been adapted in the field of AD therapeutics, and future prospects on potential molecular targets for nano-drug delivery systems.
Collapse
Affiliation(s)
- Govindarajan Karthivashan
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Diseases Konkuk University, Chungju, Republic of Korea
| | - Palanivel Ganesan
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Diseases Konkuk University, Chungju, Republic of Korea
- Nanotechnology research center, College of Biomedical and Health Science, Konkuk University, Chungju, Republic of Korea
| | - Shin-Young Park
- Department of Applied Life Science, Graduate school of Konkuk University, Chungju, Republic of Korea
| | - Joon-Soo Kim
- Department of Applied Life Science, Graduate school of Konkuk University, Chungju, Republic of Korea
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Diseases Konkuk University, Chungju, Republic of Korea
- Department of Applied Life Science, Graduate school of Konkuk University, Chungju, Republic of Korea
| |
Collapse
|
22
|
Ahlschwede KM, Curran GL, Rosenberg JT, Grant SC, Sarkar G, Jenkins RB, Ramakrishnan S, Poduslo JF, Kandimalla KK. Cationic carrier peptide enhances cerebrovascular targeting of nanoparticles in Alzheimer's disease brain. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 16:258-266. [PMID: 30300748 DOI: 10.1016/j.nano.2018.09.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 09/02/2018] [Accepted: 09/13/2018] [Indexed: 11/18/2022]
Abstract
Accumulation of amyloid beta (Aβ) peptides in the cerebral vasculature, referred to as cerebral amyloid angiopathy (CAA), is widely observed in Alzheimer's disease (AD) brain and was shown to accelerate cognitive decline. There is no effective method for detecting cerebrovascular amyloid (CVA) and treat CAA. The targeted nanoparticles developed in this study effectively migrated from the blood flow to the vascular endothelium as determined by using quartz crystal microbalance with dissipation monitoring (QCM-D) technology. We also improved the stability, and blood-brain barrier (BBB) transcytosis of targeted nanoparticles by coating them with a cationic BBB penetrating peptide (K16ApoE). The K16ApoE-Targeted nanoparticles demonstrated specific targeting of vasculotropic DutchAβ40 peptide accumulated in the cerebral vasculature. Moreover, K16ApoE-Targeted nanoparticles demonstrated significantly greater uptake into brain and provided specific MRI contrast to detect brain amyloid plaques.
Collapse
Affiliation(s)
- Kristen M Ahlschwede
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA; Molecular Neurobiology Laboratory, Departments of Neurology, Neuroscience and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA; Department of Pharmaceutical Sciences, College of Pharmacy, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Geoffry L Curran
- Molecular Neurobiology Laboratory, Departments of Neurology, Neuroscience and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Jens T Rosenberg
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, USA; Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA
| | - Samuel C Grant
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, USA; Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA
| | - Gobinda Sarkar
- Department of Experimental Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Robert B Jenkins
- Department of Experimental Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Subramanian Ramakrishnan
- The National High Magnetic Field Laboratory, Florida State University, Tallahassee, FL, USA; Department of Chemical & Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA
| | - Joseph F Poduslo
- Molecular Neurobiology Laboratory, Departments of Neurology, Neuroscience and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA; Molecular Neurobiology Laboratory, Departments of Neurology, Neuroscience and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA.
| |
Collapse
|
23
|
Magnetic Nanoparticles Applications for Amyloidosis Study and Detection: A Review. NANOMATERIALS 2018; 8:nano8090740. [PMID: 30231587 PMCID: PMC6164038 DOI: 10.3390/nano8090740] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 09/14/2018] [Accepted: 09/16/2018] [Indexed: 12/27/2022]
Abstract
Magnetic nanoparticles (MNPs) have great potential in biomedical and clinical applications because of their many unique properties. This contribution provides an overview of the MNPs mainly used in the field of amyloid diseases. The first part discusses their use in understanding the amyloid mechanisms of fibrillation, with emphasis on their ability to control aggregation of amyloidogenic proteins. The second part deals with the functionalization by various moieties of numerous MNPs’ surfaces (molecules, peptides, antibody fragments, or whole antibodies of MNPs) for the detection and the quantification of amyloid aggregates. The last part of this review focuses on the use of MNPs for magnetic-resonance-based amyloid imaging in biomedical fields, with particular attention to the application of gadolinium-based paramagnetic nanoparticles (AGuIX), which have been recently developed. Biocompatible AGuIX nanoparticles show favorable characteristics for in vivo use, such as nanometric and straightforward functionalization. Their properties have enabled their application in MRI. Here, we report that AGuIX nanoparticles grafted with the Pittsburgh compound B can actively target amyloid aggregates in the brain, beyond the blood–brain barrier, and remain the first step in observing amyloid plaques in a mouse model of Alzheimer’s disease.
Collapse
|
24
|
Fernández T, Martínez-Serrano A, Cussó L, Desco M, Ramos-Gómez M. Functionalization and Characterization of Magnetic Nanoparticles for the Detection of Ferritin Accumulation in Alzheimer's Disease. ACS Chem Neurosci 2018; 9:912-924. [PMID: 29298040 DOI: 10.1021/acschemneuro.7b00260] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Early diagnosis in Alzheimer's disease (AD), prior to the appearance of marked clinical symptoms, is critical to prevent irreversible neuronal damage and neural malfunction that lead to dementia and death. Therefore, there is an urgent need to generate new contrast agents which reveal by a noninvasive method the presence of some of the pathological signs of AD. In the present study, we demonstrate for the first time a new nanoconjugate composed of magnetic nanoparticles bound to an antiferritin antibody, which has been developed based on the existence of iron deposits and high levels of the ferritin protein present in areas with a high accumulation of amyloid plaques (particularly the subiculum in the hippocampal area) in the brain of a transgenic mouse model with five familial AD mutations. Both in vitro and after intravenous injection, functionalized magnetic nanoparticles were able to recognize and bind specifically to the ferritin protein accumulated in the subiculum area of the AD transgenic mice.
Collapse
Affiliation(s)
- Tamara Fernández
- Centre for Biomedical Technology (CTB), Universidad Politécnica de Madrid, 28223 Madrid, Spain
| | - Alberto Martínez-Serrano
- Department of Molecular Biology
and Centre for Molecular Biology “Severo Ochoa” (CBMSO),
Universidad Autónoma de Madrid and Consejo Superior de Investigaciones
Científicas, 28049 Madrid, Spain
| | - Lorena Cussó
- Departamento de Ingeniería Biomédica e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, 28911 Leganés, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Centro de Investigación
Biomédica en Red de Salud Mental (CIBERSAM), 28029 Madrid, Spain
| | - Manuel Desco
- Departamento de Ingeniería Biomédica e Ingeniería Aeroespacial, Universidad Carlos III de Madrid, 28911 Leganés, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Centro de Investigación
Biomédica en Red de Salud Mental (CIBERSAM), 28029 Madrid, Spain
| | - Milagros Ramos-Gómez
- Centre for Biomedical Technology (CTB), Universidad Politécnica de Madrid, 28223 Madrid, Spain
- CIBER de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| |
Collapse
|
25
|
Giannoni P, Badaut J, Dargazanli C, Fayd'Herbe De Maudave A, Klement W, Costalat V, Marchi N. The pericyte-glia interface at the blood-brain barrier. Clin Sci (Lond) 2018; 132:361-374. [PMID: 29439117 DOI: 10.1042/cs20171634] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 01/04/2018] [Accepted: 01/04/2018] [Indexed: 12/30/2022]
Abstract
The cerebrovasculature is a multicellular structure with varying rheological and permeability properties. The outer wall of the brain capillary endothelium is enclosed by pericytes and astrocyte end feet, anatomically assembled to guarantee barrier functions. We, here, focus on the pericyte modifications occurring in disease conditions, reviewing evidence supporting the interplay amongst pericytes, the endothelium, and glial cells in health and pathology. Deconstruction and reactivity of pericytes and glial cells around the capillary endothelium occur in response to traumatic brain injury, epilepsy, and neurodegenerative disorders, impacting vascular permeability and participating in neuroinflammation. As this represents a growing field of research, addressing the multicellular reorganization occurring at the outer wall of the blood-brain barrier (BBB) in response to an acute insult or a chronic disease could disclose novel disease mechanisms and therapeutic targets.
Collapse
Affiliation(s)
| | - Jerome Badaut
- Laboratory of Brain Molecular Imaging, CNRS UMR5287, University of Bordeaux, France
- Basic Science Departments, Loma Linda University School of Medicine, CA, U.S.A
| | - Cyril Dargazanli
- Neuroradiology, University Hospital, Montpellier, France
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Alexis Fayd'Herbe De Maudave
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Wendy Klement
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Vincent Costalat
- Neuroradiology, University Hospital, Montpellier, France
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| | - Nicola Marchi
- Laboratory of Cerebrovascular Mechanisms of Brain Disorders, Department of Neuroscience, Institute of Functional Genomics (UMR 5203 CNRS - U 1191 INSERM, University of Montpellier), Montpellier, France
| |
Collapse
|
26
|
Stem Cells in Alzheimer’s Disease: Current Standing and Future Challenges. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1079:93-102. [DOI: 10.1007/5584_2018_214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
27
|
Huang L, Hu J, Huang S, Wang B, Siaw-Debrah F, Nyanzu M, Zhang Y, Zhuge Q. Nanomaterial applications for neurological diseases and central nervous system injury. Prog Neurobiol 2017; 157:29-48. [PMID: 28743465 DOI: 10.1016/j.pneurobio.2017.07.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 07/18/2017] [Accepted: 07/18/2017] [Indexed: 12/20/2022]
Abstract
The effectiveness of noninvasive treatment for neurological disease is generally limited by the poor entry of therapeutic agents into the central nervous system (CNS). Most CNS drugs cannot permeate into the brain parenchyma because of the blood-brain barrier thus, overcoming this problem has become one of the most significant challenges in the development of neurological therapeutics. Nanotechnology has emerged as an innovative alternative for treating neurological diseases. In fact, rapid advances in nanotechnology have provided promising solutions to this challenge. This review highlights the applications of nanomaterials in the developing neurological field and discusses the evidence for their efficacies.
Collapse
Affiliation(s)
- Lijie Huang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Jiangnan Hu
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Shengwei Huang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Brian Wang
- Center for Neuroscience Discovery, Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Felix Siaw-Debrah
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Mark Nyanzu
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Yu Zhang
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China
| | - Qichuan Zhuge
- Department of Neurosurgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China; Zhejiang Provincial Key Laboratory of Aging and Neurological Disorder Research, First Affiliated Hospital, Wenzhou Medical University, Wenzhou City, Zhejiang Province, 325000, PR China.
| |
Collapse
|
28
|
Yarjanli Z, Ghaedi K, Esmaeili A, Rahgozar S, Zarrabi A. Iron oxide nanoparticles may damage to the neural tissue through iron accumulation, oxidative stress, and protein aggregation. BMC Neurosci 2017; 18:51. [PMID: 28651647 PMCID: PMC5485499 DOI: 10.1186/s12868-017-0369-9] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 06/22/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND In the recent decade, iron oxide nanoparticles (IONPs) have been proposed for several applications in the central nervous system (CNS), including targeting amyloid beta (Aβ) in the arteries, inhibiting the microglial cells, delivering drugs, and increasing contrast in magnetic resonance imaging. Conversely, a notable number of studies have reported the role of iron in neurodegenerative diseases. Therefore, this study has reviewed the recent studies to determine whether IONPs iron can threaten the cellular viability same as iron. RESULTS Iron contributes in Fenton's reaction and produces reactive oxygen species (ROS). ROS cause to damage the macromolecules and organelles of the cell via oxidative stress. Iron accumulation and oxidative stress are able to aggregate some proteins, including Aβ and α-synuclein, which play a critical role in Alzheimer's and Parkinson's diseases, respectively. Iron accumulation, oxidative stress, and protein aggregation make a positive feedback loop, which can be toxic for the cell. The release of iron ions from IONPs may result in iron accumulation in the targeted tissue, and thus, activate the positive feedback loop. However, the levels of IONPs induced toxicity depend on the size, concentration, surface charge, and the type of coating and functional groups of IONPs. CONCLUSION IONPs depending on their properties can lead to iron accumulation, oxidative stress and protein aggregation in the neural cells. Therefore, in order to apply IONPs in the CNS, the consideration of IONPs properties is crucial.
Collapse
Affiliation(s)
- Zahra Yarjanli
- Department of Biology, Faculty of Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| | - Kamran Ghaedi
- Department of Biology, Faculty of Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| | - Abolghasem Esmaeili
- Department of Biology, Faculty of Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| | - Soheila Rahgozar
- Department of Biology, Faculty of Sciences, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| | - Ali Zarrabi
- Department of Biotechnology, Faculty of Advanced Sciences and Technologies, University of Isfahan, Hezar Jerib Ave., Azadi Square, Isfahan, 81746-73441 Iran
| |
Collapse
|
29
|
Salem H, Rocha NP, Colpo GD, Teixeira AL. Moving from the Dish to the Clinical Practice: A Decade of Lessons and Perspectives from the Pre-Clinical and Clinical Stem Cell Studies for Alzheimer’s Disease. J Alzheimers Dis 2016; 53:1209-30. [DOI: 10.3233/jad-160250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Haitham Salem
- Department of Psychiatry and Behavioral Sciences, Neuropsychiatry Program, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
- Regenerative Medicine Program, University of Lübeck, Schleswig-Holstein, Germany
| | - Natalia Pessoa Rocha
- Department of Psychiatry and Behavioral Sciences, Neuropsychiatry Program, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
| | - Gabriela Delevati Colpo
- Department of Psychiatry and Behavioral Sciences, Neuropsychiatry Program, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
| | - Antonio Lucio Teixeira
- Department of Psychiatry and Behavioral Sciences, Neuropsychiatry Program, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
30
|
Imaging of Cerebral Amyloid Angiopathy with Bivalent (99m)Tc-Hydroxamamide Complexes. Sci Rep 2016; 6:25990. [PMID: 27181612 PMCID: PMC4867616 DOI: 10.1038/srep25990] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 04/26/2016] [Indexed: 11/26/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA), characterized by the deposition of amyloid aggregates in the walls of cerebral vasculature, is a major factor in intracerebral hemorrhage and vascular cognitive impairment and is also associated closely with Alzheimer’s disease (AD). We previously reported 99mTc-hydroxamamide (99mTc-Ham) complexes with a bivalent amyloid ligand showing high binding affinity for β-amyloid peptide (Aβ(1–42)) aggregates present frequently in the form in AD. In this article, we applied them to CAA-specific imaging probes, and evaluated their utility for CAA-specific imaging. In vitro inhibition assay using Aβ(1–40) aggregates deposited mainly in CAA and a brain uptake study were performed for 99mTc-Ham complexes, and all 99mTc-Ham complexes with an amyloid ligand showed binding affinity for Aβ(1–40) aggregates and very low brain uptake. In vitro autoradiography of human CAA brain sections and ex vivo autoradiography of Tg2576 mice were carried out for bivalent 99mTc-Ham complexes ([99mTc]SB2A and [99mTc]BT2B), and they displayed excellent labeling of Aβ depositions in human CAA brain sections and high affinity and selectivity to CAA in transgenic mice. These results may offer new possibilities for the development of clinically useful CAA-specific imaging probes based on the 99mTc-Ham complex.
Collapse
|
31
|
Rokka J, Snellman A, Kaasalainen M, Salonen J, Zona C, La Ferla B, Nicotra F, Re F, Masserini M, Forsback S, Lopez-Picon F, Rinne JO, Haaparanta-Solin M, Solin O. (18)F-labeling syntheses and preclinical evaluation of functionalized nanoliposomes for Alzheimer's disease. Eur J Pharm Sci 2016; 88:257-66. [PMID: 26993963 DOI: 10.1016/j.ejps.2016.03.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/17/2016] [Accepted: 03/14/2016] [Indexed: 01/06/2023]
Abstract
The aim of the present study was to synthesize functionalized (18)F-labeled NLs ((18)F-NLs) and evaluate their biological behavior in mouse models of Alzheimer's disease (AD) using positron emission tomography (PET) and ex vivo brain autoradiography. (18)F-fluorine was introduced to (18)F-NLs either by using a core forming (18)F-lipid or by encapsulating a (18)F-tracer, (18)F-treg-curcumin inside the NLs. Phosphatidic acid (PA) and curcumin derivative (Curc) functionalized (18)F-NLs with or without additional mApoE functionalization were produced using thin film hydration. The biodistribution and β-amyloid plaque-binding ability of (18)F-NLs were studied in wild type mice and AD mouse models using in vivo PET imaging and ex vivo brain autoradiography at 60min after (18)F-NL injection. Functionalized (18)F-NLs were successfully synthesized. The preclinical evaluation in mice showed that the functional group affected the biodistribution of (18)F-NLs. Further functionalization with mApoE increased the brain-to-blood ratio of (18)F-NLs but the overall brain uptake remained low with all functionalized (18)F-NLs. The liposomal encapsulation of (18)F-treg-curcumin was not successful and preclinical results of encapsulated (18)F-treg-curcumin and plain (18)F-treg-curcumin were identical. Although the studied functionalized (18)F-NLs were not suitable for PET imaging as such, the synthesis techniques introduced in this study can be utilized to modify the biological behavior of (18)F-labeled NLs.
Collapse
Affiliation(s)
- Johanna Rokka
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, Turku, Finland.
| | - Anniina Snellman
- Turku PET Centre, Preclinical Imaging, University of Turku, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | | | - Jarno Salonen
- Laboratory of Industrial Physics, University of Turku, Finland
| | - Cristiano Zona
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milano, Italy
| | - Barbara La Ferla
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milano, Italy
| | - Francesco Nicotra
- Department of Biotechnology and Bioscience, University of Milano-Bicocca, Milano, Italy
| | - Francesca Re
- Department of Health Science, University of Milano-Bicocca, Monza, Italy
| | - Massimo Masserini
- Department of Health Science, University of Milano-Bicocca, Monza, Italy
| | - Sarita Forsback
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, Turku, Finland
| | - Francisco Lopez-Picon
- Turku PET Centre, Preclinical Imaging, University of Turku, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Juha O Rinne
- Turku PET Centre, University of Turku, Turku, Finland; Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland
| | - Merja Haaparanta-Solin
- Turku PET Centre, Preclinical Imaging, University of Turku, Turku, Finland; MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Olof Solin
- Turku PET Centre, Radiopharmaceutical Chemistry Laboratory, University of Turku, Turku, Finland; Turku PET Centre, Accelerator Laboratory, Åbo Akademi University, Turku, Finland; Department of Chemistry, University of Turku, Turku, Finland
| |
Collapse
|
32
|
Migliore L, Uboldi C, Di Bucchianico S, Coppedè F. Nanomaterials and neurodegeneration. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2015; 56:149-170. [PMID: 25627719 DOI: 10.1002/em.21931] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 11/13/2014] [Indexed: 06/04/2023]
Abstract
The increasing application of nanotechnology in various industrial, environmental, and human settings raises questions surrounding the potential adverse effects induced by nanosized materials to human health, including the possible neurotoxic and neuroinflammatory properties of those substances and their capability to induce neurodegeneration. In this review, a panel of metal oxide nanoparticles (NPs), namely titanium dioxide, silicon dioxide, zinc oxide, copper oxide, iron NPs, and carbon nanotubes have been focused. An overview has been provided of the in vitro and in vivo evidence of adverse effects to the central nervous system. Research indicated that these nanomaterials (NMs) not only reach the brain, but also can cause a certain degree of brain tissue damage, including cytotoxicity, genotoxicity, induction of oxidative stress, and inflammation, all potentially involved in the onset and progression of neurodegeneration. Surface chemistry of the NMs may play an important role in their localization and subsequent effects on the brain of rodents. In addition, NM shape differences may induce varying degrees of neurotoxicity. However, one of the potential biomedical applications of NMs is nanodevices for early diagnostic and novel therapeutic approaches to counteract age related diseases. In this context, engineered NMs were promising vehicles to carry diagnostic and therapeutic compounds across the blood-brain barrier, thereby representing very timely and attractive theranostic tools in neurodegenerative diseases. Therefore, a careful assessment of the risk-benefit ratio must be taken into consideration in using nanosized materials.
Collapse
Affiliation(s)
- Lucia Migliore
- Medical Genetics Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma, 55 - 56126, Pisa, Italy
| | | | | | | |
Collapse
|
33
|
Agyare EK, Jaruszewski KM, Curran GL, Rosenberg JT, Grant SC, Lowe VJ, Ramakrishnan S, Paravastu AK, Poduslo JF, Kandimalla KK. Engineering theranostic nanovehicles capable of targeting cerebrovascular amyloid deposits. J Control Release 2014; 185:121-9. [PMID: 24735640 DOI: 10.1016/j.jconrel.2014.04.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/21/2014] [Accepted: 04/04/2014] [Indexed: 11/26/2022]
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by the deposition of amyloid beta (Aβ) proteins within the walls of the cerebral vasculature with subsequent aggressive vascular inflammation leading to recurrent hemorrhagic strokes. The objective of the study was to develop theranostic nanovehicles (TNVs) capable of a) targeting cerebrovascular amyloid; b) providing magnetic resonance imaging (MRI) contrast for the early detection of CAA; and c) treating cerebrovascular inflammation resulting from CAA. The TNVs comprised of a polymeric nanocore made from Magnevist (MRI contrast agent) conjugated chitosan. The nanocore was also loaded with cyclophosphamide (CYC), an immunosuppressant shown to reduce the cerebrovascular inflammation in CAA. Putrescine modified F(ab')2 fragment of anti-amyloid antibody, IgG4.1 (pF(ab')24.1) was conjugated to the surface of the nanocore to target cerebrovascular amyloid. The average size of the control chitosan nanoparticles (conjugated with albumin and are devoid of Magnevist, CYC, and pF(ab')24.1) was 164±1.2 nm and that of the TNVs was 239±4.1 nm. The zeta potential values of the CCNs and TNVs were 21.6±1.7 mV and 11.9±0.5 mV, respectively. The leakage of Magnevist from the TNVs was a modest 0.2% over 4 days, and the CYC release from the TNVs followed Higuchi's model that describes sustained drug release from polymeric matrices. The studies conducted in polarized human microvascular endothelial cell monolayers (hCMEC/D3) in vitro as well as in mice in vivo have demonstrated the ability of TNVs to target cerebrovascular amyloid. In addition, the TNVs provided contrast for imaging cerebrovascular amyloid using MRI and single photon emission computed tomography. Moreover, the TNVs were shown to reduce pro-inflammatory cytokine production by the Aβ challenged blood brain barrier (BBB) endothelium more effectively than the cyclophosphamide alone.
Collapse
Affiliation(s)
- Edward K Agyare
- Division of Basic Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, 1520 S. MLK BLVD, Tallahassee 32307, USA
| | - Kristen M Jaruszewski
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, 308 Harvard St. SE, Room 9-149A WDH, Minneapolis 55455, USA; Molecular Neurobiology Laboratory, Departments of Neurology, Neuroscience, and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, 200 1st Street SW, Rochester 55905, USA
| | - Geoffry L Curran
- Molecular Neurobiology Laboratory, Departments of Neurology, Neuroscience, and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, 200 1st Street SW, Rochester 55905, USA
| | - Jens T Rosenberg
- The Florida State University and National High Magnetic Field Laboratory, 1800 East Paul Dirac Drive, Tallahassee 32310, USA
| | - Samuel C Grant
- The Florida State University and National High Magnetic Field Laboratory, 1800 East Paul Dirac Drive, Tallahassee 32310, USA; Department of Chemical and Biomedical Engineering, Florida A&M University-Florida State University College of Engineering, 2525 Pottsdamer Street, Tallahassee 32310, USA
| | - Val J Lowe
- Nuclear Medicine, Department of Radiology, Mayo Clinic, 200 1st Street SW, Rochester 55905, USA
| | - Subramanian Ramakrishnan
- Department of Chemical and Biomedical Engineering, Florida A&M University-Florida State University College of Engineering, 2525 Pottsdamer Street, Tallahassee 32310, USA
| | - Anant K Paravastu
- The Florida State University and National High Magnetic Field Laboratory, 1800 East Paul Dirac Drive, Tallahassee 32310, USA; Department of Chemical and Biomedical Engineering, Florida A&M University-Florida State University College of Engineering, 2525 Pottsdamer Street, Tallahassee 32310, USA
| | - Joseph F Poduslo
- Molecular Neurobiology Laboratory, Departments of Neurology, Neuroscience, and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, 200 1st Street SW, Rochester 55905, USA
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, University of Minnesota, 308 Harvard St. SE, Room 9-149A WDH, Minneapolis 55455, USA; Molecular Neurobiology Laboratory, Departments of Neurology, Neuroscience, and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, 200 1st Street SW, Rochester 55905, USA.
| |
Collapse
|
34
|
Klohs J, Rudin M, Shimshek DR, Beckmann N. Imaging of cerebrovascular pathology in animal models of Alzheimer's disease. Front Aging Neurosci 2014; 6:32. [PMID: 24659966 PMCID: PMC3952109 DOI: 10.3389/fnagi.2014.00032] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 02/19/2014] [Indexed: 01/04/2023] Open
Abstract
In Alzheimer's disease (AD), vascular pathology may interact with neurodegeneration and thus aggravate cognitive decline. As the relationship between these two processes is poorly understood, research has been increasingly focused on understanding the link between cerebrovascular alterations and AD. This has at last been spurred by the engineering of transgenic animals, which display pathological features of AD and develop cerebral amyloid angiopathy to various degrees. Transgenic models are versatile for investigating the role of amyloid deposition and vascular dysfunction, and for evaluating novel therapeutic concepts. In addition, research has benefited from the development of novel imaging techniques, which are capable of characterizing vascular pathology in vivo. They provide vascular structural read-outs and have the ability to assess the functional consequences of vascular dysfunction as well as to visualize and monitor the molecular processes underlying these pathological alterations. This article focusses on recent in vivo small animal imaging studies addressing vascular aspects related to AD. With the technical advances of imaging modalities such as magnetic resonance, nuclear and microscopic imaging, molecular, functional and structural information related to vascular pathology can now be visualized in vivo in small rodents. Imaging vascular and parenchymal amyloid-β (Aβ) deposition as well as Aβ transport pathways have been shown to be useful to characterize their dynamics and to elucidate their role in the development of cerebral amyloid angiopathy and AD. Structural and functional imaging read-outs have been employed to describe the deleterious affects of Aβ on vessel morphology, hemodynamics and vascular integrity. More recent imaging studies have also addressed how inflammatory processes partake in the pathogenesis of the disease. Moreover, imaging can be pivotal in the search for novel therapies targeting the vasculature.
Collapse
Affiliation(s)
- Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich Zurich, Switzerland ; Neuroscience Center Zurich, University of Zurich and ETH Zurich Zurich, Switzerland
| | - Markus Rudin
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich Zurich, Switzerland ; Neuroscience Center Zurich, University of Zurich and ETH Zurich Zurich, Switzerland ; Institute of Pharmacology and Toxicology, University of Zurich Zurich, Switzerland
| | - Derya R Shimshek
- Autoimmunity, Transplantation and Inflammation/Neuroinflammation Department, Novartis Institutes for BioMedical Research Basel, Switzerland
| | - Nicolau Beckmann
- Analytical Sciences and Imaging, Novartis Institutes for BioMedical Research Basel, Switzerland
| |
Collapse
|
35
|
Hamilton AM, Mallett C, Foster PJ. High-resolution MRI and nanoparticles: the future of brain imaging. FUTURE NEUROLOGY 2014. [DOI: 10.2217/fnl.13.77] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
ABSTRACT: Cellular MRI uses superparamagnetic iron oxide nanoparticles to label cells (in vitro or in vivo) for detection in magnetic resonance images. The infiltration of inflammatory macrophages can be visualized in brain diseases, such as multiple sclerosis, stroke and Alzheimer‘s disease, and correlates with disease severity and responses to treatments. Mesenchymal stromal cells, neural stem cells and immune cells used as cell therapies in CNS diseases can be tracked in vivo over time to determine their migration and dispersion. Tracking labeled cancer cells provides information about metastasis and proliferative status in preclinical tumor models. Ongoing technical improvements come from the development of new particles, the use of fluorine-based contrast agents and the refinement of high-field MRI for cell tracking.
Collapse
Affiliation(s)
- Amanda M Hamilton
- Imaging Research Laboratories, Robarts Research Institute, London, ON, N6A 5K8, Canada
| | - Christiane Mallett
- Imaging Research Laboratories, Robarts Research Institute, London, ON, N6A 5K8, Canada
| | - Paula J Foster
- Department of Medical Biophysics, Western University, London, ON, Canada
| |
Collapse
|
36
|
Choi SS, Lee SR, Kim SU, Lee HJ. Alzheimer's disease and stem cell therapy. Exp Neurobiol 2014; 23:45-52. [PMID: 24737939 PMCID: PMC3984956 DOI: 10.5607/en.2014.23.1.45] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 02/28/2014] [Accepted: 02/28/2014] [Indexed: 12/19/2022] Open
Abstract
The loss of neuronal cells in the central nervous system may occur in many neurodegenerative diseases. Alzheimer's disease is a common senile disease in people over 65 years, and it causes impairment characterized by the decline of mental function, including memory loss and cognitive impairment, and affects the quality of life of patients. However, the current therapeutic strategies against AD are only to relieve symptoms, but not to cure it. Because there are only a few therapeutic strategies against Alzheimer's disease, we need to understand the pathogenesis of this disease. Cell therapy may be a powerful tool for the treatment of Alzheimer's disease. This review will discuss the characteristics of Alzheimer's disease and various available therapeutic strategies.
Collapse
Affiliation(s)
- Sung S Choi
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul 156-756, Korea
| | - Sang-Rae Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang 363-883, Korea
| | - Seung U Kim
- Division of Neurology, Department of Medicine, University of British Columbia, Vancouver 317-2194, Canada
| | - Hong J Lee
- Medical Research Institute, Chung-Ang University College of Medicine, Seoul 156-756, Korea
| |
Collapse
|
37
|
Jaruszewski KM, Curran GL, Swaminathan SK, Rosenberg JT, Grant SC, Ramakrishnan S, Lowe VJ, Poduslo JF, Kandimalla KK. Multimodal nanoprobes to target cerebrovascular amyloid in Alzheimer's disease brain. Biomaterials 2013; 35:1967-76. [PMID: 24331706 DOI: 10.1016/j.biomaterials.2013.10.075] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 10/27/2013] [Indexed: 12/12/2022]
Abstract
Cerebral amyloid angiopathy (CAA) results from the accumulation of Aβ proteins primarily within the media and adventitia of small arteries and capillaries of the cortex and leptomeninges. CAA affects a majority of Alzheimer's disease (AD) patients and is associated with a rapid decline in cognitive reserve. Unfortunately, there is no pre-mortem diagnosis available for CAA. Furthermore, treatment options are few and relatively ineffective. To combat this issue, we have designed nanovehicles (nanoparticles-IgG4.1) capable of targeting cerebrovascular amyloid (CVA) and serving as early diagnostic and therapeutic agents. These nanovehicles were loaded with Gadolinium (Gd) based (Magnevist(®)) magnetic resonance imaging contrast agents or single photon emission computed tomography (SPECT) agents, such as (125)I. In addition, the nanovehicles carry either anti-inflammatory and anti-amyloidogenic agents such as curcumin or immunosuppressants such as dexamethasone, which were previously shown to reduce cerebrovascular inflammation. Owing to the anti-amyloid antibody (IgG4.1) grafted on the surface, the nanovehicles are capable of specifically targeting CVA deposits. The nanovehicles effectively marginate from the blood flow to the vascular wall as determined by using quartz crystal microbalance with dissipation monitoring (QCM-D) technology. They demonstrate excellent distribution to the brain vasculature and target CVA, thus providing MRI and SPECT contrast specific to the CVA in the brain. In addition, they also display the potential to carry therapeutic agents to reduce cerebrovascular inflammation associated with CAA, which is believed to trigger hemorrhage in CAA patients.
Collapse
Affiliation(s)
- Kristen M Jaruszewski
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; Molecular Neurobiology Laboratory, Department of Neurology, Neuroscience and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA; Department of Pharmaceutics, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Geoffry L Curran
- Molecular Neurobiology Laboratory, Department of Neurology, Neuroscience and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Suresh K Swaminathan
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jens T Rosenberg
- The Florida State University and National High Magnetic Field Laboratory, Tallassee, FL 32310, USA
| | - Samuel C Grant
- The Florida State University and National High Magnetic Field Laboratory, Tallassee, FL 32310, USA; Department of Chemical and Biomedical Engineering, College of Engineering, Florida A&M University-Florida State University, Tallahassee, FL 32310, USA
| | - Subramanian Ramakrishnan
- The Florida State University and National High Magnetic Field Laboratory, Tallassee, FL 32310, USA; Department of Chemical and Biomedical Engineering, College of Engineering, Florida A&M University-Florida State University, Tallahassee, FL 32310, USA
| | - Val J Lowe
- Nuclear Medicine, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Joseph F Poduslo
- Molecular Neurobiology Laboratory, Department of Neurology, Neuroscience and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Karunya K Kandimalla
- Department of Pharmaceutics and Brain Barriers Research Center, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA; Molecular Neurobiology Laboratory, Department of Neurology, Neuroscience and Biochemistry/Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| |
Collapse
|
38
|
Sillerud LO, Solberg NO, Chamberlain R, Orlando RA, Heidrich JE, Brown DC, Brady CI, Vander Jagt TA, Garwood M, Vander Jagt DL. SPION-enhanced magnetic resonance imaging of Alzheimer's disease plaques in AβPP/PS-1 transgenic mouse brain. J Alzheimers Dis 2013; 34:349-65. [PMID: 23229079 DOI: 10.3233/jad-121171] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In our program to develop non-invasive magnetic resonance imaging (MRI) methods for the diagnosis of Alzheimer's disease (AD), we have synthesized antibody-conjugated, superparamagnetic iron oxide nanoparticles (SPIONs) for use as an in vivo agent for MRI detection of amyloid-β plaques in AD. Here we report studies in AβPP/PS1 transgenic mice, which demonstrate the ability of novel anti-AβPP conjugated SPIONs to penetrate the blood-brain barrier to act as a contrast agent for MR imaging of plaques. The conspicuity of the plaques increased from an average Z-score of 5.1 ± 0.5 to 8.3 ± 0.2 when the plaque contrast to noise ratio was compared in control AD mice with AD mice treated with SPIONs. The number of MRI-visible plaques per brain increased from 347 ± 45 in the control AD mice, to 668 ± 86 in the SPION treated mice. These results indicated that our SPION enhanced amyloid-β detection method delivers an efficacious, non-invasive MRI detection method in transgenic mice.
Collapse
Affiliation(s)
- Laurel O Sillerud
- Department of Biochemistry and Molecular Biology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zhang M, Mao X, Yu Y, Wang CX, Yang YL, Wang C. Nanomaterials for reducing amyloid cytotoxicity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2013; 25:3780-801. [PMID: 23722464 DOI: 10.1002/adma.201301210] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Indexed: 05/20/2023]
Abstract
This review is intended to reflect the recent progress on therapeutic applications of nanomaterials in amyloid diseases. The progress on anti-amyloid functions of various nanomaterials including inorganic nanoparticles, polymeric nanoparticles, carbon nanomaterials and biomolecular aggregates, is reviewed and discussed. The main functionalization strategies for general nanoparticle modifications are reviewed for potential applications of targeted therapeutics. The interaction mechanisms between amyloid peptides and nanomaterials are discussed from the perspectives of dominant interactions and kinetics. The encapsulation of anti-amyloid drugs, targeted drug delivery, controlled drug release and drug delivery crossing blood brain barrier by application of nanomaterials would also improve the therapeutics of amyloid diseases.
Collapse
Affiliation(s)
- Min Zhang
- National Center for Nanoscience and Technology, Beijing 100190, China
| | | | | | | | | | | |
Collapse
|
40
|
Sabbagh JJ, Kinney JW, Cummings JL. Alzheimer's disease biomarkers in animal models: closing the translational gap. AMERICAN JOURNAL OF NEURODEGENERATIVE DISEASE 2013; 2:108-120. [PMID: 23844335 PMCID: PMC3703123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 05/31/2013] [Indexed: 06/02/2023]
Abstract
The rising prevalence of Alzheimer's disease (AD) is rapidly becoming one of the largest health and economic challenges in the world. There is a growing need for the development and implementation of reliable biomarkers for AD that can be used to assist in diagnosis, inform disease progression, and monitor therapeutic efficacy. Preclinical models permit the evaluation of candidate biomarkers and assessment of pipeline agents before clinical trials are initiated and provide a translational opportunity to advance biomarker discovery. Fast and inexpensive data can be obtained from examination of peripheral markers, though they currently lack the sensitivity and consistency of imaging techniques such as MRI or PET. Plasma and cerebrospinal fluid (CSF) biomarkers in animal models can assist in development and implementation of similar approaches in clinical populations. These biomarkers may also be invaluable in decisions to advance a treatment to human testing. Longitudinal studies in AD models can determine initial presentation and progression of biomarkers that may also be used to evaluate disease-modifying efficacy of drugs. The refinement of biomarker approaches in preclinical systems will not only aid in drug development, but may facilitate diagnosis and disease monitoring in AD patients.
Collapse
|
41
|
Alzheimer's disease biomarkers: correspondence between human studies and animal models. Neurobiol Dis 2013; 56:116-30. [PMID: 23631871 DOI: 10.1016/j.nbd.2013.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 04/11/2013] [Accepted: 04/18/2013] [Indexed: 01/05/2023] Open
Abstract
Alzheimer's disease (AD) represents an escalating global threat as life expectancy and disease prevalence continue to increase. There is a considerable need for earlier diagnoses to improve clinical outcomes. Fluid biomarkers measured from cerebrospinal fluid (CSF) and blood, or imaging biomarkers have considerable potential to assist in the diagnosis and management of AD. An additional important utility of biomarkers is in novel therapeutic development and clinical trials to assess efficacy and side effects of therapeutic interventions. Because many biomarkers are initially examined in animal models, the extent to which markers translate from animals to humans is an important issue. The current review highlights many existing and pipeline biomarker approaches, focusing on the degree of correspondence between AD patients and animal models. The review also highlights the need for greater translational correspondence between human and animal biomarkers.
Collapse
|
42
|
Carrero I, Gonzalo M, Martin B, Sanz-Anquela J, Arévalo-Serrano J, Gonzalo-Ruiz A. Oligomers of beta-amyloid protein (Aβ1-42) induce the activation of cyclooxygenase-2 in astrocytes via an interaction with interleukin-1beta, tumour necrosis factor-alpha, and a nuclear factor kappa-B mechanism in the rat brain. Exp Neurol 2012; 236:215-27. [DOI: 10.1016/j.expneurol.2012.05.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2012] [Revised: 04/22/2012] [Accepted: 05/05/2012] [Indexed: 11/25/2022]
|
43
|
Anti-Aβ-MAb and dually decorated nanoliposomes: Effect of Aβ1-42 peptides on interaction with hCMEC/D3 cells. Eur J Pharm Biopharm 2012; 81:49-56. [DOI: 10.1016/j.ejpb.2012.02.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2011] [Revised: 02/09/2012] [Accepted: 02/10/2012] [Indexed: 11/18/2022]
|
44
|
Manook A, Yousefi BH, Willuweit A, Platzer S, Reder S, Voss A, Huisman M, Settles M, Neff F, Velden J, Schoor M, von der Kammer H, Wester HJ, Schwaiger M, Henriksen G, Drzezga A. Small-animal PET imaging of amyloid-beta plaques with [11C]PiB and its multi-modal validation in an APP/PS1 mouse model of Alzheimer's disease. PLoS One 2012; 7:e31310. [PMID: 22427802 PMCID: PMC3302888 DOI: 10.1371/journal.pone.0031310] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 01/05/2012] [Indexed: 11/18/2022] Open
Abstract
In vivo imaging and quantification of amyloid-β plaque (Aβ) burden in small-animal models of Alzheimer's disease (AD) is a valuable tool for translational research such as developing specific imaging markers and monitoring new therapy approaches. Methodological constraints such as image resolution of positron emission tomography (PET) and lack of suitable AD models have limited the feasibility of PET in mice. In this study, we evaluated a feasible protocol for PET imaging of Aβ in mouse brain with [11C]PiB and specific activities commonly used in human studies. In vivo mouse brain MRI for anatomical reference was acquired with a clinical 1.5 T system. A recently characterized APP/PS1 mouse was employed to measure Aβ at different disease stages in homozygous and hemizygous animals. We performed multi-modal cross-validations for the PET results with ex vivo and in vitro methodologies, including regional brain biodistribution, multi-label digital autoradiography, protein quantification with ELISA, fluorescence microscopy, semi-automated histological quantification and radioligand binding assays. Specific [11C]PiB uptake in individual brain regions with Aβ deposition was demonstrated and validated in all animals of the study cohort including homozygous AD animals as young as nine months. Corresponding to the extent of Aβ pathology, old homozygous AD animals (21 months) showed the highest uptake followed by old hemizygous (23 months) and young homozygous mice (9 months). In all AD age groups the cerebellum was shown to be suitable as an intracerebral reference region. PET results were cross-validated and consistent with all applied ex vivo and in vitro methodologies. The results confirm that the experimental setup for non-invasive [11C]PiB imaging of Aβ in the APP/PS1 mice provides a feasible, reproducible and robust protocol for small-animal Aβ imaging. It allows longitudinal imaging studies with follow-up periods of approximately one and a half years and provides a foundation for translational Alzheimer neuroimaging in transgenic mice.
Collapse
Affiliation(s)
- André Manook
- Nuklearmedizinische Klinik und Poliklinik, Klinikum rechts der Isar, Technische Universität München, Munich, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Gong YK, Winnik FM. Strategies in biomimetic surface engineering of nanoparticles for biomedical applications. NANOSCALE 2012; 4:360-8. [PMID: 22134705 DOI: 10.1039/c1nr11297j] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Engineered nanoparticles (NPs) play an increasingly important role in biomedical sciences and in nanomedicine. Yet, in spite of significant advances, it remains difficult to construct drug-loaded NPs with precisely defined therapeutic effects, in terms of release time and spatial targeting. The body is a highly complex system that imposes multiple physiological and cellular barriers to foreign objects. Upon injection in the blood stream or following oral administation, NPs have to bypass numerous barriers prior to reaching their intended target. A particularly successful design strategy consists in masking the NP to the biological environment by covering it with an outer surface mimicking the composition and functionality of the cell's external membrane. This review describes this biomimetic approach. First, we outline key features of the composition and function of the cell membrane. Then, we present recent developments in the fabrication of molecules that mimic biomolecules present on the cell membrane, such as proteins, peptides, and carbohydrates. We present effective strategies to link such bioactive molecules to the NPs surface and we highlight the power of this approach by presenting some exciting examples of biomimetically engineered NPs useful for multimodal diagnostics and for target-specific drug/gene delivery applications. Finally, critical directions for future research and applications of biomimetic NPs are suggested to the readers.
Collapse
Affiliation(s)
- Yong-kuan Gong
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry and Materials Science, Northwest University, Xi'an, 710069, PR China.
| | | |
Collapse
|
46
|
Kara F, Dongen ESV, Schliebs R, Buchem MAV, Groot HJMD, Alia A. Monitoring blood flow alterations in the Tg2576 mouse model of Alzheimer's disease by in vivo magnetic resonance angiography at 17.6 T. Neuroimage 2011; 60:958-66. [PMID: 22227054 DOI: 10.1016/j.neuroimage.2011.12.055] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 11/16/2011] [Accepted: 12/18/2011] [Indexed: 12/24/2022] Open
Abstract
Many neurodegenerative diseases including Alzheimer's disease are linked to abnormalities in the vascular system. In AD, the deposition of amyloid β (Aβ) peptide in the cerebral vessel walls, known as cerebral amyloid angiopathy (CAA) is frequently observed, leading to blood flow abnormalities. Visualization of the changes in vascular structure is important for early diagnosis and treatment. Blood vessels can be imaged non-invasively by magnetic resonance angiography (MRA). In this study we optimized high resolution MRA at 17.6 T to longitudinally monitor morphological changes in cerebral arteries in a Tg2576 mouse model, a widely used model of AD. Our results at 17.6 T show that MRA significantly benefits from the ultra-high magnetic field strength especially to visualize smaller vessels. Visual and quantitative analysis of MRA results revealed severe blood flow defects in large and medium sized arteries in Tg2576 mice. In particular blood flow defects were observed in the middle cerebral artery (MCA) and in the anterior communicating artery (AComA) in Tg2576 mice. Histological data show that Aβ levels in the vessel wall may be responsible for impaired cerebral blood flow, thereby contributing to the early progression of AD. To our knowledge this is the first ultra-high field MRA study monitoring blood flow alterations longitudinally in living Tg2576 mice, consequently providing a powerful tool to test new therapeutic intervention related to CAA in a mouse model of AD.
Collapse
Affiliation(s)
- F Kara
- SSNMR, Leiden Institute of Chemistry, Gorlaeus Laboratoria, Einsteinweg 55, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
47
|
Rotman M, Snoeks TJA, van der Weerd L. Pre-clinical optical imaging and MRI for drug development in Alzheimer's disease. DRUG DISCOVERY TODAY. TECHNOLOGIES 2011; 8:e117-e125. [PMID: 24990260 DOI: 10.1016/j.ddtec.2011.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Optical and magnetic resonance imaging have the potential to be complementary non-invasive imaging modalities. Yet without advances in imaging technologies and contrast agents both have short-comings that cannot be ignored. In this review we demonstrate the pre-clinical use of the two imaging techniques in Alzheimer's disease, including examples from recent applications and discuss what is needed to improve their applicability for drug discovery.:
Collapse
Affiliation(s)
- Maarten Rotman
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| | - Thomas J A Snoeks
- Department of Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
| | - Louise van der Weerd
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|