1
|
Sun X, Badachhape A, Bhandari P, Chin J, Annapragada A, Tanifum E. A dual target molecular magnetic resonance imaging probe for noninvasive profiling of pathologic alpha-synuclein and microgliosis in a mouse model of Parkinson's disease. Front Neurosci 2024; 18:1428736. [PMID: 39114484 PMCID: PMC11303179 DOI: 10.3389/fnins.2024.1428736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 07/11/2024] [Indexed: 08/10/2024] Open
Abstract
The pathogenesis of Parkinson's disease (PD) is characterized by progressive deposition of alpha-synuclein (α-syn) aggregates in dopaminergic neurons and neuroinflammation. Noninvasive in vivo imaging of α-syn aggregate accumulation and neuroinflammation can elicit the underlying mechanisms involved in disease progression and facilitate the development of effective treatment as well as disease diagnosis and prognosis. Here we present a novel approach to simultaneously profile α-syn aggregation and reactive microgliosis in vivo, by targeting oligomeric α-syn in cerebrospinal fluid with nanoparticle bearing a magnetic resonance imaging (MRI), contrast payload. In this proof-of-concept report we demonstrate, in vitro, that microglia and neuroblastoma cell lines internalize agglomerates formed by cross-linking the nanoparticles with oligomeric α-syn. Delayed in vivo MRI scans following intravenous administration of the nanoparticles in the M83 α-syn transgenic mouse line show statistically significant MR signal enhancement in test mice versus controls. The in vivo data were validated by ex-vivo immunohistochemical analysis which show strong correlation between in vivo MRI signal enhancement, Lewy pathology distribution, and microglia activity in the treated brain tissue. Furthermore, neuronal and microglial cells in brain tissue from treated mice display strong cytosolic signal originating from the nanoparticles, attributed to in vivo cell uptake of nanoparticle/oligomeric α-syn agglomerates.
Collapse
Affiliation(s)
- Xianwei Sun
- Department of Radiology, Baylor College of Medicine, Houston, TX, United States
| | - Andrew Badachhape
- Department of Radiology, Baylor College of Medicine, Houston, TX, United States
| | - Prajwal Bhandari
- Department of Radiology, Baylor College of Medicine, Houston, TX, United States
| | - Jeannie Chin
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Ananth Annapragada
- Department of Radiology, Baylor College of Medicine, Houston, TX, United States
- Department of Radiology, Texas Children’s Hospital, Houston, TX, United States
| | - Eric Tanifum
- Department of Radiology, Baylor College of Medicine, Houston, TX, United States
- Department of Radiology, Texas Children’s Hospital, Houston, TX, United States
| |
Collapse
|
2
|
Ahmed T. Lipid nanoparticle mediated small interfering RNA delivery as a potential therapy for Alzheimer's disease. Eur J Neurosci 2024; 59:2915-2954. [PMID: 38622050 DOI: 10.1111/ejn.16336] [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: 09/18/2023] [Revised: 02/21/2024] [Accepted: 03/14/2024] [Indexed: 04/17/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative condition that exhibits a gradual decline in cognitive function and is prevalent among a significant number of individuals globally. The use of small interfering RNA (siRNA) molecules in RNA interference (RNAi) presents a promising therapeutic strategy for AD. Lipid nanoparticles (LNPs) have been developed as a delivery vehicle for siRNA, which can selectively suppress target genes, by enhancing cellular uptake and safeguarding siRNA from degradation. Numerous research studies have exhibited the effectiveness of LNP-mediated siRNA delivery in reducing amyloid beta (Aβ) levels and enhancing cognitive function in animal models of AD. The feasibility of employing LNP-mediated siRNA delivery as a therapeutic approach for AD is emphasized by the encouraging outcomes reported in clinical studies for other medical conditions. The use of LNP-mediated siRNA delivery has emerged as a promising strategy to slow down or even reverse the progression of AD by targeting the synthesis of tau phosphorylation and other genes linked to the condition. Improvement of the delivery mechanism and determination of the most suitable siRNA targets are crucial for the efficacious management of AD. This review focuses on the delivery of siRNA through LNPs as a promising therapeutic strategy for AD, based on the available literature.
Collapse
Affiliation(s)
- Tanvir Ahmed
- Department of Pharmaceutical Sciences, North South University, Dhaka, Bangladesh
| |
Collapse
|
3
|
Parekh P, Badachhape AA, Tanifum EA, Annapragada AV, Ghaghada KB. Advances in nanoprobes for molecular MRI of Alzheimer's disease. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1946. [PMID: 38426638 PMCID: PMC10983770 DOI: 10.1002/wnan.1946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 01/11/2024] [Accepted: 01/30/2024] [Indexed: 03/02/2024]
Abstract
Alzheimer's disease is the most common cause of dementia and a leading cause of mortality in the elderly population. Diagnosis of Alzheimer's disease has traditionally relied on evaluation of clinical symptoms for cognitive impairment with a definitive diagnosis requiring post-mortem demonstration of neuropathology. However, advances in disease pathogenesis have revealed that patients exhibit Alzheimer's disease pathology several decades before the manifestation of clinical symptoms. Magnetic resonance imaging (MRI) plays an important role in the management of patients with Alzheimer's disease. The clinical availability of molecular MRI (mMRI) contrast agents can revolutionize the diagnosis of Alzheimer's disease. In this article, we review advances in nanoparticle contrast agents, also referred to as nanoprobes, for mMRI of Alzheimer's disease. This article is categorized under: Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease.
Collapse
Affiliation(s)
- Parag Parekh
- Department of Radiology, Baylor College of Medicine, Houston, Texas 77030
- Department of Radiology, Texas Children's Hospital, Houston, Texas 77030
| | - Andrew A. Badachhape
- Department of Radiology, Baylor College of Medicine, Houston, Texas 77030
- Department of Radiology, Texas Children's Hospital, Houston, Texas 77030
| | - Eric A. Tanifum
- Department of Radiology, Baylor College of Medicine, Houston, Texas 77030
- Department of Radiology, Texas Children's Hospital, Houston, Texas 77030
| | - Ananth V. Annapragada
- Department of Radiology, Baylor College of Medicine, Houston, Texas 77030
- Department of Radiology, Texas Children's Hospital, Houston, Texas 77030
| | - Ketan B. Ghaghada
- Department of Radiology, Baylor College of Medicine, Houston, Texas 77030
- Department of Radiology, Texas Children's Hospital, Houston, Texas 77030
| |
Collapse
|
4
|
Abidi SMS, Sharma C, Randhawa S, Shukla AK, Acharya A. A review on nanotechnological perspective of "the amyloid cascade hypothesis" for neurodegenerative diseases. Int J Biol Macromol 2023; 253:126821. [PMID: 37690655 DOI: 10.1016/j.ijbiomac.2023.126821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/03/2023] [Accepted: 09/07/2023] [Indexed: 09/12/2023]
Abstract
Neurodegenerative diseases (NDs) are characterized by progressive degeneration of neurons which deteriorates the brain functions. An early detection of the onset of NDs is utmost important, as it will provide the fast treatment strategies to prevent further progression of the disease. Conventionally, accurate diagnosis of the brain related disorders is difficult in their early phase. To solve this problem, nanotechnology based neurofunctional imaging and biomarker detection techniques have been developed which allows high specificity and sensitivity towards screening and diagnosis of NDs. Another challenge to treat the brain related disorders is to overcome the complex integrity of blood-brain-barrier (BBB) for the delivery of theranostic agents. Fortunately, utilization of nanomaterials has been pursued as promising strategy to address this challenge. Herein, we critically highlighted the recent improvements in the field of neurodiagnostic and therapeutic approaches involving innovative strategies for diagnosis, and inhibition of protein aggregates. We have provided particular emphasis on the use of nanotechnology which can push forward the blooming research growth in this field to win the battle against devastating NDs.
Collapse
Affiliation(s)
- Syed M S Abidi
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P. 176061, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Chandni Sharma
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P. 176061, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shiwani Randhawa
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P. 176061, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ashish K Shukla
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P. 176061, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amitabha Acharya
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P. 176061, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
5
|
Jo SL, Yang H, Lee HW, Hong EJ. Curcumae radix Reduces Endoplasmic Reticulum Stress in Mice with Chronic Neuroinflammation. Biomedicines 2023; 11:2107. [PMID: 37626603 PMCID: PMC10452873 DOI: 10.3390/biomedicines11082107] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 08/27/2023] Open
Abstract
Endoplasmic reticulum (ER) stress is a condition in which the ER protein-folding machinery is impaired, leading to the accumulation of improperly folded proteins and triggering an unfolded-protein response. Excessive ER stress causes cell death and contributes to the development of chronic diseases. Interestingly, there is a bidirectional relationship between ER stress and the nuclear factor-kappa B (NF-κB) pathway. Curcumin, a natural polyphenolic compound found in Curcumae radix, exerts its neuroprotective effects by regulating ER stress and inflammation. Therefore, investigating the potential protective and regulatory effects of curcumin on ER stress, inflammation, and neurodegeneration under chronic neuroinflammatory conditions is of great interest. Mice were pretreated with Curcumae radix extract (CRE) for 19 days and then treated with CRE plus lipopolysaccharide for 1 week. We monitored pro-inflammatory cytokine levels in the serum and ER stress-, inflammation-, and neurodegeneration-related markers in the mouse cerebrum and hippocampus using Western blotting and qRT-PCR. CRE reduced Interleukin-1 beta levels in the blood and brain of mice with lipopolysaccharide-induced chronic inflammation. CRE also suppressed the expression of markers related to the ER stress and NF-κB signaling pathways. The expression of neurodegeneration-related markers was reduced in the mouse cerebrum and hippocampus. CRE exerts neuroprotective effects under chronic inflammatory conditions via multifaceted anti-inflammatory and ER stress-pathway regulatory mechanisms.
Collapse
Affiliation(s)
- Seong-Lae Jo
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea;
| | - Hyun Yang
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea;
| | - Hye Won Lee
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea;
| | - Eui-Ju Hong
- College of Veterinary Medicine, Chungnam National University, Daejeon 34134, Republic of Korea;
| |
Collapse
|
6
|
Parekh P, Mu Q, Badachhape A, Bhavane R, Srivastava M, Devkota L, Sun X, Bhandari P, Eriksen JL, Tanifum E, Ghaghada K, Annapragada A. A surrogate marker for very early-stage tau pathology is detectable by molecular magnetic resonance imaging. Theranostics 2022; 12:5504-5521. [PMID: 35910789 PMCID: PMC9330526 DOI: 10.7150/thno.72258] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 07/02/2022] [Indexed: 01/30/2023] Open
Abstract
The abnormal phosphorylation of tau is a necessary precursor to the formation of tau fibrils, a marker of Alzheimer's disease. We hypothesize that hyperphosphorylative conditions may result in unique cell surface markers. We identify and demonstrate the utility of such surrogate markers to identify the hyperphosphorylative state. Methods: Cell SELEX was used to identify novel thioaptamers specifically binding hyperphosphorylative cells. Cell surface vimentin was identified as a potential binding target of the aptamer. Novel molecular magnetic resonance imaging (M-MRI) probes using these aptamers and a small molecule ligand to vimentin were used for in vivo detection of this pre-pathological state. Results: In a mouse model of pathological tau, we demonstrated in vivo visualization of the hyperphosphorylative state by M-MRI, enabling the identification at a pre-pathological stage of mice that develop frank tau pathology several months later. In vivo visualization of the hyperphosphorylative state by M-MRI was further validated in a second mouse model (APP/PS1) of Alzheimer's disease again identifying the mutants at a pre-pathological stage. Conclusions: M-MRI of the hyperphosphorylative state identifies future tau pathology and could enable extremely early-stage diagnosis of Alzheimer's disease, at a pre-patholgical stage.
Collapse
Affiliation(s)
| | - Qingshan Mu
- Baylor College of Medicine, Houston, TX, USA
| | | | | | | | | | - Xianwei Sun
- Baylor College of Medicine, Houston, TX, USA
| | | | | | - Eric Tanifum
- Texas Children's Hospital/Baylor College of Medicine, Houston, TX, USA
| | - Ketan Ghaghada
- Texas Children's Hospital/Baylor College of Medicine, Houston, TX, USA
| | - Ananth Annapragada
- Texas Children's Hospital/Baylor College of Medicine, Houston, TX, USA,✉ Corresponding author:
| |
Collapse
|
7
|
Delbreil P, Rabanel JM, Banquy X, Brambilla D. Therapeutic nanotechnologies for Alzheimer's disease: a critical analysis of recent trends and findings. Adv Drug Deliv Rev 2022; 187:114397. [PMID: 35738546 DOI: 10.1016/j.addr.2022.114397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 11/01/2022]
Abstract
Alzheimer's Disease (AD) is an irreversible neurodegenerative disease for which no disease modifying therapies are presently available. Besides the identification of pathological targets, AD presents numerous clinical and pharmacological challenges such as efficient active delivery to the central nervous system, cell targeting, and long-term dosing. Nanoparticles have been explored to overcome some of these challenges as drug delivery vehicles or drugs themselves. However, early promises have failed to materialize as no nanotechnology-based product has been able to reach the market and very few have moved past preclinical stages. In this review, we perform a critical analysis of the past decade's research on nanomedicine-based therapies for AD at the preclinical and clinical stages. The main obstacles to nanotechnology products and the most promising approaches were also identified, including renewed promise with gene editing, gene modulation, and vaccines.
Collapse
Affiliation(s)
- Philippe Delbreil
- Faculty of pharmacy, Université de Montréal, PO Box 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Jean-Michel Rabanel
- Faculty of pharmacy, Université de Montréal, PO Box 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Xavier Banquy
- Faculty of pharmacy, Université de Montréal, PO Box 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada
| | - Davide Brambilla
- Faculty of pharmacy, Université de Montréal, PO Box 6128, Succursale Centre-ville, Montréal, QC H3C 3J7, Canada.
| |
Collapse
|
8
|
Ghaghada KB, Bhavane R, Badachhape A, Tanifum E, Annapragada A. Nanoprobes for Computed Tomography and Magnetic Resonance Imaging in Atherosclerosis Research. Methods Mol Biol 2022; 2419:809-823. [PMID: 35238003 DOI: 10.1007/978-1-0716-1924-7_49] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Atheromatous lesions are characterized by intrusion into the vascular lumen, resulting in morphological changes to the blood compartment and into the vessel wall, resulting in characteristic molecular and cellular signatures in the solid tissue of the intima, tunica media, adventitia and surrounding tissue. Nanoprobes can be easily formulated to provide long blood-pool residence and molecular targeting, facilitating the imaging of atheromatous changes. Detection of nanoprobes can be accomplished by a variety of methods. We focus in this chapter on the use of cross-sectional imaging techniques, computed tomography (CT) and magnetic resonance imaging (MRI), that facilitate in vivo, noninvasive imaging of the vascular morphology and molecular/cellular signatures of the atheroma. The methods described are suitable for use in animal models, although versions of the probes are being readied for clinical trials, potentially facilitating clinical use in the future.
Collapse
Affiliation(s)
- Ketan B Ghaghada
- E.B. Singleton Department of Radiology, Texas Children's Hospital & Baylor College of Medicine, Houston, TX, USA
| | - Rohan Bhavane
- E.B. Singleton Department of Radiology, Texas Children's Hospital & Baylor College of Medicine, Houston, TX, USA
| | - Andrew Badachhape
- E.B. Singleton Department of Radiology, Texas Children's Hospital & Baylor College of Medicine, Houston, TX, USA
| | - Eric Tanifum
- E.B. Singleton Department of Radiology, Texas Children's Hospital & Baylor College of Medicine, Houston, TX, USA
| | - Ananth Annapragada
- E.B. Singleton Department of Radiology, Texas Children's Hospital & Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
9
|
Musielak M, Potoczny J, Boś-Liedke A, Kozak M. The Combination of Liposomes and Metallic Nanoparticles as Multifunctional Nanostructures in the Therapy and Medical Imaging-A Review. Int J Mol Sci 2021; 22:6229. [PMID: 34207682 PMCID: PMC8229649 DOI: 10.3390/ijms22126229] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/05/2021] [Accepted: 06/06/2021] [Indexed: 12/24/2022] Open
Abstract
Nanotechnology has introduced a new quality and has definitely developed the possibilities of treating and diagnosing various diseases. One of the scientists' interests is liposomes and metallic nanoparticles (LipoMNPs)-the combination of which has introduced new properties and applications. However, the field of creating hybrid nanostructures consisting of liposomes and metallic nanoparticles is relatively little understood. The purpose of this review was to compile the latest reports in the field of treatment and medical imaging using of LipoMNPs. The authors focused on presenting this issue in the direction of improving the used conventional treatment and imaging methods. Most of all, the nature of bio-interactions between nanostructures and cells is not sufficiently taken into account. As a result, overcoming the existing limitations in the implementation of such solutions in the clinic is difficult. We concluded that hybrid nanostructures are used in a very wide range, especially in the treatment of cancer and magnetic resonance imaging. There were also solutions that combine treatments with simultaneous imaging, creating a theragnostic approach. In the future, researchers should focus on the description of the biological interactions and the long-term effects of the nanostructures to use LipoMNPs in the treatment of patients.
Collapse
Affiliation(s)
- Marika Musielak
- Department of Electroradiology, Poznan University of Medical Sciences, 61-701 Poznań, Poland
- Radiobiology Laboratory, Department of Medical Physics, Greater Poland Cancer Centre, 61-866 Poznań, Poland
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, 61-614 Poznań, Poland; (A.B.-L.); (M.K.)
| | - Jakub Potoczny
- Heliodor Swiecicki Clinical Hospital in Poznan, 60-355 Poznań, Poland;
| | - Agnieszka Boś-Liedke
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, 61-614 Poznań, Poland; (A.B.-L.); (M.K.)
| | - Maciej Kozak
- Department of Macromolecular Physics, Faculty of Physics, Adam Mickiewicz University, 61-614 Poznań, Poland; (A.B.-L.); (M.K.)
| |
Collapse
|
10
|
Cano A, Turowski P, Ettcheto M, Duskey JT, Tosi G, Sánchez-López E, García ML, Camins A, Souto EB, Ruiz A, Marquié M, Boada M. Nanomedicine-based technologies and novel biomarkers for the diagnosis and treatment of Alzheimer's disease: from current to future challenges. J Nanobiotechnology 2021; 19:122. [PMID: 33926475 PMCID: PMC8086346 DOI: 10.1186/s12951-021-00864-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/17/2021] [Indexed: 02/07/2023] Open
Abstract
Increasing life expectancy has led to an aging population, which has consequently increased the prevalence of dementia. Alzheimer's disease (AD), the most common form of dementia worldwide, is estimated to make up 50-80% of all cases. AD cases are expected to reach 131 million by 2050, and this increasing prevalence will critically burden economies and health systems in the next decades. There is currently no treatment that can stop or reverse disease progression. In addition, the late diagnosis of AD constitutes a major obstacle to effective disease management. Therefore, improved diagnostic tools and new treatments for AD are urgently needed. In this review, we investigate and describe both well-established and recently discovered AD biomarkers that could potentially be used to detect AD at early stages and allow the monitoring of disease progression. Proteins such as NfL, MMPs, p-tau217, YKL-40, SNAP-25, VCAM-1, and Ng / BACE are some of the most promising biomarkers because of their successful use as diagnostic tools. In addition, we explore the most recent molecular strategies for an AD therapeutic approach and nanomedicine-based technologies, used to both target drugs to the brain and serve as devices for tracking disease progression diagnostic biomarkers. State-of-the-art nanoparticles, such as polymeric, lipid, and metal-based, are being widely investigated for their potential to improve the effectiveness of both conventional drugs and novel compounds for treating AD. The most recent studies on these nanodevices are deeply explained and discussed in this review.
Collapse
Affiliation(s)
- Amanda Cano
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain.
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.
- Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain.
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain.
| | - Patric Turowski
- UCL Institute of Ophthalmology, University College of London, London, UK
| | - Miren Ettcheto
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Jason Thomas Duskey
- Nanotech Lab, Te.Far.T.I, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Umberto Veronesi Foundation, 20121, Milano, Italy
| | - Giovanni Tosi
- Nanotech Lab, Te.Far.T.I, Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Sánchez-López
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Maria Luisa García
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), Barcelona, Spain
- Department of Pharmacy, Pharmaceutical Technology and Physical Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Barcelona, Spain
| | - Antonio Camins
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 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
| | - Agustín Ruiz
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Marta Marquié
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Mercè Boada
- Research Center and Memory Clinic, Fundació ACE. Institut Català de Neurociències Aplicades, International University of Catalunya (UIC), C/Marquès de Sentmenat, 57, 08029, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| |
Collapse
|
11
|
Gopalan D, Pandey A, Alex AT, Kalthur G, Pandey S, Udupa N, Mutalik S. Nanoconstructs as a versatile tool for detection and diagnosis of Alzheimer biomarkers. NANOTECHNOLOGY 2021; 32:142002. [PMID: 33238254 DOI: 10.1088/1361-6528/abcdcb] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The current review focuses towards the advancements made in the past decade in the field of nanotechnology for the early Alzheimer's disease (AD) diagnosis. This review includes the application of nanomaterials and nanosensors for the early detection of the main AD biomarkers (amyloid beta, phosphorylated tau, apolipoprotein E4 allele or APOE4, microRNAs, cholesterol, hydrogen peroxide etc) in biological fluids, to detect the biomarkers at a very low concentration ranging in pico, femto and even atto molar concentrations. The field of drug development has always aimed and is constantly working on developing disease modifying drugs, but these drugs will only succeed when given in the early disease stages. Thus, developing efficient diagnostic tools is of vital importance. Various nanomaterials such as liposomes; dendrimers; polymeric nanoparticles; coordination polymers; inorganic nanoparticles such as silica, manganese oxide, zinc oxide, iron oxide, super paramagnetic iron oxides; quantum dots, silver nanoparticles, gold nanoparticles, and carbon based nanostructures (carbon nanotubes, graphene oxide, nanofibres, nanodiamonds, carbon dots); Up-conversion nanoparticles; 2D nanomaterials; and radioactive nanoprobes have been used in constructing and improving efficiency of nano-sensors for AD biosensing at an early stage of diagnosis.
Collapse
Affiliation(s)
- Divya Gopalan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576 104, India
| | - Abhijeet Pandey
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576 104, India
| | - Angel Treasa Alex
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576 104, India
| | - Guruprasad Kalthur
- Department of Clinical Embryology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, Karnataka 576 104, India
| | - Sureshwar Pandey
- School of Pharmacy, Faculty of Medical Sciences, The university of West Indies, St. Augustine, Trinidad and Tobago, Jamaica
| | - Nayanabhirama Udupa
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576 104, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576 104, India
| |
Collapse
|
12
|
Oyarzún MP, Tapia-Arellano A, Cabrera P, Jara-Guajardo P, Kogan MJ. Plasmonic Nanoparticles as Optical Sensing Probes for the Detection of Alzheimer's Disease. SENSORS (BASEL, SWITZERLAND) 2021; 21:2067. [PMID: 33809416 PMCID: PMC7998661 DOI: 10.3390/s21062067] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/10/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD), considered a common type of dementia, is mainly characterized by a progressive loss of memory and cognitive functions. Although its cause is multifactorial, it has been associated with the accumulation of toxic aggregates of the amyloid-β peptide (Aβ) and neurofibrillary tangles (NFTs) of tau protein. At present, the development of highly sensitive, high cost-effective, and non-invasive diagnostic tools for AD remains a challenge. In the last decades, nanomaterials have emerged as an interesting and useful tool in nanomedicine for diagnostics and therapy. In particular, plasmonic nanoparticles are well-known to display unique optical properties derived from their localized surface plasmon resonance (LSPR), allowing their use as transducers in various sensing configurations and enhancing detection sensitivity. Herein, this review focuses on current advances in in vitro sensing techniques such as Surface-enhanced Raman scattering (SERS), Surface-enhanced fluorescence (SEF), colorimetric, and LSPR using plasmonic nanoparticles for improving the sensitivity in the detection of main biomarkers related to AD in body fluids. Additionally, we refer to the use of plasmonic nanoparticles for in vivo imaging studies in AD.
Collapse
Affiliation(s)
- María Paz Oyarzún
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| | - Andreas Tapia-Arellano
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| | - Pablo Cabrera
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| | - Pedro Jara-Guajardo
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| | - Marcelo J. Kogan
- Departamento de Química Farmacológica y Toxicológica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Dr. Carlos Lorca Tobar 964, Independencia, 8380000 Santiago, Chile; (M.P.O.); (A.T.-A.); (P.C.); (P.J.-G.)
- Advanced Center for Chronic Diseases (ACCDIS), Sergio Livingstone #1007, Independencia, 8380492 Santiago, Chile
| |
Collapse
|
13
|
Zhang L, Sun H, Chen Y, Wei M, Lee J, Li F, Ling D. Functional nanoassemblies for the diagnosis and therapy of Alzheimer's diseases. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1696. [PMID: 33463089 DOI: 10.1002/wnan.1696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/23/2020] [Accepted: 12/26/2020] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease that affects populations around the world. Many therapeutics have been investigated for AD diagnosis and/or therapy, but the efficacy is largely limited by the poor bioavailability of drugs and by the presence of the blood-brain barrier. Recently, the development of nanomedicines enables efficient drug delivery to the brain, but the complex pathological mechanism of AD prevents them from successful treatment. As a type of advanced nanomedicine, multifunctional nanoassemblies self-assembled from nanoscale imaging or therapeutic agents can simultaneously target multiple pathological factors, showing great potential in the diagnosis and therapy of AD. To help readers better understand this emerging field, in this review, we first introduce the pathological mechanisms and the potential drug candidates of AD, as well as the design strategies of nanoassemblies for improving AD targeting efficiency. Moreover, the progress of dynamic nanoassemblies that can diagnose and/or treat AD in response to the endogenous or exogenous stimuli will be described. Finally, we conclude with our perspectives on the future development in this field. The objective of this review is to outline the latest progress of using nanoassemblies to overcome the complex pathological environment of AD for improved diagnosis and therapy, in hopes of accelerating the future development of intelligent AD nanomedicines. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Diagnostic Tools > in vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Lingxiao Zhang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Heng Sun
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ying Chen
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Min Wei
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jiyoung Lee
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Fangyuan Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
| | - Daishun Ling
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Biomedical Engineering of the Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, China
- National Center for Translational Medicine, Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
14
|
Curcumin and Its Derivatives as Theranostic Agents in Alzheimer's Disease: The Implication of Nanotechnology. Int J Mol Sci 2020; 22:ijms22010196. [PMID: 33375513 PMCID: PMC7795367 DOI: 10.3390/ijms22010196] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/15/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Curcumin is a polyphenolic natural compound with diverse and attractive biological properties, which may prevent or ameliorate pathological processes underlying age-related cognitive decline, Alzheimer's disease (AD), dementia, or mode disorders. AD is a chronic neurodegenerative disorder that is known as one of the rapidly growing diseases, especially in the elderly population. Moreover, being the eminent cause of dementia, posing problems for families, societies as well a severe burden on the economy. There are no effective drugs to cure AD. Although curcumin and its derivatives have shown properties that can be considered useful in inhibiting the hallmarks of AD, however, they have low bioavailability. Furthermore, to combat diagnostic and therapeutic limitations, various nanoformulations have also been recognized as theranostic agents that can also enhance the pharmacokinetic properties of curcumin and other bioactive compounds. Nanocarriers have shown beneficial properties to deliver curcumin and other nutritional compounds against the blood-brain barrier to efficiently distribute them in the brain. This review spotlights the role and effectiveness of curcumin and its derivatives in AD. Besides, the gut metabolism of curcumin and the effects of nanoparticles and their possible activity as diagnostic and therapeutic agents in AD also discussed.
Collapse
|
15
|
Thangudu S, Cheng FY, Su CH. Advancements in the Blood-Brain Barrier Penetrating Nanoplatforms for Brain Related Disease Diagnostics and Therapeutic Applications. Polymers (Basel) 2020; 12:E3055. [PMID: 33419339 PMCID: PMC7766280 DOI: 10.3390/polym12123055] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022] Open
Abstract
Noninvasive treatments to treat the brain-related disorders have been paying more significant attention and it is an emerging topic. However, overcoming the blood brain barrier (BBB) is a key obstacle to most of the therapeutic drugs to enter into the brain tissue, which significantly results in lower accumulation of therapeutic drugs in the brain. Thus, administering the large quantity/doses of drugs raises more concerns of adverse side effects. Nanoparticle (NP)-mediated drug delivery systems are seen as potential means of enhancing drug transport across the BBB and to targeted brain tissue. These systems offer more accumulation of therapeutic drugs at the tumor site and prolong circulation time in the blood. In this review, we summarize the current knowledge and advancements on various nanoplatforms (NF) and discusses the use of nanoparticles for successful cross of BBB to treat the brain-related disorders such as brain tumors, Alzheimer's disease, Parkinson's disease, and stroke.
Collapse
Affiliation(s)
- Suresh Thangudu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
| | - Fong-Yu Cheng
- Department of Chemistry, Chinese Culture University, Taipei 111, Taiwan
| | - Chia-Hao Su
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming University, Taipei 112, Taiwan
| |
Collapse
|
16
|
Badachhape AA, Working PK, Srivastava M, Bhandari P, Stupin IV, Devkota L, Tanifum EA, Annapragada AV, Ghaghada KB. Pre-clinical dose-ranging efficacy, pharmacokinetics, tissue biodistribution, and toxicity of a targeted contrast agent for MRI of amyloid deposition in Alzheimer's disease. Sci Rep 2020; 10:16185. [PMID: 32999398 PMCID: PMC7527957 DOI: 10.1038/s41598-020-73233-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 09/09/2020] [Indexed: 01/30/2023] Open
Abstract
In these preclinical studies, we describe ADx-001, an Aβ-targeted liposomal macrocyclic gadolinium (Gd) imaging agent, for MRI of amyloid plaques. The targeting moiety is a novel lipid-PEG conjugated styryl-pyrimidine. An MRI-based contrast agent such as ADx-001 is attractive because of the lack of radioactivity, ease of distribution, long shelf life, and the prevalence of MRI scanners. Dose-ranging efficacy studies were performed on a 1 T MRI scanner using a transgenic APP/PSEN1 mouse model of Alzheimer's disease. ADx-001 was tested at 0.10, 0.15, and 0.20 mmol Gd/kg. Gold standard post-mortem amyloid immunostaining was used for the determination of sensitivity and specificity. ADx-001 toxicity was evaluated in rats and monkeys at doses up to 0.30 mmol Gd/kg. ADx-001 pharmacokinetics were determined in monkeys and its tissue distribution was evaluated in rats. ADx-001-enhanced MRI demonstrated significantly higher (p < 0.05) brain signal enhancement in transgenic mice relative to wild type mice at all dose levels. ADx-001 demonstrated high sensitivity at 0.20 and 0.15 mmol Gd/kg and excellent specificity at all dose levels for in vivo imaging of β amyloid plaques. ADx-001 was well tolerated in rats and monkeys and exhibited the slow clearance from circulation and tissue biodistribution typical of PEGylated nanoparticles.
Collapse
Affiliation(s)
- Andrew A Badachhape
- Department of Radiology, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - Mayank Srivastava
- The Singleton Department of Pediatric Radiology, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Prajwal Bhandari
- Department of Radiology, Baylor College of Medicine, 1102 Bates Street, Suite 850, Houston, TX, 77030, USA
| | - Igor V Stupin
- The Singleton Department of Pediatric Radiology, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Laxman Devkota
- Department of Pediatrics-Oncology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Eric A Tanifum
- The Singleton Department of Pediatric Radiology, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Ananth V Annapragada
- The Singleton Department of Pediatric Radiology, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Ketan B Ghaghada
- The Singleton Department of Pediatric Radiology, Texas Children's Hospital, Houston, TX, 77030, USA.
- Department of Radiology, Baylor College of Medicine, 1102 Bates Street, Suite 850, Houston, TX, 77030, USA.
- Edward B. Singleton Department of Radiology, Texas Children's Hospital, Houston, USA.
| |
Collapse
|
17
|
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
|
18
|
Derakhshankhah H, Sajadimajd S, Jafari S, Izadi Z, Sarvari S, Sharifi M, Falahati M, Moakedi F, Muganda WCA, Müller M, Raoufi M, Presley JF. Novel therapeutic strategies for Alzheimer's disease: Implications from cell-based therapy and nanotherapy. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 24:102149. [PMID: 31927133 DOI: 10.1016/j.nano.2020.102149] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/28/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is a multifactorial neurodegenerative disease which leads to progressive dysfunction of cognition, memory and learning in elderly people. Common therapeutic agents are not only inadequate to suppress the progression of AD pathogenesis but also produce deleterious side effects; hence, development of alternative therapies is required to specifically suppress complications of AD. The current review provides a commentary on conventional as well as novel therapeutic approaches with an emphasis on stem cell and nano-based therapies for improvement and management of AD pathogenesis. According to our overview of the current literature, AD is a multi-factorial disorder with various pathogenic trajectories; hence, a multifunctional strategy to create effective neuroprotective agents is required to treat this disorder.
Collapse
Affiliation(s)
- Hossein Derakhshankhah
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soraya Sajadimajd
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Samira Jafari
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zhila Izadi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sajad Sarvari
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Majid Sharifi
- Department of Nanotechnology, Faculty of Advance Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advance Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Faezeh Moakedi
- Health Science Center, West Virginia University, Morgantown, USA
| | | | - Mareike Müller
- Physical Chemistry I and Research Center of Micro and Nanochemistry (Cμ), University of Siegen, Siegen, Germany
| | - Mohammad Raoufi
- Physical Chemistry I and Research Center of Micro and Nanochemistry (Cμ), University of Siegen, Siegen, Germany; Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - John F Presley
- Department of Anatomy and Cell Biology, McGill University, Montreal, Canada.
| |
Collapse
|
19
|
Hettiarachchi SD, Zhou Y, Seven E, Lakshmana MK, Kaushik AK, Chand HS, Leblanc RM. Nanoparticle-mediated approaches for Alzheimer's disease pathogenesis, diagnosis, and therapeutics. J Control Release 2019; 314:125-140. [PMID: 31647979 DOI: 10.1016/j.jconrel.2019.10.034] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 10/16/2019] [Accepted: 10/18/2019] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is an irreversible and progressive neurodegenerative disorder manifested by memory loss and cognitive impairment. Deposition of the amyloid β plaques has been identified as the most common AD pathology; however, the excessive accumulation of phosphorylated or total tau proteins, reactive oxygen species, and higher acetylcholinesterase activity are also strongly associated with Alzheimer's dementia. Several therapeutic approaches targeting these pathogenic mechanisms have failed in clinical or preclinical trials, partly due to the limited bioavailability, poor cell, and blood-brain barrier penetration, and low drug half-life of current regimens. The nanoparticles (NPs)-mediated drug delivery systems improve drug solubility and bioavailability, thus renders as superior alternatives. Moreover, NPs-mediated approaches facilitate multiple drug loading and targeted drug delivery, thereby increasing drug efficacy. However, certain NPs can cause acute toxicity damaging cellular and tissue architecture, therefore, NP material should be carefully selected. In this review, we summarize the recent NPs-mediated studies that exploit various pathologic mechanisms of AD by labeling, identifying, and treating the affected brain pathologies. The disadvantages of the select NP-based deliveries and the future aspects will also be discussed.
Collapse
Affiliation(s)
- Sajini D Hettiarachchi
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Yiqun Zhou
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Elif Seven
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| | - Madepalli K Lakshmana
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
| | - Ajeet K Kaushik
- Department of Natural Sciences, Division of Sciences, Arts & Mathematics, Florida Polytechnic University, Lakeland, FL 33805-8531, USA
| | - Hitendra S Chand
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, 11200 SW 8th Street, Miami, FL 33199, USA
| | - Roger M Leblanc
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL, 33146, USA
| |
Collapse
|
20
|
Kevadiya BD, Ottemann BM, Thomas MB, Mukadam I, Nigam S, McMillan J, Gorantla S, Bronich TK, Edagwa B, Gendelman HE. Neurotheranostics as personalized medicines. Adv Drug Deliv Rev 2019; 148:252-289. [PMID: 30421721 PMCID: PMC6486471 DOI: 10.1016/j.addr.2018.10.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 12/16/2022]
Abstract
The discipline of neurotheranostics was forged to improve diagnostic and therapeutic clinical outcomes for neurological disorders. Research was facilitated, in largest measure, by the creation of pharmacologically effective multimodal pharmaceutical formulations. Deployment of neurotheranostic agents could revolutionize staging and improve nervous system disease therapeutic outcomes. However, obstacles in formulation design, drug loading and payload delivery still remain. These will certainly be aided by multidisciplinary basic research and clinical teams with pharmacology, nanotechnology, neuroscience and pharmaceutic expertise. When successful the end results will provide "optimal" therapeutic delivery platforms. The current report reviews an extensive body of knowledge of the natural history, epidemiology, pathogenesis and therapeutics of neurologic disease with an eye on how, when and under what circumstances neurotheranostics will soon be used as personalized medicines for a broad range of neurodegenerative, neuroinflammatory and neuroinfectious diseases.
Collapse
Affiliation(s)
- Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Brendan M Ottemann
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Midhun Ben Thomas
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Insiya Mukadam
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Saumya Nigam
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - JoEllyn McMillan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Santhi Gorantla
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Benson Edagwa
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, USA; Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
21
|
|
22
|
Ovais M, Zia N, Ahmad I, Khalil AT, Raza A, Ayaz M, Sadiq A, Ullah F, Shinwari ZK. Phyto-Therapeutic and Nanomedicinal Approaches to Cure Alzheimer's Disease: Present Status and Future Opportunities. Front Aging Neurosci 2018; 10:284. [PMID: 30405389 PMCID: PMC6205985 DOI: 10.3389/fnagi.2018.00284] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 08/30/2018] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by cognitive inability manifested due to the accumulation of β-amyloid, formation of hyper phosphorylated neurofibrillary tangles, and a malfunctioned cholinergic system. The degeneration integrity of the neuronal network can appear long after the onset of the disease. Nanotechnology-based interventions have opened an exciting area via theranostics of AD in terms of tailored nanomedicine, which are able to target and deliver drugs across the blood-brain barrier (BBB). The exciting interface existing between medicinal plants and nanotechnology is an emerging marvel in medicine, which has delivered promising results in the treatment of AD. In order to assess the potential applications of the medicinal plants, their derived components, and various nanomedicinal approaches, a review of literature was deemed as necessary. In the present review, numerous phytochemicals and various feats in nanomedicine for the treatment of AD have been discussed mechanistically for the first time. Furthermore, recent trends in nanotechnology such as green synthesis of metal nanoparticles with reference to the treatment of AD have been elaborated. Foreseeing the recent progress, we hope that the interface of medicinal plants and nanotechnology will lead to highly effective theranostic strategies for the treatment of AD in the near future.
Collapse
Affiliation(s)
- Muhammad Ovais
- Department of Biotechnology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- National Institute of Lasers and Optronics, Pakistan Atomic Energy Commission, Islamabad, Pakistan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Nashmia Zia
- National Institute of Lasers and Optronics, Pakistan Atomic Energy Commission, Islamabad, Pakistan
- Department of Pharmacy, University of Peshawar, Peshawar, Pakistan
| | - Irshad Ahmad
- Department of Life Sciences, King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
| | - Ali Talha Khalil
- Department of Eastern Medicine and Surgery, Qarshi University, Lahore, Pakistan
| | - Abida Raza
- National Institute of Lasers and Optronics, Pakistan Atomic Energy Commission, Islamabad, Pakistan
| | - Muhammad Ayaz
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Abdul Sadiq
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
- Department of Life Sciences and Chemistry, Faculty of Health, Jacobs University Bremen, Bremen, Germany
| | - Farhat Ullah
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Zabta Khan Shinwari
- Department of Biotechnology, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan
- Department of Eastern Medicine and Surgery, Qarshi University, Lahore, Pakistan
- Pakistan Academy of Sciences, Islamabad, Pakistan
| |
Collapse
|
23
|
Woodside DG, Tanifum EA, Ghaghada KB, Biediger RJ, Caivano AR, Starosolski ZA, Khounlo S, Bhayana S, Abbasi S, Craft JW, Maxwell DS, Patel C, Stupin IV, Bakthavatsalam D, Market RV, Willerson JT, Dixon RAF, Vanderslice P, Annapragada AV. Magnetic Resonance Imaging of Atherosclerotic Plaque at Clinically Relevant Field Strengths (1T) by Targeting the Integrin α4β1. Sci Rep 2018; 8:3733. [PMID: 29487319 PMCID: PMC5829217 DOI: 10.1038/s41598-018-21893-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 02/12/2018] [Indexed: 02/07/2023] Open
Abstract
Inflammation drives the degradation of atherosclerotic plaque, yet there are no non-invasive techniques available for imaging overall inflammation in atherosclerotic plaques, especially in the coronary arteries. To address this, we have developed a clinically relevant system to image overall inflammatory cell burden in plaque. Here, we describe a targeted contrast agent (THI0567-targeted liposomal-Gd) that is suitable for magnetic resonance (MR) imaging and binds with high affinity and selectivity to the integrin α4β1(very late antigen-4, VLA-4), a key integrin involved in recruiting inflammatory cells to atherosclerotic plaques. This liposomal contrast agent has a high T1 relaxivity (~2 × 105 mM-1s-1 on a particle basis) resulting in the ability to image liposomes at a clinically relevant MR field strength. We were able to visualize atherosclerotic plaques in various regions of the aorta in atherosclerosis-prone ApoE-/- mice on a 1 Tesla small animal MRI scanner. These enhanced signals corresponded to the accumulation of monocyte/macrophages in the subendothelial layer of atherosclerotic plaques in vivo, whereas non-targeted liposomal nanoparticles did not demonstrate comparable signal enhancement. An inflammatory cell-targeted method that has the specificity and sensitivity to measure the inflammatory burden of a plaque could be used to noninvasively identify patients at risk of an acute ischemic event.
Collapse
Affiliation(s)
- Darren G Woodside
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA.
| | - Eric A Tanifum
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Ketan B Ghaghada
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Ronald J Biediger
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Amy R Caivano
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Zbigniew A Starosolski
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Sayadeth Khounlo
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Saakshi Bhayana
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Shahrzad Abbasi
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - John W Craft
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA.,Department of Biology and Chemistry, University of Houston, 4800 Calhoun Road, Houston, Texas, 77004, USA
| | - David S Maxwell
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas, 77030, USA.,Department of Institutional Analytics and Informatics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chandreshkumar Patel
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | - Igor V Stupin
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA
| | | | - Robert V Market
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - James T Willerson
- Division of Cardiology Research, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Richard A F Dixon
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Peter Vanderslice
- Department of Molecular Cardiology, Texas Heart Institute, 6770 Bertner Avenue, Houston, Texas, 77030, USA
| | - Ananth V Annapragada
- Department of Pediatric Radiology, Texas Children's Hospital, 6621 Fannin Street, Houston, Texas, 77030, USA.
| |
Collapse
|
24
|
Kokuryo D. [3. Researches of Drug Delivery System and Theranostics Using Pre-clinical MRI]. Nihon Hoshasen Gijutsu Gakkai Zasshi 2018; 74:76-83. [PMID: 29353839 DOI: 10.6009/jjrt.2018_jsrt_74.1.76] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
25
|
Nasr SH, Kouyoumdjian H, Mallett C, Ramadan S, Zhu DC, Shapiro EM, Huang X. Detection of β-Amyloid by Sialic Acid Coated Bovine Serum Albumin Magnetic Nanoparticles in a Mouse Model of Alzheimer's Disease. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:10.1002/smll.201701828. [PMID: 29134771 PMCID: PMC5773361 DOI: 10.1002/smll.201701828] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 09/27/2017] [Indexed: 05/04/2023]
Abstract
The accumulation and formation of β-amyloid (Aβ) plaques in the brain are distinctive pathological hallmarks of Alzheimer's disease (AD). Designing nanoparticle (NP) contrast agents capable of binding with Aβ highly selectively can potentially facilitate early detection of AD. However, a significant obstacle is the blood brain barrier (BBB), which can preclude the entrance of NPs into the brain for Aβ binding. In this work, bovine serum albumin (BSA) coated NPs are decorated with sialic acid (NP-BSAx -Sia) to overcome the challenges in Aβ imaging in vivo. The NP-BSAx -Sia is biocompatible with high magnetic relaxivities, suggesting that they are suitable contrast agents for magnetic resonance imaging (MRI). The NP-BSAx -Sia binds with Aβ in a sialic acid dependent manner with high selectivities toward Aβ deposited on brains and cross the BBB in an in vitro model. The abilities of these NPs to detect Aβ in vivo in human AD transgenic mice by MRI are evaluated without the need to coinject mannitol to increase BBB permeability. T2 *-weighted MRI shows that Aβ plaques in mouse brains can be detected as aided by NP-BSAx -Sia, which is confirmed by histological analysis. Thus, NP-BSAx -Sia is a promising new tool for noninvasive in vivo detection of Aβ plaques.
Collapse
Affiliation(s)
- Seyedmehdi Hossaini Nasr
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
| | - Hovig Kouyoumdjian
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
- Department of Chemistry, York University, 4700 Keele Street, Chemistry Building 350, Toronto, ON, M3J 1P3 T, Canada
| | - Christiane Mallett
- Department of Radiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Sherif Ramadan
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
- Chemistry Department, Faculty of Science, Benha University, Benha, Qaliobiya, 13518, Egypt
| | - David C Zhu
- Department of Radiology, Michigan State University, East Lansing, MI, 48824, USA
| | - Erik M Shapiro
- Department of Radiology, Michigan State University, East Lansing, MI, 48824, USA
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, 578 S. Shaw Lane, East Lansing, MI, 48824, USA
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
26
|
Challenges and opportunities in developing targeted molecular imaging to determine inner ear defects of sensorineural hearing loss. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2017; 14:397-404. [PMID: 29074310 DOI: 10.1016/j.nano.2017.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 10/05/2017] [Accepted: 10/13/2017] [Indexed: 12/30/2022]
Abstract
The development of inner ear gene carriers and delivery systems has enabled genetic defects to be repaired and hearing to be restored in mouse models. Today, promising advances in translational therapies provide confidence that targeted molecular therapy for inner ear diseases will be developed. Unfortunately, the currently available non-invasive modalities, such as Computerized Tomography scan or Magnetic Resonance Imaging provide insufficient resolution to identify most pathologies of the human inner ear, even when the current generation of contrast agents is utilized. The development of targeted contrast agents may play a critical role in determining the cause of, and treatment for, sensorineural hearing loss. Such agents should be able to pass through the cochlea barriers, possess minimal cytotoxicity, and easily conjugate to a targeting agent, without distorting the anatomic details. This review focuses on a series of contrast agents which may fit these criteria for potential clinical application.
Collapse
|
27
|
Hajipour MJ, Santoso MR, Rezaee F, Aghaverdi H, Mahmoudi M, Perry G. Advances in Alzheimer's Diagnosis and Therapy: The Implications of Nanotechnology. Trends Biotechnol 2017; 35:937-953. [PMID: 28666544 DOI: 10.1016/j.tibtech.2017.06.002] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/12/2017] [Accepted: 06/06/2017] [Indexed: 01/03/2023]
Abstract
Alzheimer's disease (AD) is a type of dementia that causes major issues for patients' memory, thinking, and behavior. Despite efforts to advance AD diagnostic and therapeutic tools, AD remains incurable due to its complex and multifactorial nature and lack of effective diagnostics/therapeutics. Nanoparticles (NPs) have demonstrated the potential to overcome the challenges and limitations associated with traditional diagnostics/therapeutics. Nanotechnology is now offering new tools and insights to advance our understanding of AD and eventually may offer new hope to AD patients. Here, we review the key roles of nanotechnologies in the recent literature, in both diagnostic and therapeutic aspects of AD, and discuss how these achievements may improve patient prognosis and quality of life.
Collapse
Affiliation(s)
- Mohammad Javad Hajipour
- Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr 75147, Iran; Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran 13169-43551, Iran
| | - Michelle R Santoso
- Division of Cardiovascular Medicine, Stanford University, Stanford, CA 94305, USA
| | - Farhad Rezaee
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Haniyeh Aghaverdi
- Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Morteza Mahmoudi
- Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 13169-43551, Iran.
| | - George Perry
- Neurosciences Institute and Department of Biology, College of Sciences, University of Texas at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
28
|
Panahi Y, Farshbaf M, Mohammadhosseini M, Mirahadi M, Khalilov R, Saghfi S, Akbarzadeh A. Recent advances on liposomal nanoparticles: synthesis, characterization and biomedical applications. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 45:788-799. [DOI: 10.1080/21691401.2017.1282496] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Yunes Panahi
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Masoud Farshbaf
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mozhdeh Mirahadi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rovshan Khalilov
- Department of Plant Physiology, Faculty of Biology, Baku State University, Baku, Azerbaijan
- Joint Ukrainian-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems, Drohobych Ukraine & Baku, Azerbaijan
| | - Siamak Saghfi
- Department of Plant Physiology, Faculty of Biology, Baku State University, Baku, Azerbaijan
- Joint Ukrainian-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems, Drohobych Ukraine & Baku, Azerbaijan
| | - Abolfazl Akbarzadeh
- Joint Ukrainian-Azerbaijan International Research and Education Center of Nanobiotechnology and Functional Nanosystems, Drohobych Ukraine & Baku, Azerbaijan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Universal Scientific Education and Research Network (USERN), Tabriz, Iran
| |
Collapse
|
29
|
Tafoya MA, Madi S, Sillerud LO. Superparamagnetic nanoparticle-enhanced MRI of Alzheimer's disease plaques and activated microglia in 3X transgenic mouse brains: Contrast optimization. J Magn Reson Imaging 2016; 46:574-588. [PMID: 27875002 DOI: 10.1002/jmri.25563] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 11/01/2016] [Indexed: 11/05/2022] Open
Abstract
PURPOSE To optimize magnetic resonance imaging (MRI) of antibody-conjugated superparamagnetic nanoparticles for detecting amyloid-β plaques and activated microglia in a 3X transgenic mouse model of Alzheimer's disease. MATERIALS AND METHODS Ten 3X Tg mice were fed either chow or chow containing 100 ppm resveratrol. Four brains, selected from animals injected with either anti-amyloid targeted superparamagnetic iron oxide nanoparticles, or anti-Iba-1-conjugated FePt-nanoparticles, were excised, fixed with formalin, and placed in Fomblin for ex vivo MRI (11.7T) using multislice-multiecho, multiple gradient echo, rapid acquisition with relaxation enhancement, and susceptibility-weighted imaging (SWI). Aβ plaques and areas of neuroinflammation appeared as hypointense regions whose number, location, and Z-score were measured as a function of sequence type and echo time. RESULTS The MR contrast was due to the shortening of the transverse relaxation time of the plaque-adjacent tissue water. A theoretical analysis of this effect showed that the echo time was the primary determinant of plaque contrast and was used to optimize Z-scores. The Z-scores of the detected lesions varied from 21 to 34 as the echo times varied from 4 to 25 msec, with SWI providing the highest Z-score and number of detected lesions. Computation of the entire plaque and activated microglial distributions in 3D showed that resveratrol treatment led to a reduction of ∼24-fold of Aβ plaque density and ∼4-fold in microglial activation. CONCLUSION Optimized MRI of antibody-conjugated superparamagnetic nanoparticles served to reveal the 3D distributions of both Aβ plaques and activated microglia and to measure the effects of drug treatments in this 3X Tg model. LEVEL OF EVIDENCE 1 Technical Efficacy: Stage 2 J. MAGN. RESON. IMAGING 2017;46:574-588.
Collapse
Affiliation(s)
- Marissa A Tafoya
- UNM BRaIN Center, Department of Neurology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | | | - Laurel O Sillerud
- UNM BRaIN Center, Department of Neurology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| |
Collapse
|