1
|
Refaat A, Thomas P, Zhang W, Esser L, Tong Z, Beer M, Mawdsley D, Thirkettle-Watts D, Shields KA, Nicolazzo JA, Voelcker NH. An In Vitro-In Vivo Comparative Study Using Highly Sensitive Radioisotopic Assays to Assess the Predictive Power of Emerging Blood-Brain Barrier Models. SMALL METHODS 2025; 9:e2401400. [PMID: 39663724 DOI: 10.1002/smtd.202401400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/07/2024] [Indexed: 12/13/2024]
Abstract
Microfluidic BBB-on-a-chip models (μBBB) aim to recapitulate the organotypic features of the human BBB with great potential to model CNS diseases and advance CNS therapeutics. Nevertheless, their predictive capacity for drug uptake into the brain remains uncertain due to limited evaluation with only a small number of model drugs. Here, the in vivo brain uptake of a panel of nine radiolabeled compounds is evaluated in Swiss-outbred mice following a single intravenously administered dose and compared against results from the microfluidic μBBB platform and the conventional Transwell BBB model. Radioisotopic measurements are employed to calculate brain-to-plasma concentration ratios (B/P) of the compounds both in vivo and in vitro. The in vitro-in vivo correlation plots of the B/P ratios revealed a strong positive correlation (r = 0.8081, R2 = 0.6530) for the μBBB, suggesting a high degree of predictive ability for drug permeability into the brain. In contrast, the Transwell assay showed a weaker in vitro-in vivo correlation (r = 0.6467, R2 = 0.4182). Finally, brain uptake of radiolabeled, brain-targeted, angiopep2-conjugated nanoparticles (ANG2-NP) is assessed in the μBBB and results mirrored the in vivo uptake, while the Transwell model failed to resolve the differences between the targeted and non-targeted NPs.
Collapse
Affiliation(s)
- Ahmed Refaat
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
| | - Patrick Thomas
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
- Department of Defence, Defence Science and Technology Group, 506 Lorimer Street, Port Melbourne, VIC, 3207, Australia
| | - Weisen Zhang
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
- Commonwealth Scientific and Industrial Research Organization, Research Way, Melbourne, VIC, 3168, Australia
| | - Lars Esser
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
- Commonwealth Scientific and Industrial Research Organization, Research Way, Melbourne, VIC, 3168, Australia
| | - Ziqiu Tong
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
| | - Michael Beer
- Department of Defence, Defence Science and Technology Group, 506 Lorimer Street, Port Melbourne, VIC, 3207, Australia
| | - David Mawdsley
- Department of Defence, Defence Science and Technology Group, 506 Lorimer Street, Port Melbourne, VIC, 3207, Australia
| | - David Thirkettle-Watts
- Department of Defence, Defence Science and Technology Group, 506 Lorimer Street, Port Melbourne, VIC, 3207, Australia
| | - Katherine A Shields
- Department of Defence, Defence Science and Technology Group, 506 Lorimer Street, Port Melbourne, VIC, 3207, Australia
| | - Joseph A Nicolazzo
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
| | - Nicolas H Voelcker
- Department of Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Melbourne, VIC, 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Rd, Melbourne, VIC, 3168, Australia
- Department of Materials Science & Engineering, Faculty of Engineering, Monash University, 14 Alliance Ln, Melbourne, VIC, 3168, Australia
| |
Collapse
|
2
|
Saikia J, Sarkar M, Ramakrishnan V. Factors affecting the physical stability of peptide self-assembly in neurodegenerative disorders. Neuropeptides 2025; 111:102517. [PMID: 40112745 DOI: 10.1016/j.npep.2025.102517] [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: 11/08/2024] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 03/22/2025]
Abstract
Biological systems comprise of diverse biomolecules, including proteins, nucleic acids, lipids, and carbohydrates. Peptides, which are short chains of amino acids, exhibit unique properties when assembled to nano-level architectures. Self-assembling peptides possess a remarkable ability to organize into structured aggregates such as nanofibers, nanotubes, nanoribbons, and nanovesicles. These intricate structures are linked to neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Prion disease, Huntington's disease, and type II diabetes. Peptide nano assembly can be guided by external stimuli, such as temperature, pH, ultrasound, electric and magnetic fields. In this review, the discussion will be centred around the various factors that influence the self-assembly of peptides alongside therapeutic interventions that align with the fundamental principles of thermodynamics and kinetics to modulate the aggregation characteristics of peptide self-assembly.
Collapse
Affiliation(s)
- Jahnu Saikia
- Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Mouli Sarkar
- Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India
| | - Vibin Ramakrishnan
- Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India; Mehta Family School of Data Science & Artificial Intelligence, Indian Institute of Technology Guwahati, Guwahati 781039, India.
| |
Collapse
|
3
|
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] [MESH Headings] [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
|
4
|
Liu Y, Xia X, Zheng M, Shi B. Bio-Nano Toolbox for Precision Alzheimer's Disease Gene Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2314354. [PMID: 38778446 DOI: 10.1002/adma.202314354] [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: 12/30/2023] [Revised: 05/01/2024] [Indexed: 05/25/2024]
Abstract
Alzheimer's disease (AD) is the most burdensome aging-associated neurodegenerative disorder, and its treatment encounters numerous failures during drug development. Although there are newly approved in-market β-amyloid targeting antibody solutions, pathological heterogeneity among patient populations still challenges the treatment outcome. Emerging advances in gene therapies offer opportunities for more precise personalized medicine; while, major obstacles including the pathological heterogeneity among patient populations, the puzzled mechanism for druggable target development, and the precision delivery of functional therapeutic elements across the blood-brain barrier remain and limit the use of gene therapy for central neuronal diseases. Aiming for "precision delivery" challenges, nanomedicine provides versatile platforms that may overcome the targeted delivery challenges for AD gene therapy. In this perspective, to picture a toolbox for AD gene therapy strategy development, the most recent advances from benchtop to clinics are highlighted, possibly available gene therapy targets, tools, and delivery platforms are outlined, their challenges as well as rational design elements are addressed, and perspectives in this promising research field are discussed.
Collapse
Affiliation(s)
- Yang Liu
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Xue Xia
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Meng Zheng
- Department of Radiotherapy and Translational Medicine Center, Huaihe Hospital of Henan University, Henan University, Kaifeng, Henan, 475000, China
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, New South Wales, 2109, Australia
| |
Collapse
|
5
|
Malik SA, Mondal S, Atreya HS. Alpha-Synuclein Aggregation Mechanism in the Presence of Nanomaterials. Biochemistry 2024; 63:1162-1169. [PMID: 38668883 DOI: 10.1021/acs.biochem.3c00624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Parkinson's disease (PD) is characterized by the toxic oligomeric and fibrillar phases formed by monomeric alpha-synuclein (α-syn). Certain nanoparticles have been demonstrated to promote protein aggregation, while other nanomaterials have been found to prevent the process. In the current work, we use nuclear magnetic resonance spectroscopy in conjunction with isothermal titration calorimetry to investigate the cause and mechanism of these opposing effects at the amino acid protein level. The interaction of α-syn with two types of nanomaterials was considered: citrate-capped gold nanoparticles (AuNPs) and graphene oxide (GO). In the presence of AuNPs, α-syn aggregation is accelerated, whereas in the presence of GO, aggregation is prevented. The study indicates that GO sequesters the NAC region of α-syn monomers through electrostatic and hydrophobic interactions, leading to a reduced elongation rate, and AuNPs leave the NAC region exposed while binding the N-terminus, leading to higher aggregation. The protein's inclination toward quicker aggregation is explained by the binding of the N-terminus of α-syn with the gold nanoparticles. Conversely, a comparatively stronger interaction with GO causes the nucleation and growth phases to be postponed and inhibits intermolecular interactions. Our finding offers novel experimental insights at the residue level regarding the aggregation of α-syn in the presence of various nanomaterials and creates new opportunities for the development of suitably functionalized nanomaterial-based therapeutic reagents against Parkinson's and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Shahid A Malik
- Department of Solid State and Structural Chemistry Unit, Indian Institute of Science, Bangalore 560012, India
- Nuclear Magnetic Resonance Research Centre, Indian Institute of Science, Bangalore 560012, India
- The John Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Somnath Mondal
- Department of Solid State and Structural Chemistry Unit, Indian Institute of Science, Bangalore 560012, India
- Nuclear Magnetic Resonance Research Centre, Indian Institute of Science, Bangalore 560012, India
- Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Hanudatta S Atreya
- Nuclear Magnetic Resonance Research Centre, Indian Institute of Science, Bangalore 560012, India
| |
Collapse
|
6
|
Ansari MA, Tripathi T, Venkidasamy B, Monziani A, Rajakumar G, Alomary MN, Alyahya SA, Onimus O, D'souza N, Barkat MA, Al-Suhaimi EA, Samynathan R, Thiruvengadam M. Multifunctional Nanocarriers for Alzheimer's Disease: Befriending the Barriers. Mol Neurobiol 2024; 61:3042-3089. [PMID: 37966683 DOI: 10.1007/s12035-023-03730-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 10/12/2023] [Indexed: 11/16/2023]
Abstract
Neurodegenerative diseases (NDDs) have been increasing in incidence in recent years and are now widespread worldwide. Neuronal death is defined as the progressive loss of neuronal structure or function which is closely associated with NDDs and represents the intrinsic features of such disorders. Amyotrophic lateral sclerosis, frontotemporal dementia, Alzheimer's, Parkinson's, and Huntington's diseases (AD, PD, and HD, respectively) are considered neurodegenerative diseases that affect a large number of people worldwide. Despite the testing of various drugs, there is currently no available therapy that can remedy or effectively slow the progression of these diseases. Nanomedicine has the potential to revolutionize drug delivery for the management of NDDs. The use of nanoparticles (NPs) has recently been developed to improve drug delivery efficiency and is currently subjected to extensive studies. Nanoengineered particles, known as nanodrugs, can cross the blood-brain barrier while also being less invasive compared to the most treatment strategies in use. Polymeric, magnetic, carbonic, and inorganic NPs are examples of NPs that have been developed to improve drug delivery efficiency. Primary research studies using NPs to cure AD are promising, but thorough research is needed to introduce these approaches to clinical use. In the present review, we discussed the role of metal-based NPs, polymeric nanogels, nanocarrier systems such as liposomes, solid lipid NPs, polymeric NPs, exosomes, quantum dots, dendrimers, polymersomes, carbon nanotubes, and nanofibers and surfactant-based systems for the therapy of neurodegenerative diseases. In addition, we highlighted nanoformulations such as N-butyl cyanoacrylate, poly(butyl cyanoacrylate), D-penicillamine, citrate-coated peptide, magnetic iron oxide, chitosan (CS), lipoprotein, ceria, silica, metallic nanoparticles, cholinesterase inhibitors, an acetylcholinesterase inhibitors, metal chelators, anti-amyloid, protein, and peptide-loaded NPs for the treatment of AD.
Collapse
Affiliation(s)
- Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research & Medical Consultations, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441, Dammam, Saudi Arabia
| | - Takshashila Tripathi
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Baskar Venkidasamy
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Alan Monziani
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Govindasamy Rajakumar
- Department of Orthodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Mohammad N Alomary
- Advanced Diagnostic and Therapeutic Institute, King Abdulaziz City for Science and Technology, 11442, Riyadh, Saudi Arabia
| | - Sami A Alyahya
- Wellness and Preventive Medicine Institute, King Abdulaziz City for Science and Technology, 11442, Riyadh, Saudi Arabia
| | - Oriane Onimus
- Faculty of Basic and Biomedical Sciences, University of Paris, Paris, France
| | - Naomi D'souza
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL, UK
| | - Md Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin, Saudi Arabia
| | - Ebtesam A Al-Suhaimi
- Research Consultation Department, Vice Presidency for Scientific Research and Innovation, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441, Dammam, Saudi Arabia
| | - Ramkumar Samynathan
- Department of Oral and Maxillofacial Surgery, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, Tamil Nadu, India
| | - Muthu Thiruvengadam
- Department of Crop Science, College of Sanghuh Life Science, Konkuk University, Seoul, 05029, Republic of Korea.
| |
Collapse
|
7
|
Duan L, Hao Z, Ji R, Li X, Wang H, Su Y, Guan F, Ma S. Glucose-modified BSA/procyanidin C1 NPs penetrate the blood-brain barrier and alleviate neuroinflammation in Alzheimer's disease models. Int J Biol Macromol 2024; 268:131739. [PMID: 38657920 DOI: 10.1016/j.ijbiomac.2024.131739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/12/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease with high prevalence, long duration and poor prognosis. The blood-brain barrier (BBB) is a physiologic barrier in the central nervous system, which hinders the entry of most drugs into the brain from the blood, thus affecting the efficacy of drugs for AD. Natural products are recognized as one of the promising and unique therapeutic approaches to treat AD. To improve the efficiency and therapeutic effect of the drug across the BBB, a natural polyphenolic compound, procyanidin C-1 (C1) was encapsulated in glucose-functionalized bovine serum albumin (BSA) nanoparticles to construct Glu-BSA/C1 NPs in our study. Glu-BSA/C1 NPs exhibited good stability, slow release, biocompatibility and antioxidant properties. In addition, Glu-BSA/C1 NPs penetrated the BBB, accumulated in the brain by targeting Glut1, and maintained the BBB integrity both in vitro and in vivo. Moreover, Glu-BSA/C1 NPs alleviated memory impairment of 5 × FAD mice by reducing Aβ deposition and Tau phosphorylation and promoting neurogenesis. Mechanistically, Glu-BSA/C1 NPs significantly activated the PI3K/AKT pathway and inhibited the NLRP3/Caspase-1/IL-1β pathway thereby suppressing neuroinflammation. Taken together, Glu-BSA/C1 NPs could penetrate the BBB and mitigate neuroinflammation in AD, which provides a new therapeutic approach targeting AD.
Collapse
Affiliation(s)
- Linyan Duan
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Zhizhong Hao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Rong Ji
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Xingfan Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Hao Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Yujing Su
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China; Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, Henan, China.
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, Henan, China.
| |
Collapse
|
8
|
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
|
9
|
Raj A, Bandyopadhyay U. Role of lysosome in healing neurological disorders by nano-bioengineering. Front Neurosci 2024; 17:1331211. [PMID: 38260032 PMCID: PMC10800409 DOI: 10.3389/fnins.2023.1331211] [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: 10/31/2023] [Accepted: 12/08/2023] [Indexed: 01/24/2024] Open
Abstract
Lysosomes primarily recognized as center for cellular 'garbage-disposing-unit', which has recently emerged as a crucial regulator of cellular metabolism. This organelle is a well-known vital player in the pathology including neurodegenerative disorders. In pathological context, removal of intracellular damaged misfolded proteins, organelles and aggregates are ensured by 'Autophagy' pathway, which initially recognizes, engulfs and seals the toxic cargo at the cytosolic environment. Thereafter the cell completes the task of encapsulated cargo elimination upon delivery of them to the terminal compartment - lysosome, which contains acid hydrolases, that are capable of degrading the abnormal protein-lipid-repertoire. The merge between inseparable 'Autophagy' and 'Lysosomal' pathways evolved into 'Autophagy-Lysosome Pathway (ALP)', through which cell ultimately degrades and recycles bio-materials for metabolic needs. Dysregulation of any of the steps of the multi-step ALP can contribute to the development and progression of disorders including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Therefore, targeting differential steps of ALP or directly lysosomes using nano-bioengineering approaches holds great promise for therapeutic interventions. This review aims to explore the role of distal autophagy pathway and proximal lysosomal function, as cellular degradative and metabolic hubs, in healing neurological disorders and highlights the contributions of nano-bioengineering in this field. Despite multiple challenges, this review underscores the immense potential of integrating autophagy-lysosomal biology with nano-bioengineering to revolutionize the field and provide novel therapeutic avenues for tackling neurological-neurodegenerative-disorders.
Collapse
Affiliation(s)
| | - Urmi Bandyopadhyay
- Manipal Institute of Regenerative Medicine (MIRM), Bengaluru, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| |
Collapse
|
10
|
Kavoosi S, Shahraki A, Sheervalilou R. Identification of microRNA-mRNA Regulatory Networks with Therapeutic Values in Alzheimer's Disease by Bioinformatics Analysis. J Alzheimers Dis 2024; 98:671-689. [PMID: 38427479 DOI: 10.3233/jad-230966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Background Alzheimer's disease (AD) is the most prevalent neurological disorder worldwide, affecting approximately 24 million individuals. Despite more than a century of research on AD, its pathophysiology is still not fully understood. Objective Recently, genetic studies of AD have focused on analyzing the general expression profile by employing high-throughput genomic techniques such as microarrays. Current research has leveraged bioinformatics advancements in genetic science to build upon previous efforts. Methods Data from the GSE118553 dataset used in this investigation, and the analyses carried out using programs such as Limma and BioBase. Differentially expressed genes (DEGs) and differentially expressed microRNAs (DEmiRs) associated with AD identified in the studied areas of the brain. Target genes of the DEmiRs identified using the MultiMiR package. Gene ontology (GO) completed using the Enrichr website, and the protein-protein interaction (PPI) network for these genes drawn using STRING and Cytoscape software. Results The findings introduced DEGs including CTNNB1, PAK2, MAP2K1, PNPLA6, IGF1R, FOXL2, DKK3, LAMA4, PABPN1, and GDPD5, and DEmiRs linked to AD (miR-106A, miR-1826, miR-1253, miR-10B, miR-18B, miR-101-2, miR-761, miR-199A1, miR-379 and miR-668), (miR-720, miR-218-2, miR-25, miR-602, miR-1226, miR-548K, miR-H1, miR-410, miR-548F2, miR-181A2), (miR-1470, miR-651, miR-544, miR-1826, miR-195, miR-610, miR-599, miR-323, miR-587 and miR-340), and (miR-1282, miR-1914, miR-642, miR-1323, miR-373, miR-323, miR-1322, miR-612, miR-606 and miR-758) in cerebellum, frontal cortex, temporal cortex, and entorhinal cortex, respectively. Conclusions The majority of the genes and miRNAs identified by our findings may be employed as biomarkers for prediction, diagnosis, or therapy response monitoring.
Collapse
Affiliation(s)
- Sakine Kavoosi
- Department of Biology, Faculty of Science, University of Sistan and Baluchestan, Zahedan, Iran
| | - Ali Shahraki
- Department of Biology, Faculty of Science, University of Sistan and Baluchestan, Zahedan, Iran
| | | |
Collapse
|
11
|
Al Abdullah S, Najm L, Ladouceur L, Ebrahimi F, Shakeri A, Al-Jabouri N, Didar TF, Dellinger K. Functional Nanomaterials for the Diagnosis of Alzheimer's Disease: Recent Progress and Future Perspectives. ADVANCED FUNCTIONAL MATERIALS 2023; 33:2302673. [PMID: 39309539 PMCID: PMC11415277 DOI: 10.1002/adfm.202302673] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Indexed: 09/25/2024]
Abstract
Alzheimer's disease (AD) is one of the main causes of dementia worldwide, whereby neuronal death or malfunction leads to cognitive impairment in the elderly population. AD is highly prevalent, with increased projections over the next few decades. Yet current diagnostic methods for AD occur only after the presentation of clinical symptoms. Evidence in the literature points to potential mechanisms of AD induction beginning before clinical symptoms start to present, such as the formation of amyloid beta (Aβ) extracellular plaques and neurofibrillary tangles (NFTs). Biomarkers of AD, including Aβ 40, Aβ 42, and tau protein, amongst others, show promise for early AD diagnosis. Additional progress is made in the application of biosensing modalities to measure and detect significant changes in these AD biomarkers within patient samples, such as cerebral spinal fluid (CSF) and blood, serum, or plasma. Herein, a comprehensive review of the emerging nano-biomaterial approaches to develop biosensors for AD biomarkers' detection is provided. Advances, challenges, and potential of electrochemical, optical, and colorimetric biosensors, focusing on nanoparticle-based (metallic, magnetic, quantum dots) and nanostructure-based biomaterials are discussed. Finally, the criteria for incorporating these emerging nano-biomaterials in clinical settings are presented and assessed, as they hold great potential for enhancing early-onset AD diagnostics.
Collapse
Affiliation(s)
- Saqer Al Abdullah
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 East Gate City Boulevard, Greensboro, NC 27401, USA
| | - Lubna Najm
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
| | - Liane Ladouceur
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L7, Canada
| | - Farbod Ebrahimi
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 East Gate City Boulevard, Greensboro, NC 27401, USA
| | - Amid Shakeri
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L7, Canada
| | - Nadine Al-Jabouri
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| | - Tohid F Didar
- School of Biomedical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L8, Canada
- Department of Mechanical Engineering, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4L7, Canada
- Institute for Infectious Disease Research (IIDR), 1280 Main St W, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Kristen Dellinger
- Department of Nanoengineering, Joint School of Nanoscience and Nanoengineering, North Carolina A&T State University, 2907 East Gate City Boulevard, Greensboro, NC 27401, USA
| |
Collapse
|
12
|
Nguyen TT, Nguyen-Thi PT, Nguyen THA, Ho TT, Tran NMA, Van Vo T, Van Vo G. Recent Advancements in Nanomaterials: A Promising Way to Manage Neurodegenerative Disorders. Mol Diagn Ther 2023; 27:457-473. [PMID: 37217723 DOI: 10.1007/s40291-023-00654-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2023] [Indexed: 05/24/2023]
Abstract
Neurodegenerative diseases (NDs) such as dementia, Alzheimer's disease, Parkinson's disease, frontotemporal dementia, and amyotrophic lateral sclerosis are some of the most prevalent disorders currently afflicting healthcare systems. Many of these diseases share similar pathological hallmarks, including elevated oxidative stress, mitochondrial dysfunction, protein misfolding, excitotoxicity, and neuroinflammation, all of which contribute to the deterioration of the nervous system's structure and function. The development of diagnostic and therapeutic materials in the monitoring and treatment of these diseases remains challenging. One of the biggest challenges facing therapeutic and diagnostic materials is the blood-brain barrier (BBB). The BBB is a multifunctional membrane possessing a plethora of biochemical, cellular, and immunological features that ensure brain homeostasis by preventing the entry and accumulation of unwanted compounds. With regards to neurodegenerative diseases, the recent application of tailored nanomaterials (nanocarriers and nanoparticles) has led to advances in diagnostics and therapeutics. In this review, we provide an overview of commonly used nanoparticles and their applications in NDs, which may offer new therapeutic strategies for the prevention and treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Thuy Trang Nguyen
- Faculty of Chemical Engineering, Industrial University of Ho Chi Minh City, Ho Chi Minh City, 71420, Vietnam
| | | | - Thi Hong Anh Nguyen
- Ho Chi Minh City University of Food Industry (HUFI), 140 Le Trong Tan Street, Tay Thanh Ward, Tan Phu District, Ho Chi Minh City, 700000, Vietnam
| | - Thanh-Tam Ho
- Institute for Global Health Innovations, Duy Tan University, Da Nang, 550000, Vietnam.
- Faculty of Pharmacy, Duy Tan University, Da Nang, 550000, Vietnam.
| | - Nguyen-Minh-An Tran
- Faculty of Chemical Engineering, Industrial University of Ho Chi Minh City, Ho Chi Minh City, 71420, Vietnam
| | - Toi Van Vo
- Tissue Engineering and Regenerative Medicine Department, School of Biomedical Engineering, International University, Ho Chi Minh City, 700000, Vietnam.
- Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam.
| | - Giau Van Vo
- Department of Biomedical Engineering, School of Medicine, Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam.
- Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, Vietnam National University, Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam.
- Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam.
| |
Collapse
|
13
|
Resmi AN, Rekha CR, Dhushyandhun ME, Elangovan S, Shenoy SJ, Gulia KK, Jayasree RS. Bifunctional cysteine gold nanoclusters for β-amyloid fibril inhibition and fluorescence imaging: a distinctive approach to manage Alzheimer's disease. J Mater Chem B 2023; 11:4715-4724. [PMID: 37171084 DOI: 10.1039/d2tb02802f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Alzheimer's disease (AD) is a progressive complex neurodegenerative disorder affecting millions of individuals worldwide. Currently, there is no effective treatment for AD. AD is characterized by the deposition of amyloid plaques/fibrils. One major strategy for managing this disease is by slowing the progression of AD using different drugs which could potentially limit free-radical formation, oxidative stress and lipid peroxidation and promote the survival of neurons exposed to β-amyloid. Inhibition of amyloid fibrillization and clearance of amyloid plaques/fibrils are essential for the prevention and treatment of AD. The thiophilic interaction between the side chain of an aromatic residue in a polypeptide and a sulphur atom of the compound can effectively inhibit amyloid fibril formation. In this work, we have synthesized cysteine-capped gold nanoclusters (Cy-AuNCs) which exhibit inherent red emission and can disintegrate amyloid fibrils through the aforementioned thiophilic interactions. Herein, we also used molecular docking to study the thiophilic interactions between the sulphur atom of Cy-AuNCs and the aromatic rings of the protein. Finally, the gold cluster was functionalized with a brain targeting molecule, Levodopa (AuCs-LD), to specifically target the brain and to facilitate passage through the blood brain barrier (BBB). Both Cy-AuNCs and AuCs-LD showed good biocompatibility and the inherent fluorescence properties of nanoclusters enabled real time imaging. The efficacy of the nanoclusters to disintegrate amyloid fibrils and their ability to cross the BBB were demonstrated both in vitro and in vivo in the BBB model and the AD animal model respectively. Our results imply that nanoparticle-based artificial molecular chaperones may offer a promising therapeutic approach for AD.
Collapse
Affiliation(s)
- A N Resmi
- Division of Biophotonics and Imaging, Department of Biomaterial Sciences and Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum 695012, India.
| | - C R Rekha
- Division of Biophotonics and Imaging, Department of Biomaterial Sciences and Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum 695012, India.
| | - M E Dhushyandhun
- Division of Biophotonics and Imaging, Department of Biomaterial Sciences and Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum 695012, India.
| | - Sarathkumar Elangovan
- Division of Biophotonics and Imaging, Department of Biomaterial Sciences and Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum 695012, India.
| | - Sachin J Shenoy
- Division of in vivo Models and Testing, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum 695012, India
| | - Kamalesh K Gulia
- Division of Sleep Research, Department of Applied Biology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum 695012, India
| | - Ramapurath S Jayasree
- Division of Biophotonics and Imaging, Department of Biomaterial Sciences and Technology, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (SCTIMST), Trivandrum 695012, India.
| |
Collapse
|
14
|
Duan L, Li X, Ji R, Hao Z, Kong M, Wen X, Guan F, Ma S. Nanoparticle-Based Drug Delivery Systems: An Inspiring Therapeutic Strategy for Neurodegenerative Diseases. Polymers (Basel) 2023; 15:2196. [PMID: 37177342 PMCID: PMC10181407 DOI: 10.3390/polym15092196] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Neurodegenerative diseases are common, incurable neurological disorders with high prevalence, and lead to memory, movement, language, and intelligence impairments, threatening the lives and health of patients worldwide. The blood-brain barrier (BBB), a physiological barrier between the central nervous system and peripheral blood circulation, plays an important role in maintaining the homeostasis of the intracerebral environment by strictly regulating the transport of substances between the blood and brain. Therefore, it is difficult for therapeutic drugs to penetrate the BBB and reach the brain, and this affects their efficacy. Nanoparticles (NPs) can be used as drug transport carriers and are also known as nanoparticle-based drug delivery systems (NDDSs). These systems not only increase the stability of drugs but also facilitate the crossing of drugs through the BBB and improve their efficacy. In this article, we provided an overview of the types and administration routes of NPs, highlighted the preclinical and clinical studies of NDDSs in neurodegenerative diseases, and summarized the combined therapeutic strategies in the management of neurodegenerative diseases. Finally, the prospects and challenges of NDDSs in recent basic and clinical research were also discussed. Above all, NDDSs provide an inspiring therapeutic strategy for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Linyan Duan
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (L.D.); (X.L.); (R.J.); (Z.H.)
| | - Xingfan Li
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (L.D.); (X.L.); (R.J.); (Z.H.)
| | - Rong Ji
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (L.D.); (X.L.); (R.J.); (Z.H.)
| | - Zhizhong Hao
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (L.D.); (X.L.); (R.J.); (Z.H.)
| | - Mingyue Kong
- NHC Key Laboratory of Birth Defects Prevention, Henan Key Laboratory of Population Defects Prevention, Zhengzhou 450002, China;
| | - Xuejun Wen
- Department of Chemical and Life Science Engineering, School of Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (L.D.); (X.L.); (R.J.); (Z.H.)
- Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China
| | - Shanshan Ma
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China; (L.D.); (X.L.); (R.J.); (Z.H.)
- NHC Key Laboratory of Birth Defects Prevention, Henan Key Laboratory of Population Defects Prevention, Zhengzhou 450002, China;
- Institute of Neuroscience, Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
15
|
Dahri M, Miri Jahromi A, Nikzad A, Mohammadgholian M, Rahmanian M, Abolmaali SS, Maleki R. Novel bioengineered MBenes for the treatment of Alzheimer's disease: An in-Sillico study. J Biomol Struct Dyn 2022; 40:12268-12276. [PMID: 34427178 DOI: 10.1080/07391102.2021.1969288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease is a neurodegenerative disease caused by the deposition and accumulation of amyloid-β (Aβ) peptides in the brain neurons. Current medications are not a definitive cure for this disease, but they can hamper the signs and symptoms of Alzheimer's disease. Therefore, prevention is the best way to deal with this disease. In this study, the novel structures based on MBenes (such as Cd2B, Mo2B, Cu2B, and Ta2B) are proposed to prevent amyloid-β accumulation in Alzheimer's disease. Regarding the remarkable MBene properties such as tunability, biocompatibility, and low manufacturing cost, the effect of these structures on amyloid-β deformation was explored using molecular dynamics simulation. To provide an atomic analysis of Beta-amyloid behavior in the presence of these structures, the compaction, contact area, and stability of Beta-amyloid were investigated. The results indicated the satisfactory performance of MBenes on the destabilization of amyloid-β structures. Moreover, given the higher interactions between Cd2B and amyloid-β, the instability, compaction, and the contact area of amyloid-β particles were investigated in this complex. The findings confirmed Cd2B as the best structure to prevent amyloid-β accumulation. The results of this investigation paved the way for the development of these structures as a medicinal agent to prevent Alzheimer's disease.
Collapse
Affiliation(s)
- Mohammad Dahri
- Computational Biology and Chemistry Group (CBCG), Universal Scientific Education and Research Network (USERN), Department of Physics, Tehran University, Tehran, Iran.,Center for Nanotechnology in Drug Delivery, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Miri Jahromi
- Computational Biology and Chemistry Group (CBCG), Universal Scientific Education and Research Network (USERN), Department of Physics, Tehran University, Tehran, Iran
| | - Arash Nikzad
- The University of British Columbia, Vancouver, Canada
| | - Maryam Mohammadgholian
- Computational Biology and Chemistry Group (CBCG), Universal Scientific Education and Research Network (USERN), Department of Physics, Tehran University, Tehran, Iran
| | - Mohammad Rahmanian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Samira Sadat Abolmaali
- Center for Nanotechnology in Drug Delivery, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Maleki
- Computational Biology and Chemistry Group (CBCG), Universal Scientific Education and Research Network (USERN), Department of Physics, Tehran University, Tehran, Iran
| |
Collapse
|
16
|
Lai Y, Dang Y, Sun Q, Pan J, Yu H, Zhang W, Xu Z. Design of an activatable NIR-II nanoprobe for the in vivo elucidation of Alzheimer's disease-related variations in methylglyoxal concentrations. Chem Sci 2022; 13:12511-12518. [PMID: 36349272 PMCID: PMC9628982 DOI: 10.1039/d2sc05242c] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 10/07/2022] [Indexed: 12/19/2024] Open
Abstract
Clear elucidation of the changes in Alzheimer's disease (AD)-related methylglyoxal (MGO) levels in vivo is significant yet highly challenging. Fluorescence imaging in the second near-infrared region (NIR-II, 1000-1700 nm) has gained increasing attention as an observation method in living organisms, but an MGO-activatable fluorescent probe that emits in this region for in vivo brain imaging is lacking because of the existence of the blood-brain barrier (BBB). Herein, a biocompatible Fe3O4 nanoparticle (IONP)-conjugated MGO-activatable NIR-II fluorescent probe (MAM) modified with the peptide T7 (HAIYPRH) (named TM-IONP) is reported for the in situ detection of MGO in a transgenic AD mouse model. In this system, the T7 peptide enhances BBB crossing and brain accumulation by specifically targeting transferrin receptors on the BBB. Due to the MAM probe, TM-IONPs emit fluorescence in the NIR-II region and display high selectivity with an MGO detection limit of 72 nM and a 10-fold increase in the fluorescence signal. After intravenous administration, the TM-IONPs are easily delivered to the brain and pass through the BBB without intervention, and as a result, the brains of AD mice can be noninvasively imaged for the first time by the in situ detection of MGO with a 24.2-fold enhancement in NIR-II fluorescence intensity compared with wild-type mice. Thus, this MGO-activated NIR-II-emitting nanoprobe is potentially useful for early AD diagnosis in clinic.
Collapse
Affiliation(s)
- Yi Lai
- School of Chemistry and Molecular Engineering, East China Normal University 500 Dongchuan Road Shanghai 200241 China
| | - Yijing Dang
- School of Chemistry and Molecular Engineering, East China Normal University 500 Dongchuan Road Shanghai 200241 China
| | - Qian Sun
- School of Chemistry and Molecular Engineering, East China Normal University 500 Dongchuan Road Shanghai 200241 China
| | - Jiaxing Pan
- State Key Laboratory of Drug Research, Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences 501 Haike Road Shanghai 201203 China
| | - Haijun Yu
- State Key Laboratory of Drug Research, Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences 501 Haike Road Shanghai 201203 China
| | - Wen Zhang
- School of Chemistry and Molecular Engineering, East China Normal University 500 Dongchuan Road Shanghai 200241 China
| | - Zhiai Xu
- School of Chemistry and Molecular Engineering, East China Normal University 500 Dongchuan Road Shanghai 200241 China
| |
Collapse
|
17
|
Significance of native PLGA nanoparticles in the treatment of Alzheimer's disease pathology. Bioact Mater 2022; 17:506-525. [PMID: 36330076 PMCID: PMC9614411 DOI: 10.1016/j.bioactmat.2022.05.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 04/26/2022] [Accepted: 05/23/2022] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is believed to be triggered by increased levels/aggregation of β-amyloid (Aβ) peptides. At present, there is no effective disease-modifying treatment for AD. Here, we evaluated the therapeutic potential of FDA-approved native poly(d,l-lactide-co-glycolide) (PLGA) nanoparticles on Aβ aggregation and in cellular/animal models of AD. Our results showed that native PLGA can not only suppress the spontaneous aggregation but can also trigger disassembly of preformed Aβ aggregates. Spectroscopic studies, molecular dynamics simulations and biochemical analyses revealed that PLGA, by interacting with the hydrophobic domain of Aβ1-42, prevents a conformational shift towards the β-sheet structure, thus precluding the formation and/or triggering disassembly of Aβ aggregates. PLGA-treated Aβ samples can enhance neuronal viability by reducing phosphorylation of tau protein and its associated signaling mechanisms. Administration of PLGA can interact with Aβ aggregates and attenuate memory deficits as well as Aβ levels/deposits in the 5xFAD mouse model of AD. PLGA can also protect iPSC-derived neurons from AD patients against Aβ toxicity by decreasing tau phosphorylation. These findings provide unambiguous evidence that native PLGA, by targeting different facets of the Aβ axis, can have beneficial effects in mouse neurons/animal models as well as on iPSC-derived AD neurons - thus signifying its unique therapeutic potential in the treatment of AD pathology. PLGA nanoparticles by interacting with hydrophobic domain inhibit Aβ aggregation. PLGA-mediated inhibition of Aβ aggregation can increase viability of mouse neurons. PLGA administration can attenuate cognitive deficits/pathology in 5xFAD AD mouse model. PLGA can protect iPSC-derived neurons from AD patients against Aβ toxicity.
Collapse
|
18
|
Shiravandi A, Yari F, Tofigh N, Kazemi Ashtiani M, Shahpasand K, Ghanian MH, Shekari F, Faridbod F. Earlier Detection of Alzheimer's Disease Based on a Novel Biomarker cis P-tau by a Label-Free Electrochemical Immunosensor. BIOSENSORS 2022; 12:879. [PMID: 36291017 PMCID: PMC9599477 DOI: 10.3390/bios12100879] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/01/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
Early detection of cis phosphorylated tau (cis P-tau) may help as an effective treatment to control the progression of Alzheimer's disease (AD). Recently, we introduced for the first time a monoclonal antibody (mAb) with high affinity against cis P-tau. In this study, the cis P-tau mAb was utilized to develop a label-free immunosensor. The antibody was immobilized onto a gold electrode and the electrochemical responses to the analyte were acquired by electrochemical impedance spectroscopy (EIS), cyclic voltammetry (CV), and differential pulse voltammetry (DPV). The immunosensor was capable of selective detection of cis P-tau among non-specific targets like trans P-tau and major plasma proteins. A wide concentration range (10 × 10-14 M-3.0 × 10-9 M) of cis P-tau was measured in PBS and human serum matrices with a limit of detection of 0.02 and 0.05 pM, respectively. Clinical applicability of the immunosensor was suggested by its long-term storage stability and successful detection of cis P-tau in real samples of cerebrospinal fluid (CSF) and blood serum collected from human patients at different stages of AD. These results suggest that this simple immunosensor may find great application in clinical settings for early detection of AD which is an unmet urgent need in today's healthcare services.
Collapse
Affiliation(s)
- Ayoub Shiravandi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Farzaneh Yari
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran P.O. Box 14155-6455, Iran
| | - Nahid Tofigh
- Laboratory of Neuro-Organic Chemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 1417935840, Iran
| | - Mohammad Kazemi Ashtiani
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Koorosh Shahpasand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Mohammad-Hossein Ghanian
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Faezeh Shekari
- Advanced Therapy Medicinal Product Technology Development Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research (ACECR), Tehran 1665659911, Iran
| | - Farnoush Faridbod
- Center of Excellence in Electrochemistry, School of Chemistry, College of Science, University of Tehran, Tehran P.O. Box 14155-6455, Iran
| |
Collapse
|
19
|
Hou T, Zhang N, Yan C, Ding M, Niu H, Guan P, Hu X. Curcumin-loaded protein imprinted mesoporous nanosphere for inhibiting amyloid aggregation. Int J Biol Macromol 2022; 221:334-345. [PMID: 36084870 DOI: 10.1016/j.ijbiomac.2022.08.185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/18/2022] [Accepted: 08/29/2022] [Indexed: 11/24/2022]
Abstract
Some natural variants of human lysozyme are associated with systemic non-neurological amyloidosis that leads to amyloid protein fibril deposition in different tissues. Inhibition of amyloid fibrillation by nanomaterials is considered to be an effective approach to treating amyloidosis. Here, we prepared a targeted, highly loaded curcumin lysozyme-imprinted nanosphere (CUR-MIMS) that could effectively inhibit the aggregation of lysozyme with lysozyme adsorption capacity of 193.57 mg g-1 and the imprinting factor (IF) of 3.72. CUR-MIMS could bind to lysozyme through hydrophobic interactions and effectively reduce the hydrophobicity of the total solvent-exposed surface in lysozyme fibrillation, thus reducing the self-assembly process triggered by hydrophobic interactions. Thioflavin T (ThT) analysis demonstrated that CUR-MIMS inhibited the aggregation of amyloid fibrils in a dose-dependent manner (inhibition efficiency of 56.07 %). Circular dichroism (CD) spectrum further illustrated that CUR-MIMS could significantly inhibit the transition of lysozyme from α-helix structure to β-sheet. More importantly, biological experiments proved the good biocompatibility of CUR-MIMS, which indicated the potential of our system as a future therapeutic platform for amyloidosis.
Collapse
Affiliation(s)
- Tongtong Hou
- Department of Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, PR China
| | - Nan Zhang
- Department of Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, PR China
| | - Chaoren Yan
- Department of Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, PR China
| | - Minling Ding
- Department of Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, PR China
| | - Huizhe Niu
- Department of Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, PR China
| | - Ping Guan
- Department of Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, PR China.
| | - Xiaoling Hu
- Department of Chemistry, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an 710072, PR China.
| |
Collapse
|
20
|
Wu Q, Karthivashan G, Nakhaei-Nejad M, Anand BG, Giuliani F, Kar S. Native PLGA nanoparticles regulate APP metabolism and protect neurons against β-amyloid toxicity: Potential significance in Alzheimer's disease pathology. Int J Biol Macromol 2022; 219:1180-1196. [PMID: 36030976 DOI: 10.1016/j.ijbiomac.2022.08.148] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/05/2022]
Abstract
Biodegradable poly(lactic-co-glycolic acid)(PLGA) nanoparticles have been used extensively in delivering drugs to target tissues due to their excellent biocompatibility. Evidence suggests that PLGA-conjugated drugs/agents can attenuate pathology in cellular/animal models of Alzheimer's disease (AD), which is initiated by increased level/aggregation of amyloid β (Aβ) peptide generated from amyloid precursor protein (APP). The beneficial effects were attributed to conjugated-drugs rather than to PLGA nanoparticles. Interestingly, we recently reported that PLGA without any drug/agent (native PLGA) can suppress Aβ aggregation/toxicity. However, very little is known about the internalization, subcellular localization or effects of PLGA in neurons. In this study, using primary mouse cortical neurons, we first showed that native PLGA is internalized by an energy-mediated clathrin-dependent/-independent pathway and is localized in endosomal-lysosomal-autophagic vesicles. By attenuating internalization, PLGA can protect neurons against Aβ-mediated toxicity. Additionally, PLGA treatment altered expression profiles of certain AD-associated genes and decreased the levels of APP, its cleaved products α-/β-CTFs and Aβ peptides in mouse as well as iPSC-derived neurons from control and AD patients. Collectively, these results suggest that native PLGA not only protects neurons against Aβ-induced toxicity but also influences the expression of AD-related genes/proteins - highlighting PLGA's implication in normal and AD-related pathology.
Collapse
Affiliation(s)
- Qi Wu
- Departments of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada.
| | - Govindarajan Karthivashan
- Departments of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada.
| | - Maryam Nakhaei-Nejad
- Departments of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada.
| | - Bibin G Anand
- Departments of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada.
| | - Fabrizio Giuliani
- Departments of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada.
| | - Satyabrata Kar
- Departments of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
21
|
A field-deployable diagnostic assay for the visual detection of misfolded prions. Sci Rep 2022; 12:12246. [PMID: 35851406 PMCID: PMC9293997 DOI: 10.1038/s41598-022-16323-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/08/2022] [Indexed: 11/30/2022] Open
Abstract
Diagnostic tools for the detection of protein-misfolding diseases (i.e., proteopathies) are limited. Gold nanoparticles (AuNPs) facilitate sensitive diagnostic techniques via visual color change for the identification of a variety of targets. In parallel, recently developed quaking-induced conversion (QuIC) assays leverage protein-amplification and fluorescent signaling for the accurate detection of misfolded proteins. Here, we combine AuNP and QuIC technologies for the visual detection of amplified misfolded prion proteins from tissues of wild white-tailed deer infected with chronic wasting disease (CWD), a prion disease of cervids. Our newly developed assay, MN-QuIC, enables both naked-eye and light-absorbance measurements for detection of misfolded prions. MN-QuIC leverages basic laboratory equipment that is cost-effective and portable, thus facilitating real-time prion diagnostics across a variety of settings. In addition to laboratory-based tests, we deployed to a rural field-station in southeastern Minnesota and tested for CWD on site. We successfully demonstrated that MN-QuIC is functional in a non-traditional laboratory setting by performing a blinded analysis in the field and correctly identifying all CWD positive and CWD not-detected deer at the field site in 24 h, thus documenting the portability of the assay. White-tailed deer tissues used to validate MN-QuIC included medial retropharyngeal lymph nodes, parotid lymph nodes, and palatine tonsils. Importantly, all of the white-tailed deer (n = 63) were independently tested using ELISA, IHC, and/or RT-QuIC technologies and results secured with MN-QuIC were 95.7% and 100% consistent with these tests for positive and non-detected animals, respectively. We hypothesize that electrostatic forces help govern the AuNP/prion interactions and conclude that MN-QuIC has great potential for sensitive, field-deployable diagnostics for CWD, with future potential diagnostic applications for a variety of proteopathies.
Collapse
|
22
|
Huang R, Zhou X, Chen G, Su L, Liu Z, Zhou P, Weng J, Min Y. Advances of functional nanomaterials for magnetic resonance imaging and biomedical engineering applications. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1800. [PMID: 35445588 DOI: 10.1002/wnan.1800] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 11/12/2022]
Abstract
Functional nanomaterials have been widely used in biomedical fields due to their good biocompatibility, excellent physicochemical properties, easy surface modification, and easy regulation of size and morphology. Functional nanomaterials for magnetic resonance imaging (MRI) can target specific sites in vivo and more easily detect disease-related specific biomarkers at the molecular and cellular levels than traditional contrast agents, achieving a broad application prospect in MRI. This review focuses on the basic principles of MRI, the classification, synthesis and surface modification methods of contrast agents, and their clinical applications to provide guidance for designing novel contrast agents and optimizing the contrast effect. Furthermore, the latest biomedical advances of functional nanomaterials in medical diagnosis and disease detection, disease treatment, the combination of diagnosis and treatment (theranostics), multi-model imaging and nanozyme are also summarized and discussed. Finally, the bright application prospects of functional nanomaterials in biomedicine are emphasized and the urgent need to achieve significant breakthroughs in the industrial transformation and the clinical translation is proposed. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Diagnostic Tools > Diagnostic Nanodevices Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Ruijie Huang
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Xingyu Zhou
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Guiyuan Chen
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Lanhong Su
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Zhaoji Liu
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Peijie Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jianping Weng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yuanzeng Min
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Department of Chemistry, University of Science and Technology of China, Hefei, China
| |
Collapse
|
23
|
Future of Alzheimer’s Disease: Nanotechnology-Based Diagnostics and Therapeutic Approach. BIONANOSCIENCE 2022. [DOI: 10.1007/s12668-022-00998-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
24
|
Xu L, Wang H, Xu Y, Cui W, Ni W, Chen M, Huang H, Stewart C, Li L, Li F, Han J. Machine Learning-Assisted Sensor Array Based on Poly(amidoamine) (PAMAM) Dendrimers for Diagnosing Alzheimer's Disease. ACS Sens 2022; 7:1315-1322. [PMID: 35584464 DOI: 10.1021/acssensors.2c00132] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent neurodegenerative disorder, and the early diagnosis of AD remains challenging. Here we have developed a fluorescent sensor array composed of three modified polyamidoamine dendrimers. Proteins of various properties were differentiated via this array with 100% accuracy, proving the rationality of the array's design. The mechanism of the fluorescence response was discussed. Furthermore, the robust three-element array enables parallel detection of multiple Aβ40/Aβ42 aggregates (0.5 μM) in diverse interferents, serum media, and cerebrospinal fluid (CSF) with high accuracy, through machine learning algorithms, demonstrating the tremendous potential of the sensor array in Alzheimer's disease diagnosis.
Collapse
Affiliation(s)
- Lian Xu
- State Key Laboratory of Natural Medicines and National R&D Center for Chinese Herbal Medicine Processing, Department of Food Quality and Safety, College of Engineering, China Pharmaceutical University, Nanjing 211109, China
| | - Hao Wang
- State Key Laboratory of Natural Medicines and National R&D Center for Chinese Herbal Medicine Processing, Department of Food Quality and Safety, College of Engineering, China Pharmaceutical University, Nanjing 211109, China
| | - Yu Xu
- State Key Laboratory of Natural Medicines and National R&D Center for Chinese Herbal Medicine Processing, Department of Food Quality and Safety, College of Engineering, China Pharmaceutical University, Nanjing 211109, China
| | - Wenyu Cui
- State Key Laboratory of Natural Medicines and National R&D Center for Chinese Herbal Medicine Processing, Department of Food Quality and Safety, College of Engineering, China Pharmaceutical University, Nanjing 211109, China
| | - Weiwei Ni
- State Key Laboratory of Natural Medicines and National R&D Center for Chinese Herbal Medicine Processing, Department of Food Quality and Safety, College of Engineering, China Pharmaceutical University, Nanjing 211109, China
| | - Mingqi Chen
- State Key Laboratory of Natural Medicines and National R&D Center for Chinese Herbal Medicine Processing, Department of Food Quality and Safety, College of Engineering, China Pharmaceutical University, Nanjing 211109, China
| | - Hui Huang
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Callum Stewart
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Linxian Li
- Ming Wai Lau Centre for Reparative Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Fei Li
- State Key Laboratory of Natural Medicines and National R&D Center for Chinese Herbal Medicine Processing, Department of Food Quality and Safety, College of Engineering, China Pharmaceutical University, Nanjing 211109, China
| | - Jinsong Han
- State Key Laboratory of Natural Medicines and National R&D Center for Chinese Herbal Medicine Processing, Department of Food Quality and Safety, College of Engineering, China Pharmaceutical University, Nanjing 211109, China
| |
Collapse
|
25
|
Yao L, Zhou Z, Wang S, Zou Q, Wang HX, Ma LX, Wang S, Zhang X. Phosphorylation of covalent organic framework nanospheres for inhibition of amyloid-β peptide fibrillation. Chem Sci 2022; 13:5902-5912. [PMID: 35685783 PMCID: PMC9132083 DOI: 10.1039/d2sc00253a] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/21/2022] [Indexed: 12/27/2022] Open
Abstract
The development and exploration of new nanostructural inhibitors against Alzheimer's disease (AD)-associated amyloid-β (Aβ) fibrillation have attracted extensive attention and become a new frontier in nanomedicine. However, focusing on finding an effective nanostructure is one of the most challenging parts of the therapeutics task. Herein, nanoscale spherical covalent organic frameworks (COFs) via post-synthetic functionalization with sodium phosphate (SP) groups on the channel networks were found to efficiently inhibit Aβ fibrillation. The as-prepared uniform SP-COF nanospheres with high surface area, good crystallinity, and chemical stability were characterized by multifarious microscopic and spectroscopic techniques. Moreover, molecular dynamics simulation together with fibrillation kinetics and cytotoxicity assay experiments shows that there were restricted-access adsorption channels in the SP-COFs which were formed by the cavities with size and functional groups accommodated to the Aβ peptide sequence and significantly affected the fibrillation and cytotoxicity of Aβ. Transmission electron microscopy (TEM), dynamic light scattering (DLS) monitoring, isothermal titration calorimetry (ITC), Fourier transform infrared (FT-IR) and circular dichroism (CD) spectra measurements, and confocal imaging observation were performed to understand the inhibition mechanism and influencing factors of the SP-COFs. To our knowledge, our strategy is the first exploration of COF-based anti-amyloidogenic nanomaterials with high affinity and specific targeting, which are crucial for the inhibition of Aβ fibrillation for AD prevention and treatment. Nanoscale spherical COFs via phosphorylation functionalization were found to efficiently inhibit fibrillation of the Alzheimer's disease-associated Aβ peptide.![]()
Collapse
Affiliation(s)
- Linli Yao
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University Wuhan 430062 China
| | - Zhe Zhou
- Department of Neurology, The First Hospital of Lanzhou University Lanzhou 730000 China
| | - Suxiao Wang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University Wuhan 430062 China
| | - Qichao Zou
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University Wuhan 430062 China
| | - Hang-Xing Wang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University Wuhan 430062 China
| | - Li-Xin Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University Wuhan 430062 China
| | - Shengfu Wang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University Wuhan 430062 China
| | - Xiuhua Zhang
- Collaborative Innovation Center for Advanced Organic Chemical Materials Co-constructed by the Province and Ministry, College of Chemistry and Chemical Engineering, Hubei University Wuhan 430062 China
| |
Collapse
|
26
|
Cheng G, Liu Y, Ma R, Cheng G, Guan Y, Chen X, Wu Z, Chen T. Anti-Parkinsonian Therapy: Strategies for Crossing the Blood-Brain Barrier and Nano-Biological Effects of Nanomaterials. NANO-MICRO LETTERS 2022; 14:105. [PMID: 35426525 PMCID: PMC9012800 DOI: 10.1007/s40820-022-00847-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 03/12/2022] [Indexed: 05/08/2023]
Abstract
Parkinson's disease (PD), a neurodegenerative disease that shows a high incidence in older individuals, is becoming increasingly prevalent. Unfortunately, there is no clinical cure for PD, and novel anti-PD drugs are therefore urgently required. However, the selective permeability of the blood-brain barrier (BBB) poses a huge challenge in the development of such drugs. Fortunately, through strategies based on the physiological characteristics of the BBB and other modifications, including enhancement of BBB permeability, nanotechnology can offer a solution to this problem and facilitate drug delivery across the BBB. Although nanomaterials are often used as carriers for PD treatment, their biological activity is ignored. Several studies in recent years have shown that nanomaterials can improve PD symptoms via their own nano-bio effects. In this review, we first summarize the physiological features of the BBB and then discuss the design of appropriate brain-targeted delivery nanoplatforms for PD treatment. Subsequently, we highlight the emerging strategies for crossing the BBB and the development of novel nanomaterials with anti-PD nano-biological effects. Finally, we discuss the current challenges in nanomaterial-based PD treatment and the future trends in this field. Our review emphasizes the clinical value of nanotechnology in PD treatment based on recent patents and could guide researchers working in this area in the future.
Collapse
Affiliation(s)
- Guowang Cheng
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People's Republic of China
| | - Yujing Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Rui Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Guopan Cheng
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Yucheng Guan
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China
| | - Xiaojia Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, People's Republic of China
| | - Zhenfeng Wu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, People's Republic of China.
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, People's Republic of China.
| |
Collapse
|
27
|
Gao KX, Zhou Z, Yao L, Wang S, Zhang Y, Zou Q, Ma LX, Wang HX. Aspartic Acid-Assisted Size-Controllable Synthesis of Nanoscale Spherical Covalent Organic Frameworks with Chiral Interfaces for Inhibiting Amyloid-β Fibrillation. ACS APPLIED BIO MATERIALS 2022; 5:1210-1221. [PMID: 35191674 DOI: 10.1021/acsabm.1c01245] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Covalent organic framework nanospheres (COF NSs) have garnered special attention due to their uniform sphere morphology, adjustable particle size, and mesoporous microenvironment. However, methods to control an optimal particle size scale while achieving solution dispersibility and specific surface properties remain underdeveloped, which precludes many of the biomedical applications. Here, we propose and develop a general strategy to access simultaneous size control and surface functionalization of uniform spherical COF NSs in a single step using aspartic acid (d-/l-Asp) that plays center roles in an acid catalyst, hydrophilicity, size-controllable synthesis, and chiral enantiomer. In this study, for the first time, we have employed a surface chemistry engineering study to create a variety of nanoscale spherical COFs and subsequently measure parameters to evaluate the effectiveness of Asp in the regulation of the particle size. Moreover, the potential utilization of the d/l-enantiomeric Asp-COF NSs in preventing β-amyloid (Aβ) aggregation is investigated by analyzing their interactions with Aβ amyloids using a multitechnique experimental approach. To our knowledge, our strategy is the first synthesis of hydrophilic COF NSs with an optimal length scale and a chiral-selective targeting surface, which are crucial for the inhibition of Aβ fibrillation for Alzheimer's disease prevention.
Collapse
Affiliation(s)
- Kai-Xiang Gao
- College of Chemistry and Chemical Engineering, Hubei University, No. 368 Youyi Avenue, Wuchang District, Wuhan 430062, China
| | - Zhe Zhou
- Department of Neurology, The First Hospital of Lanzhou University, No. 1 Donggang West Road, Chengguan District, Lanzhou 730000, China
| | - Linli Yao
- College of Chemistry and Chemical Engineering, Hubei University, No. 368 Youyi Avenue, Wuchang District, Wuhan 430062, China
| | - Suxiao Wang
- College of Chemistry and Chemical Engineering, Hubei University, No. 368 Youyi Avenue, Wuchang District, Wuhan 430062, China
| | - Yuexing Zhang
- College of Chemistry and Chemical Engineering, Hubei University, No. 368 Youyi Avenue, Wuchang District, Wuhan 430062, China
| | - Qichao Zou
- College of Chemistry and Chemical Engineering, Hubei University, No. 368 Youyi Avenue, Wuchang District, Wuhan 430062, China
| | - Li-Xin Ma
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, Hubei University, No. 368, Youyi Avenue, Wuchang District, Wuhan 430062, China
| | - Hang-Xing Wang
- College of Chemistry and Chemical Engineering, Hubei University, No. 368 Youyi Avenue, Wuchang District, Wuhan 430062, China
| |
Collapse
|
28
|
Paul PS, Cho JY, Wu Q, Karthivashan G, Grabovac E, Wille H, Kulka M, Kar S. Unconjugated PLGA nanoparticles attenuate temperature-dependent β-amyloid aggregation and protect neurons against toxicity: implications for Alzheimer's disease pathology. J Nanobiotechnology 2022; 20:67. [PMID: 35120558 PMCID: PMC8817552 DOI: 10.1186/s12951-022-01269-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 01/16/2022] [Indexed: 12/26/2022] Open
Abstract
Conversion of β-amyloid (Aβ) peptides from soluble random-coil to aggregated protein enriched with β-sheet-rich intermediates has been suggested to play a role in the degeneration of neurons and development of Alzheimer's disease (AD) pathology. Aggregation of Aβ peptide can be prompted by a variety of environmental factors including temperature which can influence disease pathogenesis. Recently, we reported that FDA-approved unconjugated poly (D,L-lactide-co-glycolide) (PLGA) nanoparticles can have beneficial effects in cellular and animal models of AD by targeting different facets of the Aβ axis. In this study, using biochemical, structural and spectroscopic analyses, we evaluated the effects of native PLGA on temperature-dependent Aβ aggregation and its ability to protect cultured neurons from degeneration. Our results show that the rate of spontaneous Aβ1-42 aggregation increases with a rise in temperature from 27 to 40 °C and PLGA with 50:50 resomer potently inhibits Aβ aggregation at all temperatures, but the effect is more profound at 27 °C than at 40 °C. It appears that native PLGA, by interacting with the hydrophobic domain of Aβ1-42, prevents a conformational shift towards β-sheet structure, thus precluding the formation of Aβ aggregates. Additionally, PLGA triggers disassembly of matured Aβ1-42 fibers at a faster rate at 40 °C than at 27 °C. PLGA-treated Aβ samples can significantly enhance viability of cortical cultured neurons compared to neurons treated with Aβ alone by attenuating phosphorylation of tau protein. Injection of native PLGA is found to influence the breakdown/clearance of Aβ peptide in the brain. Collectively, these results suggest that PLGA nanoparticles can inhibit Aβ aggregation and trigger disassembly of Aβ aggregates at temperatures outside the physiological range and can protect neurons against Aβ-mediated toxicity thus validating its unique therapeutic potential in the treatment of AD pathology.
Collapse
Affiliation(s)
- Pallabi Sil Paul
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M8 Canada
| | - Jae-Young Cho
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, AB T6G 2M9 Canada
| | - Qi Wu
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M8 Canada
| | - Govindarajan Karthivashan
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M8 Canada
| | - Emily Grabovac
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, AB T6G 2M9 Canada
| | - Holger Wille
- Department of Biochemistry, Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M8 Canada
| | - Mariana Kulka
- Nanotechnology Research Centre, National Research Council Canada, Edmonton, AB T6G 2M9 Canada
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB T6G 2E1 Canada
| | - Satyabrata Kar
- Department of Medicine (Neurology), Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M8 Canada
- Departments of Medicine (Neurology) and Psychiatry, Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB T6G 2M8 Canada
| |
Collapse
|
29
|
Fateh Basharzad S, Hamidi M, Maleki A, Karami Z, Mohamadpour H, Reza Saghatchi Zanjani M. Polysorbate-coated mesoporous silica nanoparticles as an efficient carrier for improved rivastigmine brain delivery. Brain Res 2022; 1781:147786. [PMID: 35041841 DOI: 10.1016/j.brainres.2022.147786] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 11/11/2021] [Accepted: 01/10/2022] [Indexed: 12/16/2022]
Abstract
Targeted delivery of neurological therapeutic to the brain has been attracting more and more attention to the treatment of central nervous system (CNS) diseases. Nonetheless, the main obstacle in this road map is the existence of a blood-brain barrier (BBB) which limits the penetration efficiency of most CNS drugs into the brain parenchyma. This present investigation describes a facile synthetic strategy to prepare a highly biocompatible calcium-doped mesoporous silica nanoparticles (MSNs) functionalized by polysorbate-80 (PS) as targeting ligand to deliver rivastigmine (RV) into the brain via crossing the BBB. The developed nanosystem was characterized by scanning electron microscopy (SEM), Fourier transform infrared spectroscopy (FT-IR), transmission electron microscopy (TEM), Zeta potential, and N2-adsorption-desorption analysis. In vitro hemolysis studies were carried out to confirm the biocompatibility of the nanocarriers. Our in vivo studies in an animal model of rats showed that the RV-loaded nanosystem was able to enhance the brain-to-plasma concentration ratio, brain uptake clearance, and plasma elimination half-life of the drug compared to the free one drug following intravenous (IV) administration. The results revealed that functionalization of MSNs by PS is crucial to deliver RV into the brain, suggesting PS-functionalized MSNs could be an effective carrier to deliver RV to the brain while overcoming BBB.
Collapse
Affiliation(s)
- Samaneh Fateh Basharzad
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mehrdad Hamidi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.
| | - Aziz Maleki
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Zahra Karami
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran; Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hamed Mohamadpour
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
| | | |
Collapse
|
30
|
Ogbodo JO, Agbo CP, Njoku UO, Ogugofor MO, Egba SI, Ihim SA, Echezona AC, Brendan KC, Upaganlawar AB, Upasani CD. Alzheimer's Disease: Pathogenesis and Therapeutic Interventions. Curr Aging Sci 2022; 15:2-25. [PMID: 33653258 DOI: 10.2174/1874609814666210302085232] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/04/2020] [Accepted: 11/20/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Alzheimer's Disease (AD) is the most common cause of dementia. Genetics, excessive exposure to environmental pollutants, as well as unhealthy lifestyle practices are often linked to the development of AD. No therapeutic approach has achieved complete success in treating AD; however, early detection and management with appropriate drugs are key to improving prognosis. INTERVENTIONS The pathogenesis of AD was extensively discussed in order to understand the reasons for the interventions suggested. The interventions reviewed include the use of different therapeutic agents and approaches, gene therapy, adherence to healthy dietary plans (Mediterranean diet, Okinawan diet and MIND diet), as well as the use of medicinal plants. The potential of nanotechnology as a multidisciplinary and interdisciplinary approach in the design of nano-formulations of AD drugs and the use of Superparamagnetic Iron Oxide Nanoparticles (SPIONs) as theranostic tools for early detection of Alzheimer's disease were also discussed.
Collapse
Affiliation(s)
- John O Ogbodo
- Department of Science Laboratory Technology, University of Nigeria, Nsukka, Nigeria
| | - Chinazom P Agbo
- Department of Pharmaceutics, University of Nigeria, Nsukka, Nigeria
| | - Ugochi O Njoku
- Department of Biochemistry, University of Nigeria, Nsukka, Nigeria
| | | | - Simeon I Egba
- Department of Biochemistry, Michael Okpara University of Agriculture, Umudike, Nigeria
| | - Stella A Ihim
- Department of Pharmacology and Toxicology, University of Nigeria, Nsukka, Nigeria
| | | | | | - Aman B Upaganlawar
- Department of Pharmacology, Sureshdada Shriman\'s College of Pharmacy, New Dehli, India
| | | |
Collapse
|
31
|
Chen L, Zhou Z, Zhang Y, Pan J, Wang K, Wang HX. Near-infrared Irradiation Controlled Thermo-Switchable Polymeric Photosensitizer against β-Amyloid Fibrillation. J Mater Chem B 2022; 10:4832-4839. [DOI: 10.1039/d2tb00372d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Photothermal therapy (PTT) as an emerging paradigm toward degradation of amyloid-β (Aβ) aggregations has become an effective way of treating Alzheimer’s disease (AD). A promising PTT therapeutic option requires control...
Collapse
|
32
|
Nguyen NTT, Nguyen LM, Nguyen TTT, Nguyen TT, Nguyen DTC, Tran TV. Formation, antimicrobial activity, and biomedical performance of plant-based nanoparticles: a review. ENVIRONMENTAL CHEMISTRY LETTERS 2022; 20:2531-2571. [PMID: 35369682 PMCID: PMC8956152 DOI: 10.1007/s10311-022-01425-w] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/24/2022] [Indexed: 05/09/2023]
Abstract
Because many engineered nanoparticles are toxic, there is a need for methods to fabricate safe nanoparticles such as plant-based nanoparticles. Indeed, plant extracts contain flavonoids, amino acids, proteins, polysaccharides, enzymes, polyphenols, steroids, and reducing sugars that facilitate the reduction, formation, and stabilization of nanoparticles. Moreover, synthesizing nanoparticles from plant extracts is fast, safe, and cost-effective because it does not consume much energy, and non-toxic derivatives are generated. These nanoparticles have diverse and unique properties of interest for applications in many fields. Here, we review the synthesis of metal/metal oxide nanoparticles with plant extracts. These nanoparticles display antibacterial, antifungal, anticancer, and antioxidant properties. Plant-based nanoparticles are also useful for medical diagnosis and drug delivery.
Collapse
Affiliation(s)
- Ngoan Thi Thao Nguyen
- Institute of Environmental Technology and Sustainable Development, Nguyen Tat Thanh University, 298-300A Nguyen Tat Thanh, District 4, Ho Chi Minh City, 755414 Vietnam
- Department of Chemical Engineering and Processing, Nong Lam University, Thu Duc District, Ho Chi Minh City, 700000 Vietnam
| | - Luan Minh Nguyen
- Institute of Environmental Technology and Sustainable Development, Nguyen Tat Thanh University, 298-300A Nguyen Tat Thanh, District 4, Ho Chi Minh City, 755414 Vietnam
- Department of Chemical Engineering and Processing, Nong Lam University, Thu Duc District, Ho Chi Minh City, 700000 Vietnam
| | - Thuy Thi Thanh Nguyen
- Department of Chemical Engineering and Processing, Nong Lam University, Thu Duc District, Ho Chi Minh City, 700000 Vietnam
- Faculty of Science, Nong Lam University, Thu Duc District, Ho Chi Minh City, 700000 Vietnam
| | - Thuong Thi Nguyen
- Institute of Environmental Technology and Sustainable Development, Nguyen Tat Thanh University, 298-300A Nguyen Tat Thanh, District 4, Ho Chi Minh City, 755414 Vietnam
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, 298-300A Nguyen Tat Thanh, District 4, Ho Chi Minh City, 755414 Vietnam
| | - Duyen Thi Cam Nguyen
- Institute of Environmental Technology and Sustainable Development, Nguyen Tat Thanh University, 298-300A Nguyen Tat Thanh, District 4, Ho Chi Minh City, 755414 Vietnam
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, 298-300A Nguyen Tat Thanh, District 4, Ho Chi Minh City, 755414 Vietnam
| | - Thuan Van Tran
- Institute of Environmental Technology and Sustainable Development, Nguyen Tat Thanh University, 298-300A Nguyen Tat Thanh, District 4, Ho Chi Minh City, 755414 Vietnam
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, 298-300A Nguyen Tat Thanh, District 4, Ho Chi Minh City, 755414 Vietnam
| |
Collapse
|
33
|
Guo Y, Hu Z, Wang Z. Recent Advances in the Application Peptide and Peptoid in Diagnosis Biomarkers of Alzheimer's Disease in Blood. Front Mol Neurosci 2021; 14:778955. [PMID: 35002620 PMCID: PMC8733658 DOI: 10.3389/fnmol.2021.778955] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most common neurodegenerative diseases with irreversible damage of the brain and a continuous pathophysiological process. Early detection and accurate diagnosis are essential for the early intervention of AD. Precise detection of blood biomarkers related to AD could provide a shortcut to identifying early-stage patients before symptoms. In recent years, targeting peptides or peptoids have been chosen as recognition elements in nano-sensors or fluorescence detection to increase the targeting specificity, while peptide-based probes were also developed considering their specific advantages. Peptide-based sensors and probes have been developed according to different strategies, such as natural receptors, high-throughput screening, or artificial design for AD detection. This review will briefly summarize the recent developments and trends of AD diagnosis platforms based on peptide and peptoid as recognition elements and provide insights into the application of peptide and peptoid with different sources and characteristics in the diagnosis of AD biomarkers.
Collapse
Affiliation(s)
- Yuxin Guo
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhiyuan Hu
- CAS Key Laboratory of Standardization and Measurement for Nanotechnology, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
- School of Nanoscience and Technology, Sino-Danish College, University of Chinese Academy of Sciences, Beijing, China
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, China
| | - Zihua Wang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
34
|
Zhou Z, Gu YQ, Wang HX. Artificial Chiral Interfaces against Amyloid-β Peptide Aggregation: Research Progress and Challenges. ACS Chem Neurosci 2021; 12:4236-4248. [PMID: 34724384 DOI: 10.1021/acschemneuro.1c00544] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by an imbalance between the production and clearance of amyloid-β (Aβ) species. AD not only influences the life quality of the patients but also heavily burdens the families and society. Therefore, it is an urgent mission to research and develop some new anti-amyloid aggregation drugs. In recent years, there were research and development of engineered nanostructures as Aβ amyloid inhibitors have attracted extensive attention and become a new frontier in nanomedicine. The effects of nanostructural surface properties (e.g., morphology, charge, hydrophobicity) on inhibition of Aβ aggregation are modulated by adsorbed Aβ peptides. Nevertheless, chirality has been seldom considered in recognition of Aβ species and modulation of Aβ aggregations. Moreover, a more relevant question for chiral inhibitors is little known about the molecular mechanism of how to interface chiral effects Aβ targeting recognition and effective mitigation of amyloidosis at the molecular level. Herein, we review recent experimental and theoretical results acquired in the specific areas of artificial chiral nanostructure inhibitors. This article will be essential to provide a microlevel insight into the effects of chiral nanointerfaces on amyloidosis processes as well as the development of chiral inhibitor drugs against Aβ fibrillation.
Collapse
Affiliation(s)
- Zhe Zhou
- Department of Neurology, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - You-Quan Gu
- Department of Neurology, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Hang-Xing Wang
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| |
Collapse
|
35
|
Yan X, Pan Y, Ji L, Gu J, Hu Y, Xia Y, Li C, Zhou X, Yang D, Yu Y. Multifunctional Metal-Organic Framework as a Versatile Nanoplatform for Aβ Oligomer Imaging and Chemo-Photothermal Treatment in Living Cells. Anal Chem 2021; 93:13823-13834. [PMID: 34609144 DOI: 10.1021/acs.analchem.1c02459] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In view of the close association of β-amyloid oligomers (AβO) with the clinical development of Alzheimer's disease (AD) symptoms, it is urgent to design a promising sensing and therapeutic strategy that can target AβO for preventing or delaying the onset of AD. Herein, a core-shell nanocomposite CeONP-Res-PCM@ZIF-8/polydopamine (PDA) was synthesized through an in situ encapsulated strategy, in which resveratrol (Res), ceria nanoparticles (CeONPs), and PCM (tetradecanol) were embedded into the ZIF-8/PDA matrix via a water-based mild approach. Using the AβO aptamer, the ability of CeONP-Res-PCM@ZIF-8/PDA/Apt as the fluorescent sensing platform for AβO detection and intracellular imaging was demonstrated. The nanocomposite was high in Res loading (27.5%) and could be activated to release the encapsulated Res upon illumination with NIR through PCM regulation. Moreover, due to the synergetic interactions of PDA, CeONPs, and Res in one system, CeONP-Res-PCM@ZIF-8/PDA/Apt nanocomposites exhibited multifunctional effects on inhibiting Aβ aggregation, degrading Aβ fibrils, and alleviating Aβ-induced oxidative stress and neural apoptosis. These therapeutic effects could be enhanced under NIR irradiation by virtue of the excellent photothermal property of PDA. As far as we know, there is no report of using ZIF-8-based materials for simultaneous sensing and therapeutic applications. This work boosted the development of multifunctional nanoagents for biomedical research studies.
Collapse
Affiliation(s)
- Xueyan Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu 221004, Xuzhou, China
| | - Yixin Pan
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Affiliated with Shanghai Jiao Tong University School of Medicine, Ruijin Hospital, 197 Ruijin Er Road, Shanghai 200025, P. R. China
| | - Liang Ji
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu 221004, Xuzhou, China
| | - Jinyu Gu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu 221004, Xuzhou, China
| | - Yuanyuan Hu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu 221004, Xuzhou, China
| | - Yi Xia
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu 221004, Xuzhou, China
| | - Chenglin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu 221004, Xuzhou, China
| | - Xinguang Zhou
- Shenzhen NTEK Testing Technology Co., Ltd., Shenzhen 518000, Guangdong, P. R. China
| | - Dongzhi Yang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu 221004, Xuzhou, China
| | - Yanyan Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Jiangsu 221004, Xuzhou, China
| |
Collapse
|
36
|
Cui W, Fu W, Lin Y, Zhang T. Application of Nanomaterials in Neurodegenerative Diseases. Curr Stem Cell Res Ther 2021; 16:83-94. [PMID: 32213159 DOI: 10.2174/1574888x15666200326093410] [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: 11/25/2019] [Revised: 01/07/2020] [Accepted: 02/04/2020] [Indexed: 02/08/2023]
Abstract
Neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, and Huntington's disease are very harmful brain lesions. Due to the difficulty in obtaining therapeutic drugs, the best treatment for neurodegenerative diseases is often not available. In addition, the bloodbrain barrier can effectively prevent the transfer of cells, particles and macromolecules (such as drugs) in the brain, resulting in the failure of the traditional drug delivery system to provide adequate cellular structure repair and connection modes, which are crucial for the functional recovery of neurodegenerative diseases. Nanomaterials are designed to carry drugs across the blood-brain barrier for targets. Nanotechnology uses engineering materials or equipment to interact with biological systems at the molecular level to induce physiological responses through stimulation, response and target site interactions, while minimizing the side effects, thus revolutionizing the treatment and diagnosis of neurodegenerative diseases. Some magnetic nanomaterials play a role as imaging agents or nanoprobes for Magnetic Resonance Imaging to assist in the diagnosis of neurodegenerative diseases. Although the current research on nanomaterials is not as useful as expected in clinical applications, it achieves a major breakthrough and guides the future development direction of nanotechnology in the application of neurodegenerative diseases. This review briefly discusses the application and advantages of nanomaterials in neurodegenerative diseases. Data for this review were identified by searches of PubMed, and references from relevant articles published in English between 2015 and 2019 using the search terms "nanomaterials", "neurodegenerative diseases" and "blood-brain barrier".
Collapse
Affiliation(s)
- Weitong Cui
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Wei Fu
- Department of Neurosurgery, West China Hospital of Sichuan University, Chengdu, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Tianxu Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
37
|
Zhang L, Sun H, Zhao J, Lee J, Ee Low L, Gong L, Chen Y, Wang N, Zhu C, Lin P, Liang Z, Wei M, Ling D, Li F. Dynamic nanoassemblies for imaging and therapy of neurological disorders. Adv Drug Deliv Rev 2021; 175:113832. [PMID: 34146626 DOI: 10.1016/j.addr.2021.113832] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/07/2021] [Accepted: 06/11/2021] [Indexed: 02/07/2023]
Abstract
The past decades have witnessed an increased incidence of neurological disorders (NDs) such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, ischemic stroke, and epilepsy, which significantly lower patients' life quality and increase the economic and social burden. Recently, nanomedicines composed of imaging and/or therapeutic agents have been explored to diagnose and/or treat NDs due to their enhanced bioavailability, blood-brain barrier (BBB) permeability, and targeting capacity. Intriguingly, dynamic nanoassemblies self-assembled from functional nanoparticles to simultaneously interfere with multiple pathogenic substances and pathological changes, have been regarded as one of the foremost candidates to improve the diagnostic and therapeutic efficacy of NDs. To help readers better understand this emerging field, in this review, the pathogenic mechanism of different types of NDs is briefly introduced, then the functional nanoparticles used as building blocks in the construction of dynamic nanoassemblies for NDs theranostics are summarized. Furthermore, dynamic nanoassemblies that can actively cross the BBB to target brain lesions, sensitively and efficiently diagnose or treat NDs, and effectively promote neuroregeneration are highlighted. Finally, we conclude with our perspectives on the future development in this field.
Collapse
|
38
|
Pourhamzeh M, Joghataei MT, Mehrabi S, Ahadi R, Hojjati SMM, Fazli N, Nabavi SM, Pakdaman H, Shahpasand K. The Interplay of Tau Protein and β-Amyloid: While Tauopathy Spreads More Profoundly Than Amyloidopathy, Both Processes Are Almost Equally Pathogenic. Cell Mol Neurobiol 2021; 41:1339-1354. [PMID: 32696288 PMCID: PMC11448628 DOI: 10.1007/s10571-020-00906-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 06/15/2020] [Indexed: 11/29/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder, in which amyloid precursor protein (APP) misprocessing and tau protein hyperphosphorylation are well-established pathogenic cascades. Despite extensive considerations, the central mediator of neuronal cell death upon AD remains under debate. Therefore, we examined the direct interplay between tauopathy and amyloidopathy processes. We employed primary culture neurons and examined pathogenic P-tau and Aβ oligomers upon hypoxia treatment by immunofluorescence and immunoblotting. We observed both tauopathy and amyloidopathy processes upon the hypoxia condition. We also applied Aβ1-42 or P-tau onto primary cultured neurons. We overexpressed P-tau in SH-SY5Y cells and found Aβ accumulation. Furthermore, adult male rats received Aβ1-42 or pathogenic P-tau in the dorsal hippocampus and were examined for 8 weeks. Learning and memory performance, as well as anxiety behaviors, were assessed by Morris water maze and elevated plus-maze tests. Both Aβ1-42 and pathogenic P-tau significantly induced learning and memory deficits and enhanced anxiety behavior after treatment 2 weeks. Aβ administration induced robust tauopathy distribution in the cortex, striatum, and corpus callosum as well as CA1. On the other hand, P-tau treatment developed Aβ oligomers in the cortex and CA1 only. Our findings indicate that Aβ1-42 and pathogenic P-tau may induce each other and cause almost identical neurotoxicity in a time-dependent manner, while tauopathy seems to be more distributable than amyloidopathy.
Collapse
Affiliation(s)
- Mahsa Pourhamzeh
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Taghi Joghataei
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Soraya Mehrabi
- Department of Physiology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Ahadi
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Nasrin Fazli
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyed Massood Nabavi
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Pakdaman
- Brain Mapping Research Center, Department of Neurology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Koorosh Shahpasand
- Department of Brain and Cognitive Sciences, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
39
|
Hanif S, Muhammad P, Niu Z, Ismail M, Morsch M, Zhang X, Li M, Shi B. Nanotechnology‐Based Strategies for Early Diagnosis of Central Nervous System Disorders. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202100008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Sumaira Hanif
- Henan-Macquarie University Joint Centre for Biomedical Innovation School of Life Sciences Henan University Kaifeng Henan 475004 China
| | - Pir Muhammad
- Henan-Macquarie University Joint Centre for Biomedical Innovation School of Life Sciences Henan University Kaifeng Henan 475004 China
| | - Zheng Niu
- Province's Key Lab of Brain Targeted Bionanomedicine School of Pharmacy Henan University Kaifeng Henan 475004 China
| | - Muhammad Ismail
- Henan-Macquarie University Joint Centre for Biomedical Innovation School of Life Sciences Henan University Kaifeng Henan 475004 China
| | - Marco Morsch
- Department of Biomedical Sciences Macquarie University Centre for Motor Neuron Disease Research Macquarie University NSW 2109 Australia
| | - Xiaoju Zhang
- Department of Respiratory and Critical Care Medicine Henan Provincial People's Hospital Zhengzhou Henan 450003 China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine The Third Affiliated Hospital Sun Yat-sen University Guangzhou Guangdong 510630 China
| | - Bingyang Shi
- Department of Biomedical Sciences Faculty of Medicine & Health & Human Sciences Macquarie University NSW 2109 Australia
| |
Collapse
|
40
|
Ling TS, Chandrasegaran S, Xuan LZ, Suan TL, Elaine E, Nathan DV, Chai YH, Gunasekaran B, Salvamani S. The Potential Benefits of Nanotechnology in Treating Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5550938. [PMID: 34285915 PMCID: PMC8275379 DOI: 10.1155/2021/5550938] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 06/24/2021] [Indexed: 11/17/2022]
Abstract
Alzheimer's disease is a neurodegenerative disorder that is caused by the accumulation of beta-amyloid plaques in the brain. Currently, there is no definitive cure available to treat Alzheimer's disease. The available medication in the market has the ability to only slow down its progression. However, nanotechnology has shown its superiority that can be applied for medical usage and it has a great potential in the therapy of Alzheimer's disease, specifically in the disease diagnosis and providing an alternative approach to treat Alzheimer's disease. This is done by increasing the efficiency of drug delivery by penetrating and overcoming the blood-brain barrier. Having said that, there are limitations that need to be further investigated and researched in order to minimize the adverse effects and potential toxicity and to improve drug bioavailability. The recent advances in the treatment of Alzheimer's disease using nanotechnology include the regeneration of stem cells, nanomedicine, and neuroprotection. In this review, we will discuss the advancement of nanotechnology which helps in the diagnosis and treatment of neurodegenerative disorders such as Alzheimer's disease as well as its challenges.
Collapse
Affiliation(s)
- Tan Sook Ling
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 1, Jalan Puncak Menara Gading, Taman Connaught, 56000 Kuala Lumpur, Malaysia
| | - Shanthini Chandrasegaran
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 1, Jalan Puncak Menara Gading, Taman Connaught, 56000 Kuala Lumpur, Malaysia
| | - Low Zhi Xuan
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 1, Jalan Puncak Menara Gading, Taman Connaught, 56000 Kuala Lumpur, Malaysia
| | - Tong Li Suan
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 1, Jalan Puncak Menara Gading, Taman Connaught, 56000 Kuala Lumpur, Malaysia
| | - Elaine Elaine
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 1, Jalan Puncak Menara Gading, Taman Connaught, 56000 Kuala Lumpur, Malaysia
| | - Durrgashini Visva Nathan
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| | - Yam Hok Chai
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 1, Jalan Puncak Menara Gading, Taman Connaught, 56000 Kuala Lumpur, Malaysia
| | - Baskaran Gunasekaran
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, 1, Jalan Puncak Menara Gading, Taman Connaught, 56000 Kuala Lumpur, Malaysia
| | - Shamala Salvamani
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Bukit Jalil, 57000 Kuala Lumpur, Malaysia
| |
Collapse
|
41
|
Sanati M, Aminyavari S, Khodagholi F, Hajipour MJ, Sadeghi P, Noruzi M, Moshtagh A, Behmadi H, Sharifzadeh M. PEGylated superparamagnetic iron oxide nanoparticles (SPIONs) ameliorate learning and memory deficit in a rat model of Alzheimer's disease: Potential participation of STIMs. Neurotoxicology 2021; 85:145-159. [PMID: 34058247 DOI: 10.1016/j.neuro.2021.05.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 10/21/2022]
Abstract
The amyloid-beta (Aβ) fibrillation process seems to execute a principal role in the neuropathology of Alzheimer's disease (AD). Accordingly, novel therapeutic plans have concentrated on the inhibition or degradation of Aβ oligomers and fibrils. Biocompatible nanoparticles (NPs), e.g., gold and iron oxide NPs, take a unique capacity in redirecting Aβ fibrillation kinetics; nevertheless, their impacts on AD-related memory impairment have not been adequately evaluated in vivo. Here, we examined the effect of commercial PEGylated superparamagnetic iron oxide nanoparticles (SPIONs) on the learning and memory of an AD-animal model. The outcomes demonstrated the dose-dependent effect of SPIONs on Aβ fibrillation and learning and memory processes. In vitro and in vivo findings revealed that Low doses of SPIONs inhibited Aβ aggregation and ameliorated learning and memory deficit in the AD model, respectively. Enhanced level of hippocampal proteins, including brain-derived neurotrophic factor, BDNF, phosphorylated-cAMP response element-binding protein, p-CREB, and stromal interaction molecules, e.g., STIM1 and STIM2, were also observed. However, at high doses, SPIONs did not improve the detrimental impacts of Aβ fibrillation on spatial memory and hippocampal proteins expression. Overall, we revealed the potential capacity of SPIONs on retrieval of behavioral and molecular manifestations of AD in vivo, which needs further investigations to determine the mechanistic effect of SPIONs in the AD conundrum.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Samaneh Aminyavari
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Hajipour
- The Persian Gulf Biomedical Sciences Research Institute, Persian Gulf Marine Biotechnology Research Center, Bushehr University of Medical Sciences, Bushehr, 47263, Iran; Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| | - Payam Sadeghi
- Department of Plastic Surgery, Cleveland Clinic, OH, USA
| | - Marzieh Noruzi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| | - Aynaz Moshtagh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| | - Homayoon Behmadi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1416753955, Iran
| | - Mohammad Sharifzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 1416753955, Iran.
| |
Collapse
|
42
|
Tandon A, Singh SJ, Chaturvedi RK. Nanomedicine against Alzheimer's and Parkinson's Disease. Curr Pharm Des 2021; 27:1507-1545. [PMID: 33087025 DOI: 10.2174/1381612826666201021140904] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/06/2020] [Accepted: 08/18/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's and Parkinson's are the two most rampant neurodegenerative disorders worldwide. Existing treatments have a limited effect on the pathophysiology but are unable to fully arrest the progression of the disease. This is due to the inability of these therapeutic molecules to efficiently cross the blood-brain barrier. We discuss how nanotechnology has enabled researchers to develop novel and efficient nano-therapeutics against these diseases. The development of nanotized drug delivery systems has permitted an efficient, site-targeted, and controlled release of drugs in the brain, thereby presenting a revolutionary therapeutic approach. Nanoparticles are also being thoroughly studied and exploited for their role in the efficient and precise diagnosis of neurodegenerative conditions. We summarize the role of different nano-carriers and RNAi-conjugated nanoparticle-based therapeutics for their efficacy in pre-clinical studies. We also discuss the challenges underlying the use of nanomedicine with a focus on their route of administration, concentration, metabolism, and any toxic effects for successful therapeutics in these diseases.
Collapse
Affiliation(s)
- Ankit Tandon
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Sangh J Singh
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| | - Rajnish K Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow 226001, Uttar Pradesh, India
| |
Collapse
|
43
|
Zeng H, Qi Y, Zhang Z, Liu C, Peng W, Zhang Y. Nanomaterials toward the treatment of Alzheimer’s disease: Recent advances and future trends. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.01.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
44
|
Ahlawat J, Masoudi Asil S, Guillama Barroso G, Nurunnabi M, Narayan M. Application of carbon nano onions in the biomedical field: recent advances and challenges. Biomater Sci 2021; 9:626-644. [PMID: 33241797 DOI: 10.1039/d0bm01476a] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Carbon nano onions (CNOs) are carbonaceous nanostructures composed of multiple concentric shells of fullerenes. These cage-within-cage structures remain as one of the most exciting and fascinating carbon forms, along with graphene and its derivatives, due to their unique chemical and physical properties. Their exceptional biocompatibility and biosafety make them an attractive choice in a wide range of areas, including biological systems. This nanomaterial displays low toxicity, high dispersity in aqueous solutions (upon surface functionalization), and high pharmaceutical efficiency. Even though CNOs were discovered almost simultaneously along with carbon nanotubes (CNTs), their potential in biomedical applications still appears unrealized. The existence of CNOs is equally important, just like any other carbon nanostructures such as CNTs and fullerenes, because they display the ability of carbon to form another unique nanostructure with wonderful properties. Therefore, this mini-review summarizes recent studies geared towards developing CNOs for various biomedical applications, including sensing, drug delivery, imaging, tissue engineering, and as a therapeutic drug. It concludes by discussing other potential applications of this unique nanomaterial.
Collapse
Affiliation(s)
- Jyoti Ahlawat
- The Department of Chemistry & Biochemistry, The University of Texas at El Paso, TX: 79968, USA.
| | - Shima Masoudi Asil
- The Department of Environmental Science & Engineering, The University of Texas at El Paso, TX: 79968, USA
| | | | - Md Nurunnabi
- The Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas at El Paso, TX: 79968, USA
| | - Mahesh Narayan
- The Department of Chemistry & Biochemistry, The University of Texas at El Paso, TX: 79968, USA.
| |
Collapse
|
45
|
El-Hawwary SS, Abd Almaksoud HM, Saber FR, Elimam H, Sayed AM, El Raey MA, Abdelmohsen UR. Green-synthesized zinc oxide nanoparticles, anti-Alzheimer potential and the metabolic profiling of Sabal blackburniana grown in Egypt supported by molecular modelling. RSC Adv 2021; 11:18009-18025. [PMID: 35480186 PMCID: PMC9033216 DOI: 10.1039/d1ra01725j] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/19/2021] [Indexed: 01/18/2023] Open
Abstract
Nowadays, the biosynthesis of metal nanoparticles, particularly from plants, has been gaining interest. In the present work, the methanolic extracts of leaves, fruits, and the pollen grains of Sabal blackburniana were used for the green synthesis of ZnO nanoparticles, which were early detected by the formation of precipitate and further confirmed by UV-vis spectroscopy, transmission electron microscopy (TEM), X-ray diffraction (XRD), Fourier transform infra-red (FT-IR) spectroscopy and zeta potential (ZP) studies. TEM analysis has shown different shapes, predominantly irregular small spherical narrow particles included in hexagonal structures with size ranging from 2.23 to 49.56 nm. The XRD pattern confirmed that all synthesized ZnO nanoparticles have wurtzite hexagonal structure with crystalline nature. The average particle crystallite sizes were 47.21, 47.67 and 47.8 nm. The UV-visible spectra showed λ max in the range of 354-368 nm, which indicated the presence of ZnO nanoparticles. The FT-IR analysis identifies the characteristic functional groups present on the surface of ZnO nanoparticles. The ZP determination demonstrated that all representative selected synthesized ZnONPs exhibited acceptable ZP values of -30.8 to -45.9 mV, which indicated their good stability. In addition, the anti-Alzheimer potential of the selected extracts and ZnONPs was evaluated by assessing acetylcholinesterase inhibitory activity in vitro according to the improved Ellman method. The results indicated that the selected extracts have acetylcholinesterase inhibitory activity, and highlighted the promising inhibitory potential of green-synthesized ZnONPs using pollen grains, fruits and leaves extracts; they exhibited a potent inhibitory effect with IC50 values 63.78 ± 1.04651, 81.985 ± 3.075 and 117.95 ± 6.858 ng ml-1 respectively in comparison to donepezil as standard (IC50 = 50.7 ± 5.769 ng ml-1). Dereplication analysis of the selected extracts was performed using LC-MS; metabolic profiling revealed the presence of 41 compounds belonging to various chemical classes: flavonoids, steroidal saponins, terpenoids, alkaloids, lignans, sterols and fatty acids. Docking these dereplicated metabolites against the human AChE showed that the non-glycosylated flavonoid class of compounds was able to achieve interesting binding modes inside the AChE active site; they are suggested to be associated with the observed anti-AChE activity of Sabal extracts. This study is the first report to shed light on the acetylcholinesterase inhibitory activity of green-synthesized ZnO nanoparticles of S. blackburniana metabolites.
Collapse
Affiliation(s)
- Seham S El-Hawwary
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University Cairo 11562 Egypt
| | | | - Fatema R Saber
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University Cairo 11562 Egypt
| | - Hanan Elimam
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City Sadat City 32897 Egypt
| | - Ahmed M Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University Beni-Suef 62513 Egypt
| | - Mohamed A El Raey
- Phytochemistry and Plant systematics Department, Pharmaceutical Division, National Research Centre Dokki Cairo Egypt
| | - Usama Ramadan Abdelmohsen
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University Universities Zone, 61111 New Minia City Minia Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University 61519 Minia Egypt
| |
Collapse
|
46
|
Anand BG, Wu Q, Karthivashan G, Shejale KP, Amidian S, Wille H, Kar S. Mimosine functionalized gold nanoparticles (Mimo-AuNPs) suppress β-amyloid aggregation and neuronal toxicity. Bioact Mater 2021; 6:4491-4505. [PMID: 34027236 PMCID: PMC8131740 DOI: 10.1016/j.bioactmat.2021.04.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/18/2021] [Accepted: 04/19/2021] [Indexed: 12/27/2022] Open
Abstract
Evidence suggests that increased level/aggregation of beta-amyloid (Aβ) peptides initiate neurodegeneration and subsequent development of Alzheimer's disease (AD). At present, there is no effective treatment for AD. In this study, we reported the effects of gold nanoparticles surface-functionalized with a plant-based amino acid mimosine (Mimo-AuNPs), which is found to cross the blood-brain barrier, on the Aβ fibrillization process and toxicity. Thioflavin T kinetic assays, fluorescence imaging and electron microscopy data showed that Mimo-AuNPs were able to suppress the spontaneous and seed-induced Aβ1-42 aggregation. Spectroscopic studies, molecular docking and biochemical analyses further revealed that Mimo-AuNPs stabilize Aβ1-42 to remain in its monomeric state by interacting with the hydrophobic domain of Aβ1-42 (i.e., Lys16 to Ala21) there by preventing a conformational shift towards the β-sheet structure. Additionally, Mimo-AuNPs were found to trigger the disassembly of matured Aβ1-42 fibers and increased neuronal viability by reducing phosphorylation of tau protein and the production of oxyradicals. Collectively, these results reveal that the surface-functionalization of gold nanoparticles with mimosine can attenuate Aβ fibrillization and neuronal toxicity. Thus, we propose Mimo-AuNPs may be used as a potential treatment strategy towards AD-related pathologies. Mimosine functionalized with gold nanoparticles (Mimo-AuNPs) can cross blood-brain barrier. Mimo-AuNPs inhibit aggregation of Aβ peptides by interacting with its hydrophobic domain. Mimo-AuNPs can trigger disassembly of pre-aggregated Aβ fibers. Mimo-AuNPs can protect neurons against Aβ toxicity by attenuating intracellular signaling.
Collapse
Affiliation(s)
- Bibin G Anand
- Departments of Medicine and University of Alberta, Edmonton, Alberta, T6G 2M8, Canada.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| | - Qi Wu
- Departments of Medicine and University of Alberta, Edmonton, Alberta, T6G 2M8, Canada.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| | - Govindarajan Karthivashan
- Departments of Medicine and University of Alberta, Edmonton, Alberta, T6G 2M8, Canada.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| | - Kiran P Shejale
- Department of Metallurgical Engineering and Materials Science, Indian Institute of Technology Bombay, Powai, India
| | - Sara Amidian
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada.,Departments of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| | - Holger Wille
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada.,Departments of Biochemistry, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| | - Satyabrata Kar
- Departments of Medicine and University of Alberta, Edmonton, Alberta, T6G 2M8, Canada.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, T6G 2M8, Canada
| |
Collapse
|
47
|
Zhang Y, Ding C, Li C, Wang X. Advances in fluorescent probes for detection and imaging of amyloid-β peptides in Alzheimer's disease. Adv Clin Chem 2021; 103:135-190. [PMID: 34229849 DOI: 10.1016/bs.acc.2020.08.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Amyloid plaques generated from the accumulation of amyloid-β peptides (Aβ) fibrils in the brain is one of the main hallmarks of Alzheimer's disease (AD), a most common neurodegenerative disorder. Aβ aggregation can produce neurotoxic oligomers and fibrils, which has been widely accepted as the causative factor in AD pathogenesis. Accordingly, both soluble oligomers and insoluble fibrils have been considered as diagnostic biomarkers for AD. Among the existing analytical methods, fluorometry using fluorescent probes has exhibited promising potential in quantitative detection and imaging of both soluble and insoluble Aβ species, providing a valuable approach for the diagnosis and drug development of AD. In this review, the most recent advances in the fluorescent probes for soluble or insoluble Aβ aggregates are discussed in terms of design strategy, probing mechanism, and potential applications. In the end, future research directions of fluorescent probes for Aβ species are also proposed.
Collapse
Affiliation(s)
- Yunhua Zhang
- College of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, PR China
| | - Cen Ding
- College of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, PR China
| | - Changhong Li
- College of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, PR China
| | - Xiaohui Wang
- College of Chemistry and Molecular Engineering, Nanjing Tech University, Nanjing, PR China; State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing, PR China.
| |
Collapse
|
48
|
Altinoglu G, Adali T. Alzheimer's Disease Targeted Nano-Based Drug Delivery Systems. Curr Drug Targets 2021; 21:628-646. [PMID: 31744447 DOI: 10.2174/1389450120666191118123151] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 11/07/2019] [Accepted: 11/11/2019] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, and is part of a massive and growing health care burden that is destroying the cognitive function of more than 50 million individuals worldwide. Today, therapeutic options are limited to approaches with mild symptomatic benefits. The failure in developing effective drugs is attributed to, but not limited to the highly heterogeneous nature of AD with multiple underlying hypotheses and multifactorial pathology. In addition, targeted drug delivery to the central nervous system (CNS), for the diagnosis and therapy of neurological diseases like AD, is restricted by the challenges posed by blood-brain interfaces surrounding the CNS, limiting the bioavailability of therapeutics. Research done over the last decade has focused on developing new strategies to overcome these limitations and successfully deliver drugs to the CNS. Nanoparticles, that are capable of encapsulating drugs with sustained drug release profiles and adjustable physiochemical properties, can cross the protective barriers surrounding the CNS. Thus, nanotechnology offers new hope for AD treatment as a strong alternative to conventional drug delivery mechanisms. In this review, the potential application of nanoparticle based approaches in Alzheimer's disease and their implications in therapy is discussed.
Collapse
Affiliation(s)
- Gülcem Altinoglu
- Department of Biomedical Engineering, Faculty of Engineering, Near East University, P.O. Box: 99138, North Cyprus via Mersin 10, Turkey.,Tissue Engineering and Biomaterials Research Centre, Centre of Excellence, Near East University, P.O. Box: 99138, North Cyprus via Mersin 10 Turkey
| | - Terin Adali
- Department of Biomedical Engineering, Faculty of Engineering, Near East University, P.O. Box: 99138, North Cyprus via Mersin 10, Turkey.,Tissue Engineering and Biomaterials Research Centre, Centre of Excellence, Near East University, P.O. Box: 99138, North Cyprus via Mersin 10 Turkey
| |
Collapse
|
49
|
Ribarič S. Nanotechnology Therapy for Alzheimer's Disease Memory Impairment Attenuation. Int J Mol Sci 2021; 22:ijms22031102. [PMID: 33499311 PMCID: PMC7865945 DOI: 10.3390/ijms22031102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 11/16/2022] Open
Abstract
Currently, there is no cure for Alzheimer's disease (AD) in humans; treatment is symptomatic only. Aging of the population, together with an unhealthy diet and lifestyle, contribute to the steady, global increase of AD patients. This increase creates significant health, societal and economical challenges even for the most developed countries. AD progresses from an asymptomatic stage to a progressively worsening cognitive impairment. The AD cognitive impairment is underpinned by progressive memory impairment, an increasing inability to recall recent events, to execute recently planned actions, and to learn. These changes prevent the AD patient from leading an independent and fulfilling life. Nanotechnology (NT) enables a new, alternative pathway for development of AD treatment interventions. At present, the NT treatments for attenuation of AD memory impairment are at the animal model stage. Over the past four years, there has been a steady increase in publications of AD animal models with a wide variety of original NT treatment interventions, able to attenuate memory impairment. NT therapy development, in animal models of AD, is faced with the twin challenges of the nature of AD, a chronic impairment, unique to human, of the tau protein and A β peptides that regulate several key physiological brain processes, and the incomplete understanding of AD's aetiology. This paper reviews the state-of-the-art in NT based treatments for AD memory impairment in animal models and discusses the future work for translation to the successful treatment of AD cognitive impairment in human.
Collapse
Affiliation(s)
- Samo Ribarič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
50
|
Guckeisen T, Hosseinpour S, Peukert W. Effect of pH and urea on the proteins secondary structure at the water/air interface and in solution. J Colloid Interface Sci 2021; 590:38-49. [PMID: 33524719 DOI: 10.1016/j.jcis.2021.01.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 01/09/2023]
Abstract
HYPOTHESIS The secondary structure of proteins affects their functionality and performance in physiological environments or industrial applications. Change of the solution pH or the presence of protein denaturants are the main chemical means that can alter the secondary structure of proteins or lead to protein denaturation. Since proteins in the bulk solution and those residing at the solution/air interface experience different local environments, their response to chemical denaturation can be different. EXPERIMENTS We utilize circular dichroism and chiral/achiral sum frequency generation spectroscopy to study the secondary structure of selected proteins as a function of the solution pH or in the presence of 8 M urea in the bulk solution and at the solution/air interface, respectively. FINDINGS The liquid/air interface can enhance or decrease protein conformation stability. The change in the secondary structure of the surface adsorbed proteins in alkaline solutions occurs at pH values lower than those denaturing the studied proteins in the bulk solution. In contrast, while 8 M urea completely denatures the studied proteins in the bulk solution, the liquid/air interface prevents the urea-induced denaturation of the surface adsorbed proteins by limiting the access of urea to the hydrophobic side chains of proteins protruding to air.
Collapse
Affiliation(s)
- Tobias Guckeisen
- Institute of Particle Technology (LFG), Friedrich-Alexander-Universität-Erlangen-Nürnberg (FAU), Cauerstraße 4, 91058 Erlangen, Germany.
| | - Saman Hosseinpour
- Institute of Particle Technology (LFG), Friedrich-Alexander-Universität-Erlangen-Nürnberg (FAU), Cauerstraße 4, 91058 Erlangen, Germany.
| | - Wolfgang Peukert
- Institute of Particle Technology (LFG), Friedrich-Alexander-Universität-Erlangen-Nürnberg (FAU), Cauerstraße 4, 91058 Erlangen, Germany.
| |
Collapse
|