1
|
Masroor A, Zaidi N, Nabi F, Malik S, Zehra S, Arjmand F, Naseem N, Khan RH. Biophysical insight into anti-amyloidogenic nature of novel ionic Co(II)(phen)(H 2O) 4] +[glycinate] - chemotherapeutic drug candidate against human lysozyme aggregation. Biophys Chem 2024; 308:107214. [PMID: 38428228 DOI: 10.1016/j.bpc.2024.107214] [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: 11/21/2023] [Revised: 02/10/2024] [Accepted: 02/25/2024] [Indexed: 03/03/2024]
Abstract
In the recent past, there has been an ever-increasing interest in the search for metal-based therapeutic drug candidates for protein misfolding disorders (PMDs) particularly neurodegenerative disorders such as Alzheimer's, Parkinson's, Prion's diseases, and amyotrophic lateral sclerosis. Also, different amyloidogenic variants of human lysozyme (HL) are involved in hereditary systemic amyloidosis. Metallo-therapeutic agents are extensively studied as antitumor agents, however, they are relatively unexplored for the treatment of non-neuropathic amyloidoses. In this work, inhibition potential of a novel ionic cobalt(II) therapeutic agent (CoTA) of the formulation [Co(phen)(H2O)4]+[glycinate]- is evaluated against HL fibrillation. Various biophysical techniques viz., dye-binding assays, dynamic light scattering (DLS), differential scanning calorimetry (DSC), electron microscopy, and molecular docking experiments validate the proposed mechanism of inhibition of HL fibrillation by CoTA. The experimental corroborative results of these studies reveal that CoTA can suppress and slow down HL fibrillation at physiological temperature and pH. DLS and 1-anilino-8-naphthalenesulfonate (ANS) assay show that reduced fibrillation in the presence of CoTA is marked by a significant decrease in the size and hydrophobicity of the aggregates. Fluorescence quenching and molecular docking results demonstrate that CoTA binds moderately to the aggregation-prone region of HL (Kb = 6.6 × 104 M-1), thereby, inhibiting HL fibrillation. In addition, far-UV CD and DSC show that binding of CoTA to HL does not cause any change in the stability of HL. More importantly, CoTA attenuates membrane damaging effects of HL aggregates against RBCs. This study identifies inorganic metal complexes as a therapeutic intervention for systemic amyloidosis.
Collapse
Affiliation(s)
- Aiman Masroor
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, U.P 202002, India
| | - Nida Zaidi
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, U.P 202002, India
| | - Faisal Nabi
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, U.P 202002, India
| | - Sadia Malik
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, U.P 202002, India
| | - Siffeen Zehra
- Department of Chemistry, Aligarh Muslim University, Aligarh, U.P 202002, India
| | - Farukh Arjmand
- Department of Chemistry, Aligarh Muslim University, Aligarh, U.P 202002, India
| | - Nida Naseem
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, U.P 202002, India
| | - Rizwan Hasan Khan
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh, U.P 202002, India.
| |
Collapse
|
2
|
Fatima J, Siddique YH. Application of Nanocomposites and Nanoparticles in Treating Neurodegenerative Disorders. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1217-1233. [PMID: 38288843 DOI: 10.2174/0118715273283338240104112106] [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: 09/20/2023] [Revised: 12/02/2023] [Accepted: 12/12/2023] [Indexed: 08/28/2024]
Abstract
Neurodegenerative diseases represent a formidable global health challenge, affecting millions and imposing substantial burdens on healthcare systems worldwide. Conditions, like Alzheimer's, Parkinson's, and Huntington's diseases, among others, share common characteristics, such as neuronal loss, misfolded protein aggregation, and nervous system dysfunction. One of the major obstacles in treating these diseases is the presence of the blood-brain barrier, limiting the delivery of therapeutic agents to the central nervous system. Nanotechnology offers promising solutions to overcome these challenges. In Alzheimer's disease, NPs loaded with various compounds have shown remarkable promise in preventing amyloid-beta (Aβ) aggregation and reducing neurotoxicity. Parkinson's disease benefits from improved dopamine delivery and neuroprotection. Huntington's disease poses its own set of challenges, but nanotechnology continues to offer innovative solutions. The promising developments in nanoparticle-based interventions for neurodegenerative diseases, like amyotrophic lateral sclerosis (ALS) and multiple sclerosis (MS), have offered new avenues for effective treatment. Nanotechnology represents a promising frontier in biomedical research, offering tailored solutions to the complex challenges posed by neurodegenerative diseases. While much progress has been made, ongoing research is essential to optimize nanomaterial designs, improve targeting, and ensure biocompatibility and safety. Nanomaterials possess unique properties that make them excellent candidates for targeted drug delivery and neuroprotection. They can effectively bypass the blood-brain barrier, opening doors to precise drug delivery strategies. This review explores the extensive research on nanoparticles (NPs) and nanocomposites in diagnosing and treating neurodegenerative disorders. These nanomaterials exhibit exceptional abilities to target neurodegenerative processes and halt disease progression.
Collapse
Affiliation(s)
- Javeria Fatima
- Laboratory of Alternative Animal Models, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| | - Yasir Hasan Siddique
- Laboratory of Alternative Animal Models, Section of Genetics, Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, 202002, Uttar Pradesh, India
| |
Collapse
|
3
|
Abujamai J, Satar R, Ansari SA. Designing and Formulation of Nanocarriers for "Alzheimer's and Parkinson's" Early Detection and Therapy. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1251-1262. [PMID: 38351689 DOI: 10.2174/0118715273297024240201055550] [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: 12/06/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 08/28/2024]
Abstract
The potential of nanotechnology in advancing the diagnosis and treatment of neurodegenerative diseases is explored in this comprehensive literature review. The findings of these studies suggest that nanotechnology has the capacity to improve existing therapeutic approaches, create novel and safe compounds, and develop more precise imaging techniques and diagnostic methods for neurodegenerative diseases. With the emergence of the nanomedicine era, a new and innovative approach of diagnosing and treating these conditions has been introduced. Notably, the researchers' development of a nanocarrier drug delivery tool demonstrates immense potential compared to conventional therapy, as it maximizes therapeutic efficacy and minimizes undesirable as side effects.
Collapse
Affiliation(s)
- Jakleen Abujamai
- Medicine Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia
| | - Rukhsana Satar
- Division of Biochemistry, Department of Physiology and Biochemistry, Ibn Sina National College for Medical Studies, Jeddah 22421, Saudi Arabia
| | - Shakeel Ahmed Ansari
- Department of Biochemistry, Medicine Program, Batterjee Medical College, Jeddah 21442, Saudi Arabia
| |
Collapse
|
4
|
Aricov L, Precupas A, Tudose M, Baltag D, Trică B, Sandu R, Leonties AR. Trametes versicolor laccase activity modulated by the interaction with gold nanoparticles. ENVIRONMENTAL RESEARCH 2023; 237:116920. [PMID: 37597828 DOI: 10.1016/j.envres.2023.116920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/08/2023] [Accepted: 08/17/2023] [Indexed: 08/21/2023]
Abstract
In this study, the impact of gold nanoparticles (AuNPs) on the structure and activity of laccase from Trametes versicolor (Lc) was described. Fluorescence experiments revealed that AuNPs efficiently quench Lc's tryptophan fluorescence by a static and dynamic process. By using differential scanning microcalorimetry and circular dichroism spectroscopy, it was determined how the concentration of nanoparticles and the composition of the medium affected the secondary structure of Lc. The data revealed that upon binding with AuNPs, conformational changes take place mainly in presence of high amounts of nanoparticles. The complex kinetic analysis unveiled the Lc activity enhancement at low concentrations of AuNPs as opposed to the concentrated regime, where it can be reduced by up to 55%. The Michaelis-Menten tests highlighted that the activity of the biocatalyst is closely related to the concentration of AuNPs, while the Selwyn analysis demonstrated that even in a concentrated regime of Lc it is not deactivated regardless of the amount of AuNPs added. The thermal parameters improved by twofold in the presence of low AuNPs concentration, whereas the activation energy increased with AuNPs content, implying that not all collisions are beneficial to the enzyme structure. The effect of AuNPs on the decomposition of a recalcitrant dye (naphthol green B, NG) by Lc was also evaluated, and the Michaelis-Menten model revealed that only the high AuNPs content influenced negatively the Lc activity. The isothermal titration calorimetry revealed that hydrogen bonds are the main intermolecular forces between Lc and AuNPs, while electrostatic interactions are responsible for NG adsorption to AuNPs. The results of the docking analysis show the binding of NG near the copper T1 site of Lc with hydrogen bonds, electrostatic and hydrophobic interactions. The findings of this work provide important knowledge for laccase-based bio-nanoconjugates and their use in the field of environmental remediation.
Collapse
Affiliation(s)
- Ludmila Aricov
- "Ilie Murgulescu" Institute of Physical Chemistry, Romanian Academy, Spl. Independentei 202, 060021, Bucharest, Romania
| | - Aurica Precupas
- "Ilie Murgulescu" Institute of Physical Chemistry, Romanian Academy, Spl. Independentei 202, 060021, Bucharest, Romania.
| | - Madalina Tudose
- "Ilie Murgulescu" Institute of Physical Chemistry, Romanian Academy, Spl. Independentei 202, 060021, Bucharest, Romania
| | - Dragos Baltag
- Department of Physical Chemistry, Faculty of Chemistry, University of Bucharest, Bd. Elisabeta 4-12, 030018, Bucharest, Romania; National Institute for Research and Development in Chemistry and Petrochemistry - ICECHIM, Spl. Independentei 202, 060021, Bucharest, Romania
| | - Bogdan Trică
- National Institute for Research and Development in Chemistry and Petrochemistry - ICECHIM, Spl. Independentei 202, 060021, Bucharest, Romania
| | - Romica Sandu
- "Ilie Murgulescu" Institute of Physical Chemistry, Romanian Academy, Spl. Independentei 202, 060021, Bucharest, Romania
| | - Anca Ruxandra Leonties
- "Ilie Murgulescu" Institute of Physical Chemistry, Romanian Academy, Spl. Independentei 202, 060021, Bucharest, Romania.
| |
Collapse
|
5
|
Sisubalan N, Shalini R, Ramya S, Sivamaruthi BS, Chaiyasut C. Recent advances in nanomaterials for neural applications: opportunities and challenges. Nanomedicine (Lond) 2023; 18:1979-1994. [PMID: 38078433 DOI: 10.2217/nnm-2023-0261] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023] Open
Abstract
Nanomedicines are promising for delivering drugs to the central nervous system, though their precision is still being improved. Fortifying nanoparticles with vital molecules can interact with the blood-brain barrier, enabling access to brain tissue. This study summarizes recent advances in nanomedicine to treat neurological complications. The integration of nanotechnology into cell biology aids in the study of brain cells' interactions. Magnetic microhydrogels have exhibited superior neuron activation compared with superparamagnetic iron oxide nanoparticles and hold promise for neuropsychiatric disorders. Nanomaterials have shown notable results, such as tackling neurodegenerative diseases by hindering harmful protein buildup and regulating cellular processes. However, further studies of the safety and effectiveness of nanoparticles in managing neurological diseases and disorders are still required.
Collapse
Affiliation(s)
- Natarajan Sisubalan
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Ramadoss Shalini
- Department of Botany, Bishop Heber College (Autonomous), Affiliated to Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620017, India
| | - Sakthivel Ramya
- Department of Botany, Bishop Heber College (Autonomous), Affiliated to Bharathidasan University, Tiruchirappalli, Tamil Nadu, 620017, India
| | - Bhagavathi Sundaram Sivamaruthi
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand
| |
Collapse
|
6
|
Dinçer HA, Ağıldere AM, Gökçay D. T1 relaxation time is prolonged in healthy aging: a whole brain study. Turk J Med Sci 2023; 53:675-684. [PMID: 37476907 PMCID: PMC10387954 DOI: 10.55730/1300-0144.5630] [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: 05/31/2022] [Accepted: 01/07/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND : Measurement of tissue characteristics such as the longitudinal relaxation time (T1) provides complementary information to the volumetric and surface based structural analyses. We aimed to investigate T1 relaxation time characteristics in healthy aging via an exploratory design in the whole brain. The data processing pipeline was designed to minimize errors related to aging effects such as atrophy. METHODS Sixty healthy participants underwent MRI scanning (28 F, 32 M, age range: 18-78, 30 young and 30 old) in November 2017-March 2018 at the Bilkent University UMRAM Center. Four images with varying flip angles with FLASH (fast low angle shot magnetic resonance imaging) sequence and a high-resolution structural image with MP-RAGE (Magnetization Prepared - RApid Gradient Echo) were acquired. T1 relaxation times of the entire brain were mapped by using the region of interest (ROI) based method on 134 brain areas in young and old populations. RESULTS T1 prolongation was observed in various subcortical (bilateral hippocampus, caudate and thalamus) and cortical brain structures (bilateral precentral gyrus, bilateral middle frontal gyrus, bilateral supplementary motor area (SMA), left middle occipital gyrus, bilateral postcentral gyrus and bilateral Heschl's gyrus) as well as cerebellar regions (GM regions of cerebellum: bilateral cerebellum III, cerebellum IV V, cerebellum X, cerebellar vermis u 4 5, cerebellar vermis u 9 and WM cerebellar regions: left cerebellum IX, bilateral cerebellum X and cerebellar vermis u 4 5). DISCUSSION T1 mapping provides a practical quantitative MRI (qMRI) methodology for studying the tissue characteristics in healthy aging. T1 values are significantly increased in the aging group among half of the studied ROIs (57 ROIs out of 134).
Collapse
Affiliation(s)
- Hayriye Aktaş Dinçer
- Department of Biomedical Engineering, Institute of Natural and Applied Sciences, Middle East Technical University, Ankara, Turkey
| | | | - Didem Gökçay
- Department of Medical Informatics, Informatics Institute, Middle East Technical University, Ankara, Turkey
| |
Collapse
|
7
|
Dendrimers in Neurodegenerative Diseases. Processes (Basel) 2023. [DOI: 10.3390/pr11020319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Neurodegenerative diseases (NDs), such as Parkinson’s Disease (PD), Alzheimer’s Disease (AD), Multiple Sclerosis (MS) and amyotrophic lateral sclerosis (ALS), are characterized by progressive loss of structure or function of neurons. Current therapies for NDs are only symptomatic and long-term ineffective. This challenge has promoted the development of new therapies against relevant targets in these pathologies. In this review, we will focus on the most promising therapeutic approaches based on dendrimers (DDs) specially designed for the treatment and diagnosis of NDs. DDs are well-defined polymeric structures that provide a multifunctional platform for developing different nanosystems for a myriad of applications. DDs have been proposed as interesting drug delivery systems with the ability to cross the blood–brain barrier (BBB) and increase the bioavailability of classical drugs in the brain, as well as genetic material, by reducing the synthesis of specific targets, as β-amyloid peptide. Moreover, DDs have been shown to be promising anti-amyloidogenic systems against amyloid-β peptide (Aβ) and Tau aggregation, powerful agents for blocking α-synuclein (α-syn) fibrillation, exhibit anti-inflammatory properties, promote cellular uptake to certain cell types, and are potential tools for ND diagnosis. In summary, DDs have emerged as promising alternatives to current ND therapies since they may limit the extent of damage and provide neuroprotection to the affected tissues.
Collapse
|
8
|
Ashraf H, Cossu D, Ruberto S, Noli M, Jasemi S, Simula ER, Sechi LA. Latent Potential of Multifunctional Selenium Nanoparticles in Neurological Diseases and Altered Gut Microbiota. MATERIALS (BASEL, SWITZERLAND) 2023; 16:699. [PMID: 36676436 PMCID: PMC9862321 DOI: 10.3390/ma16020699] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 12/31/2022] [Accepted: 01/06/2023] [Indexed: 06/17/2023]
Abstract
Neurological diseases remain a major concern due to the high world mortality rate and the absence of appropriate therapies to cross the blood-brain barrier (BBB). Therefore, the major focus is on the development of such strategies that not only enhance the efficacy of drugs but also increase their permeability in the BBB. Currently, nano-scale materials seem to be an appropriate approach to treating neurological diseases based on their drug-loading capacity, reduced toxicity, targeted delivery, and enhanced therapeutic effect. Selenium (Se) is an essential micronutrient and has been of remarkable interest owing to its essential role in the physiological activity of the nervous system, i.e., signal transmission, memory, coordination, and locomotor activity. A deficiency of Se leads to various neurological diseases such as Parkinson's disease, epilepsy, and Alzheimer's disease. Therefore, owing to the neuroprotective role of Se (selenium) nanoparticles (SeNPs) are of particular interest to treat neurological diseases. To date, many studies investigate the role of altered microbiota with neurological diseases; thus, the current review focused not only on the recent advancement in the field of nanotechnology, considering SeNPs to cure neurological diseases, but also on investigating the potential role of SeNPs in altered microbiota.
Collapse
Affiliation(s)
- Hajra Ashraf
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Davide Cossu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Stefano Ruberto
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Marta Noli
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Seyedesomaye Jasemi
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Elena Rita Simula
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Leonardo A. Sechi
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Complex Structure of Microbiology and Virology, AOU Sassari, 07100 Sassari, Italy
| |
Collapse
|
9
|
Ashraf H, Solla P, Sechi LA. Current Advancement of Immunomodulatory Drugs as Potential Pharmacotherapies for Autoimmunity Based Neurological Diseases. Pharmaceuticals (Basel) 2022; 15:ph15091077. [PMID: 36145298 PMCID: PMC9504155 DOI: 10.3390/ph15091077] [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: 07/27/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Dramatic advancement has been made in recent decades to understand the basis of autoimmunity-mediated neurological diseases. These diseases create a strong influence on the central nervous system (CNS) and the peripheral nervous system (PNS), leading to various clinical manifestations and numerous symptoms. Multiple sclerosis (MS) is the most prevalent autoimmune neurological disease while NMO spectrum disorder (NMOSD) is less common. Furthermore, evidence supports the presence of autoimmune mechanisms contributing to the pathogenesis of amyotrophic lateral sclerosis (ALS), which is a neurodegenerative disorder characterized by the progressive death of motor neurons. Additionally, autoimmunity is believed to be involved in the basis of Alzheimer’s and Parkinson’s diseases. In recent years, the prevalence of autoimmune-based neurological disorders has been elevated and current findings strongly suggest the role of pharmacotherapies in controlling the progression of autoimmune diseases. Therefore, this review focused on the current advancement of immunomodulatory drugs as novel approaches in the management of autoimmune neurological diseases and their future outlook.
Collapse
Affiliation(s)
- Hajra Ashraf
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Paolo Solla
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Leonardo Atonio Sechi
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
- Complex Structure of Microbology and Virology, AOU Sassari, 07100 Sassari, Italy
- Correspondence:
| |
Collapse
|
10
|
Zhang Y, Zou Z, Liu S, Miao S, Liu H. Nanogels as Novel Nanocarrier Systems for Efficient Delivery of CNS Therapeutics. Front Bioeng Biotechnol 2022; 10:954470. [PMID: 35928954 PMCID: PMC9343834 DOI: 10.3389/fbioe.2022.954470] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
Nanogels have come out as a great potential drug delivery platform due to its prominently high colloidal stability, high drug loading, core-shell structure, good permeation property and can be responsive to environmental stimuli. Such nanoscopic drug carriers have more excellent abilities over conventional nanomaterials for permeating to brain parenchyma in vitro and in vivo. Nanogel-based system can be nanoengineered to bypass physiological barriers via non-invasive treatment, rendering it a most suitable platform for the management of neurological conditions such as neurodegenerative disorders, brain tumors, epilepsy and ischemic stroke, etc. Therapeutics of central nervous system (CNS) diseases have shown marked limited site-specific delivery of CNS by the poor access of various drugs into the brain, due to the presences of the blood-brain barrier (BBB) and blood-cerebrospinal fluid barrier (BCSFB). Hence, the availability of therapeutics delivery strategies is considered as one of the most major challenges facing the treatment of CNS diseases. The primary objective of this review is to elaborate the newer advances of nanogel for CNS drugs delivery, discuss the early preclinical success in the field of nanogel technology and highlight different insights on its potential neurotoxicity.
Collapse
Affiliation(s)
| | | | | | | | - Haiyan Liu
- Department of Anatomy, College of Basic Medicine Sciences, Jilin University, Changchun, China
| |
Collapse
|
11
|
Improvement of synaptic plasticity by nanoparticles and the related mechanisms: Applications and prospects. J Control Release 2022; 347:143-163. [PMID: 35513209 DOI: 10.1016/j.jconrel.2022.04.049] [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: 01/26/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/20/2022]
Abstract
Synaptic plasticity is an important basis of learning and memory and participates in brain network remodelling after different types of brain injury (such as that caused by neurodegenerative diseases, cerebral ischaemic injury, posttraumatic stress disorder (PTSD), and psychiatric disorders). Therefore, improving synaptic plasticity is particularly important for the treatment of nervous system-related diseases. With the rapid development of nanotechnology, increasing evidence has shown that nanoparticles (NPs) can cross the blood-brain barrier (BBB) in different ways, directly or indirectly act on nerve cells, regulate synaptic plasticity, and ultimately improve nerve function. Therefore, to better elucidate the effect of NPs on synaptic plasticity, we review evidence showing that NPs can improve synaptic plasticity by regulating different influencing factors, such as neurotransmitters, receptors, presynaptic membrane proteins and postsynaptic membrane proteins, and further discuss the possible mechanism by which NPs improve synaptic plasticity. We conclude that NPs can improve synaptic plasticity and restore the function of damaged nerves by inhibiting neuroinflammation and oxidative stress, inducing autophagy, and regulating ion channels on the cell membrane. By reviewing the mechanism by which NPs regulate synaptic plasticity and the applications of NPs for the treatment of neurological diseases, we also propose directions for future research in this field and provide an important reference for follow-up research.
Collapse
|
12
|
Ferdosh S, Banerjee S, Singh J, Barat C. Amyloid protein-induced sequestration of the eukaryotic ribosome: effect of stoichiometry and polyphenolic inhibitors. FEBS Lett 2022; 596:1190-1202. [PMID: 35114013 DOI: 10.1002/1873-3468.14308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/12/2022] [Accepted: 01/22/2022] [Indexed: 11/07/2022]
Abstract
Alzheimer's disease (AD) is characterized by the appearance of neurofibrillary tangles comprising of the Tau protein and aggregation of amyloid-β peptides (Aβ 1-40 and Aβ 1-42). A concomitant loss of the ribosomal population is also observed in AD-affected neurons. Our studies demonstrate that, similarly to Tau protein aggregation, in vitro aggregation of Aβ peptides in the vicinity of the yeast 80S ribosome can induce co-aggregation of ribosomal components. The RNA-stimulated aggregation of Aβ peptides and the Tau-K18 variant is dependent on the RNA:protein stoichiometric ratio. A similar effect of stoichiometry is also observed on the ribosome-protein co-aggregation process. Polyphenolic inhibitors of amyloid aggregation, such as rosmarinic acid and myricetin, inhibit RNA-stimulated Aβ and Tau-K18 aggregation and can mitigate the co-aggregation of ribosomal components.
Collapse
Affiliation(s)
- Sehnaz Ferdosh
- Department of Biotechnology, St. Xavier's College, Kolkata, India
| | - Senjuti Banerjee
- Department of Biotechnology, St. Xavier's College, Kolkata, India
| | - Jayshree Singh
- Department of Biotechnology, St. Xavier's College, Kolkata, India
| | - Chandana Barat
- Department of Biotechnology, St. Xavier's College, Kolkata, India
| |
Collapse
|
13
|
Low KJY, Venkatraman A, Mehta JS, Pervushin K. Molecular mechanisms of amyloid disaggregation. J Adv Res 2022; 36:113-132. [PMID: 35127169 PMCID: PMC8799873 DOI: 10.1016/j.jare.2021.05.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 04/13/2021] [Accepted: 05/16/2021] [Indexed: 12/17/2022] Open
Abstract
Importance of disaggregation mechanism and innate disaggregation in living systems. Different types and mechanism of disaggregation reported in literature. Structural details of the interactions and the disaggregation mechanisms. Amyloid disaggregation in protein aggregation disorders as a potential treatment. Proposed amyloid disaggregation mechanism of an ATP-independent chaperone (L-PGDS).
Introduction Protein aggregation and deposition of uniformly arranged amyloid fibrils in the form of plaques or amorphous aggregates is characteristic of amyloid diseases. The accumulation and deposition of proteins result in toxicity and cause deleterious effects on affected individuals known as amyloidosis. There are about fifty different proteins and peptides involved in amyloidosis including neurodegenerative diseases and diseases affecting vital organs. Despite the strenuous effort to find a suitable treatment option for these amyloid disorders, very few compounds had made it to unsuccessful clinical trials. It has become a compelling challenge to understand and manage amyloidosis with the increased life expectancy and ageing population. Objective While most of the currently available literature and knowledge base focus on the amyloid inhibitory mechanism as a treatment option, it is equally important to organize and understand amyloid disaggregation strategies. Disaggregation strategies are important and crucial as they are present innately functional in many living systems and dissolution of preformed amyloids may provide a direct benefit in many pathological conditions. In this review, we have compiled the known amyloid disaggregation mechanism, interactions, and possibilities of using disaggregases as a treatment option for amyloidosis. Methods We have provided the structural details using protein-ligand docking models to visualize the interaction between these disaggregases with amyloid fibrils and their respective proposed amyloid disaggregation mechanisms. Results After reviewing and comparing the different amyloid disaggregase systems and their proposed mechanisms, we presented two different hypotheses for ATP independent disaggregases using L-PGDS as a model. Conclusion Finally, we have highlighted the importance of understanding the underlying disaggregation mechanisms used by these chaperones and organic compounds before the implementation of these disaggregases as a potential treatment option for amyloidosis.
Collapse
|
14
|
Jiang P, Gan M, Yen SH, Dickson DW. Nanoparticles With Affinity for α-Synuclein Sequester α-Synuclein to Form Toxic Aggregates in Neurons With Endolysosomal Impairment. Front Mol Neurosci 2021; 14:738535. [PMID: 34744624 PMCID: PMC8565355 DOI: 10.3389/fnmol.2021.738535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/21/2021] [Indexed: 11/13/2022] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases. It is characterized pathologically by the aggregation of α-synuclein (αS) in the form of Lewy bodies and Lewy neurites. A major challenge in PD therapy is poor efficiency of drug delivery to the brain due to the blood-brain barrier (BBB). For this reason, nanomaterials, with significant advantages in drug delivery, have gained attention. On the other hand, recent studies have shown that nanoparticles can promote αS aggregation in salt solution. Therefore, we tested if nanoparticles could have the same effect in cell models. We found that nanoparticle can induce cells to form αS inclusions as shown in immunocytochemistry, and detergent-resistant αS aggregates as shown in biochemical analysis; and nanoparticles of smaller size can induce more αS inclusions. Moreover, the induction of αS inclusions is in part dependent on endolysosomal impairment and the affinity of αS to nanoparticles. More importantly, we found that the abnormally high level of endogenous lysosomotropic biomolecules (e.g., sphingosine), due to impairing the integrity of endolysosomes could be a determinant factor for the susceptibility of cells to nanoparticle-induced αS aggregation; and deletion of GBA1 gene to increase the level of intracellular sphingosine can render cultured cells more susceptible to the formation of αS inclusions in response to nanoparticle treatment. Ultrastructural examination of nanoparticle-treated cells revealed that the induced inclusions contained αS-immunopositive membranous structures, which were also observed in inclusions seeded by αS fibrils. These results suggest caution in the use of nanoparticles in PD therapy. Moreover, this study further supports the role of endolysosomal impairment in PD pathogenesis and suggests a possible mechanism underlying the formation of membrane-associated αS pathology.
Collapse
Affiliation(s)
- Peizhou Jiang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Ming Gan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, United States
| | - Shu-Hui Yen
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
15
|
Neuroprotective effect of bromelain in 6-hydroxydopamine induced in vitro model of Parkinson's disease. Mol Biol Rep 2021; 48:7711-7717. [PMID: 34643923 DOI: 10.1007/s11033-021-06779-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/20/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND This study was designed to investigate the neuroprotective effects of bromelain, which is known to have anti-oxidant and anti-inflammatory properties, against the neurotoxicity (induced by 6-OHDA) in SH-SY5Y cells. METHODS AND RESULTS To establish Parkinson's Disease (PD) model in cell culture conditions, SH-SY5Y cells were exposed to 200 µM 6-OHDA for 1 day. Prior to 6-OHDA treatment, SH-SY5Y cells had been pre-treated with bromelain (25 µg/mL, 50 µg/mL, 75 µg/mL and 100 µg/mL). After 1 day, cell viability was determined with the 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) and lactate dehydrogenase (LDH) assays. Oxidative stress was assessed with total antioxidant capacity (TAC), total oxidant status (TOS), glutathione reductase (GR) and malondialdehyde (MDA) analyses. The effect of the bromelain in SH-SY5Ycells was also examined by 4',6-diamidino-2-phenylindole (DAPI) staining. We found that 6-OHDA increased LDH leakage, and cellular apoptosis in SH-SY5Y cells. 6-OHDA aggravated oxidative stress by increasing TOS, MDA and GR and eventually promoted apoptosis in SH-SY5Y cells, while pretreatment with bromelain attenuated these toxic effects of 6-OHDA. CONCLUSIONS These findings indicated that bromelain, with its neuroprotective features can be useful for neuroprotection in PD.
Collapse
|
16
|
Andrikopoulos N, Song Z, Wan X, Douek AM, Javed I, Fu C, Xing Y, Xin F, Li Y, Kakinen A, Koppel K, Qiao R, Whittaker AK, Kaslin J, Davis TP, Song Y, Ding F, Ke PC. Inhibition of Amyloid Aggregation and Toxicity with Janus Iron Oxide Nanoparticles. CHEMISTRY OF MATERIALS : A PUBLICATION OF THE AMERICAN CHEMICAL SOCIETY 2021; 33:6484-6500. [PMID: 34887621 PMCID: PMC8651233 DOI: 10.1021/acs.chemmater.1c01947] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Amyloid aggregation is a ubiquitous form of protein misfolding underlying the pathologies of Alzheimer's disease (AD), Parkinson's disease (PD) and type 2 diabetes (T2D), three primary forms of human amyloid diseases. While much has been learned about the origin, diagnosis and management of these neurological and metabolic disorders, no cure is currently available due in part to the dynamic and heterogeneous nature of the toxic oligomers induced by amyloid aggregation. Here we synthesized beta casein-coated iron oxide nanoparticles (βCas IONPs) via a BPA-P(OEGA-b-DBM) block copolymer linker. Using a thioflavin T kinetic assay, transmission electron microscopy, Fourier transform infrared spectroscopy, discrete molecular dynamics simulations and cell viability assays, we examined the Janus characteristics and the inhibition potential of βCas IONPs against the aggregation of amyloid beta (Aβ), alpha synuclein (αS) and human islet amyloid polypeptide (IAPP) which are implicated in the pathologies of AD, PD and T2D. Incubation of zebrafish embryos with the amyloid proteins largely inhibited hatching and elicited reactive oxygen species, which were effectively rescued by the inhibitor. Furthermore, Aβ-induced damage to mouse brain was mitigated in vivo with the inhibitor. This study revealed the potential of Janus nanoparticles as a new nanomedicine against a diverse range of amyloid diseases.
Collapse
Affiliation(s)
- Nicholas Andrikopoulos
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Zhiyuan Song
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Xulin Wan
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Food Science, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing, 400715, China
| | - Alon M. Douek
- Australian Regenerative Medicine Institute, Monash University, 15 Innovation Walk, Clayton, VIC 3800, Australia
| | - Ibrahim Javed
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane Qld 4072, Australia
| | - Changkui Fu
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane Qld 4072, Australia
| | - Yanting Xing
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Fangyun Xin
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- School of Science, Dalian Maritime University, Dalian 116026, China
| | - Yuhuan Li
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, 200032, China
| | - Aleksandr Kakinen
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane Qld 4072, Australia
| | - Kairi Koppel
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Ruirui Qiao
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane Qld 4072, Australia
| | - Andrew K. Whittaker
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane Qld 4072, Australia
| | - Jan Kaslin
- Australian Regenerative Medicine Institute, Monash University, 15 Innovation Walk, Clayton, VIC 3800, Australia
| | - Thomas P. Davis
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane Qld 4072, Australia
- Corresponding Authors: Thomas P. Davis: ; Yang Song, ; Feng Ding: ; Pu Chun Ke:
| | - Yang Song
- Key Laboratory of Luminescence Analysis and Molecular Sensing, Ministry of Education, College of Pharmaceutical Sciences, Southwest University, 2 Tiansheng Rd, Beibei District, Chongqing 400715, China
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
- Corresponding Authors: Thomas P. Davis: ; Yang Song, ; Feng Ding: ; Pu Chun Ke:
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
- Corresponding Authors: Thomas P. Davis: ; Yang Song, ; Feng Ding: ; Pu Chun Ke:
| | - Pu Chun Ke
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane Qld 4072, Australia
- The GBA National Institute for Nanotechnology Innovation, 136 Kaiyuan Avenue, Guangzhou, 510700, China
- Corresponding Authors: Thomas P. Davis: ; Yang Song, ; Feng Ding: ; Pu Chun Ke:
| |
Collapse
|
17
|
Li A, Tyson J, Patel S, Patel M, Katakam S, Mao X, He W. Emerging Nanotechnology for Treatment of Alzheimer's and Parkinson's Disease. Front Bioeng Biotechnol 2021; 9:672594. [PMID: 34113606 PMCID: PMC8185219 DOI: 10.3389/fbioe.2021.672594] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/06/2021] [Indexed: 01/21/2023] Open
Abstract
The prevalence of the two most common neurodegenerative diseases, Parkinson's disease (PD) and Alzheimer's Disease (AD), are expected to rise alongside the progressive aging of society. Both PD and AD are classified as proteinopathies with misfolded proteins α-synuclein, amyloid-β, and tau. Emerging evidence suggests that these misfolded aggregates are prion-like proteins that induce pathological cell-to-cell spreading, which is a major driver in pathogenesis. Additional factors that can further affect pathology spreading include oxidative stress, mitochondrial damage, inflammation, and cell death. Nanomaterials present advantages over traditional chemical or biological therapeutic approaches at targeting these specific mechanisms. They can have intrinsic properties that lead to a decrease in oxidative stress or an ability to bind and disaggregate fibrils. Additionally, nanomaterials enhance transportation across the blood-brain barrier, are easily functionalized, increase drug half-lives, protect cargo from immune detection, and provide a physical structure that can support cell growth. This review highlights emergent nanomaterials with these advantages that target oxidative stress, the fibrillization process, inflammation, and aid in regenerative medicine for both PD and AD.
Collapse
Affiliation(s)
- Amanda Li
- Washington University School of Medicine, St. Louis, MO, United States
| | - Joel Tyson
- Department of Chemical, Biochemical and Environmental Engineering, University of Maryland Baltimore County, Baltimore, MD, United States
| | - Shivni Patel
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Meer Patel
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sruthi Katakam
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xiaobo Mao
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Weiwei He
- Key Laboratory of Micro-Nano Materials for Energy Storage and Conversion of Henan Province, Henan Joint International Research Laboratory of Nanomaterials for Energy and Catalysis, College of Chemical and Materials Engineering, Institute of Surface Micro and Nano Materials, Xuchang University, Xuchang, China
| |
Collapse
|
18
|
Pichla M, Bartosz G, Stefaniuk I, Sadowska-Bartosz I. pH-Responsive Redox Nanoparticles Protect SH-SY5Y Cells at Lowered pH in a Cellular Model of Parkinson's Disease. Molecules 2021; 26:543. [PMID: 33494255 PMCID: PMC7864521 DOI: 10.3390/molecules26030543] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/17/2021] [Accepted: 01/20/2021] [Indexed: 11/25/2022] Open
Abstract
The damage to SH-SY5Y cells by 6-hydroxydopamine (6-OHDA) is an established cellular model of Parkinson's disease (PD). Redox nanoparticles are a promising tool for therapy, including neurodegenerative diseases. As pH of the brain tissue at sites affected by PD is lowered down to 6.5, we studied the effect of pH-responsive redox nanoparticles (poly(ethylene glycol)-b-poly[4-(2,2,6,6-tetramethylpiperidine-1-oxyl)aminomethylstyrene]), which change their structure in a pH-dependent manner and become active below pH 7 (NRNPs pH), on the viability of SH-SY5Y cells treated with 6-OHDA at pH 6.5 and 7.4. Pretreatment of the cells with NRNPs pH (15-75 μM) prior to the 6-OHDA treatment increased their survival in a concentration-dependent manner at pH 6.5, but not at pH 7.4. Among several parameters studied (ATP and GSH content, the level of reactive oxygen species, mitochondrial potential, mitochondrial mass), only the mitochondrial mass was dose-dependently protected by NRNPs pH at pH 6.5, but not at pH 7.4. These results indicate that the action of NRNPs pH on mitochondria underlies their protective effect in this cellular model of PD. These results may have potential importance for future applications of NRNPs pH in preclinical and perhaps clinical studies.
Collapse
Affiliation(s)
- Monika Pichla
- Laboratory of Analytical Biochemistry, Institute of Food Technology and Nutrition, College of Natural Sciences, Rzeszow University, 4 Zelwerowicza Street, 35-601 Rzeszow, Poland;
| | - Grzegorz Bartosz
- Department of Bioenergetics, Food Analysis and Microbiology, Institute of Food Technology and Nutrition, College of Natural Sciences, Rzeszow University, 4 Zelwerowicza Street, 35-601 Rzeszow, Poland;
| | - Ireneusz Stefaniuk
- Teaching and Research Center of Microelectronics and Nanotechnology, College of Natural Sciences, University of Rzeszow, 35-959 Rzeszow, Poland;
| | - Izabela Sadowska-Bartosz
- Laboratory of Analytical Biochemistry, Institute of Food Technology and Nutrition, College of Natural Sciences, Rzeszow University, 4 Zelwerowicza Street, 35-601 Rzeszow, Poland;
| |
Collapse
|