1
|
Takeuchi LE, Kalia LV, Simmons CA. Vascular models of Alzheimer's disease: An overview of recent in vitro models of the blood-brain barrier. Neurobiol Dis 2025; 208:106864. [PMID: 40089165 DOI: 10.1016/j.nbd.2025.106864] [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: 11/03/2024] [Revised: 02/18/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Alzheimer's disease (AD) remains an overwhelming epidemiologic and economic burden on our healthcare systems, affecting an estimate of 11 % of individuals aged 65 years and older. Increasing evidence of the role of the blood-brain barrier (BBB) in AD pathology lends support to the vascular hypothesis of AD, which posits that damage to cerebral vasculature and impairments to cerebral blood flow are major contributors to neurodegeneration in AD. While the question remains whether the dysfunction of the BBB is the cause or consequence of the disease, understanding of the relationship between vascular pathology and AD is growing increasingly complex, warranting the need for better tools to study vasculature in AD. This review provides an overview of AD models in the context of studying vascular impairments and their relevance in pathology. Specifically, we summarize opportunities in in vitro models, cell sources, and phenotypic observations in sporadic and familial forms of AD. Further, we describe recent advances in generating models which recapitulate in vivo characteristics of the BBB in AD through the use of microfluidics, induced pluripotent stem cells (iPSC), and organoid technologies. Finally, we provide a searchable database of reported cell-based models of pathogenic AD gene variants.
Collapse
Affiliation(s)
- Lily E Takeuchi
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5G 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada.
| | - Lorraine V Kalia
- Division of Neurology, Department of Medicine, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON M5T 2S8, Canada; Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada; Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Craig A Simmons
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5G 3G9, Canada; Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada; Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada.
| |
Collapse
|
2
|
Hau Gan C, Chen HJ, Yu-Syun Fan R, Pan SC, Voon Kong K. A Multifunctional Ru-Re Complex for Photodynamic Therapy and CO-Mediated Therapeutics in Alzheimer's Disease. Chemistry 2025; 31:e202403666. [PMID: 39714883 DOI: 10.1002/chem.202403666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/13/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
The development of multifunctional therapeutic agents is crucial for addressing complex diseases such as Alzheimer's disease. Herein, we report a ruthenium-rhenium (Ru-Re) complex that combines photodynamic therapy (PDT) and carbon monoxide (CO) generation capabilities. The Ru-Re complex shows promising photophysical property and significant therapeutic potential. Our studies reveal the complex's ability to generate singlet oxygen (1O2), CO, inhibit tau aggregation, and enhance mitochondrial respiration. These multifunctional properties position the Ru-Re complex as a versatile tool for therapeutic applications in Alzheimer's disease.
Collapse
Affiliation(s)
- Chun Hau Gan
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | - Hsin-Jou Chen
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | - Ricky Yu-Syun Fan
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
| | - Siao-Cian Pan
- Immune Research Core, Department of Medical Research, National Taiwan University Hospital, No.7, Chung Shan S. Rd., Zhongzheng Dist., Taipei City, 100225, Taiwan
| | - Kien Voon Kong
- Department of Chemistry, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
- Center for Emerging Material and Advanced Devices, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd., Taipei, 10617, Taiwan
| |
Collapse
|
3
|
Shankar G, Kumar P, Rai S, Ghosh A, Varma T, Wani MA, Kumar S, Mandloi U, Singh GK, Garg P, Kulkarni O, Srikrishna S, Kumar S, Modi G. Discovery of novel hybrid tryptamine-rivastigmine molecules as potent AChE and BChE inhibitors exhibiting multifunctional properties for the management of Alzheimer's disease. Eur J Med Chem 2025; 283:117066. [PMID: 39667052 DOI: 10.1016/j.ejmech.2024.117066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 11/06/2024] [Accepted: 11/14/2024] [Indexed: 12/14/2024]
Abstract
Contemporary research evidence has corroborated a gradual loss of central cholinergic neurons in Alzheimer's Disease (AD). This progressive deterioration leads to cognitive dysfunction and impaired motor activity, culminating in the brain cell's death in the disease. The approved drugs for AD treatment can only offer relief from symptoms without addressing the underlying pathological hallmarks of the disease. To address the limitations associated with rivastigmine (RIV), a marketed drug for AD, a series of tryptamine derivatives was designed, synthesized, and evaluated in various in-vitro and in-vivo AD models. Enzyme inhibition studies identified compounds 6d and 6e as the lead molecules with potent inhibitors against AChE (6d, IC50: 0.99 ± 0.009 nM and 6e IC50: 7.97 ± 0.016 nM and BChE (6d, IC50: 27.79 ± 0.21 nM and 6e, IC50: 0.79 ± 0.005 nM), compared to the marketed drug Riv (AChE, IC50: 6630 ± 0.76 nM, BChE IC50 = 91 ± 0.40 nM). The molecular docking and dynamics studies corroborated the enzyme inhibition studies. The PAMPA assay strongly suggested the BBB crossing ability of the lead molecules. Further, 6d and 6e demonstrated the capability to counteract oxidative stress and Aβ1-42 in various in-vitro studies. Compound 6e exhibited remarkable radical scavenging activity in the DPPH assay (IC50: 22.91 ± 1.73 μM) compared to rivastigmine (% radical scavenging activity: 3.71 ± 0.09 at 200 μM). Interestingly, 6d and 6e exhibited promising activity in the AD Drosophila model by protecting eye phenotypes from degeneration induced by Aβ1-42 toxicity and reduced mitochondrial and cellular oxidative stress in this model. Furthermore, upon oral administration, 6d and 6e could reverse scopolamine-induced amnesia by improving spatial and cognitive memory in mice at 0.3 and 0.5 mg/kg compared to rivastigmine at 3 mg/kg and were found to have potent ex-vivo anti-ChEs properties, which are correlated with the observed pro-cognitive effects in the Morris Water Maze, likely mediated through the inhibition of both cholinesterases. The expression of various neuroprotection markers, such as BDNF and TRKB, was significantly overexpressed compared to the disease control group.
Collapse
Affiliation(s)
- Gauri Shankar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P, 221005, India
| | - Prabhat Kumar
- Cell and Neurobiology Laboratory, Department of Biochemistry, Institute of Science, BHU, Varanasi, 221005, India
| | - Sanskriti Rai
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Aparajita Ghosh
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Jawaharnagar Shamirpet Mandal, 500078, Hyderabad, India
| | - Tanmaykumar Varma
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, 160062, India
| | - Mushtaq Ahmad Wani
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, 160062, India
| | - Sunil Kumar
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P, 221005, India
| | - Upesh Mandloi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P, 221005, India
| | - Gireesh Kumar Singh
- Department of Pharmacy, School of Health Science, Central University of South Bihar, Gaya, 824236, India
| | - Prabha Garg
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, 160062, India
| | - Onkar Kulkarni
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Jawaharnagar Shamirpet Mandal, 500078, Hyderabad, India
| | - Saripella Srikrishna
- Cell and Neurobiology Laboratory, Department of Biochemistry, Institute of Science, BHU, Varanasi, 221005, India
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India; Department of Health, Education and Technology, Lulea University of Technology, Lulea, Sweden
| | - Gyan Modi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, U.P, 221005, India.
| |
Collapse
|
4
|
Florio D, Annunziata A, Panzetta V, Netti PA, Ruffo F, Marasco D. η 6-Arene Ru(II) Complexes as Modulators of Amyloid Aggregation. Inorg Chem 2024; 63:16001-16010. [PMID: 39129368 DOI: 10.1021/acs.inorgchem.4c02456] [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: 08/13/2024]
Abstract
Inorganic medicinal compounds represent a unique and versatile source of potential therapeutics in many diseases and, more recently, in neurodegeneration. Herein we investigated the effects of two η6-arene Ru(II) complexes on the self-aggregation processes of several amyloidogenic peptides endowed with different kinetics and primary sequences. The Ru(II) complexes exhibit, around the metal ion, two chlorides, one NHC = N-heterocyclic carbene, with a glucosyl and a methyl substituent and separately a hexamethylbenzene, which is named Ru1, and one benzene, named Ru2. Both complexes were demonstrated to bind monomeric amyloids suppressing aggregation as evidenced in thioflavin T (ThT) binding assays and autofluorescence experiments. Electrospray ionization mass spectrometry (ESI-MS) indicated the formation of direct adducts between amyloid and metal complexes, which determined the marked conformational variation of peptides and a rescue of cellular viability in SH-SY5Y cells. The complex Ru2 was demonstrated to be a more potent inhibitor of amyloid aggregation compared to Ru1 likely because of the less hindrance of the arene moiety. The presented data strongly support the in vitro ability of η6-arene Ru(II) complexes to suppress amyloid aggregation, providing insights into their potential application as novel therapeutics in neurodegenerative diseases.
Collapse
Affiliation(s)
- Daniele Florio
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Alfonso Annunziata
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
- Department of Chemical, Materials, and Industrial Production Engineering (DICMaPI), University of Naples Federico II, 80125 Naples, Italy
| | - Valeria Panzetta
- Department of Chemical, Materials, and Industrial Production Engineering (DICMaPI), University of Naples Federico II, 80125 Naples, Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| | - Paolo A Netti
- Department of Chemical, Materials, and Industrial Production Engineering (DICMaPI), University of Naples Federico II, 80125 Naples, Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| | - Francesco Ruffo
- Department of Chemical Sciences, University of Naples Federico II, 80126 Naples, Italy
| | - Daniela Marasco
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
5
|
Micocci S, Stefania R, Garello F, Fasoglio U, Hawala I, Tei L, Geninatti Crich S, Digilio G. Synthesis of fluorinated curcumin derivatives for detecting amyloid plaques by 19F-MRI. Org Biomol Chem 2024; 22:5948-5959. [PMID: 38979663 DOI: 10.1039/d4ob00730a] [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: 07/10/2024]
Abstract
The most prominent pathophysiological hallmark of Alzheimer's disease is the aggregation of amyloid-β (Aβ) peptides into senile plaques. Curcumin and its derivatives exhibit a high affinity for binding to Aβ fibrils, effectively inhibiting their growth. This property holds promise for both therapeutic applications and diagnostic molecular imaging. In this study, curcumin was functionalized with perfluoro-tert-butyl groups to create candidate molecular probes specifically targeted to Aβ fibrils for use in 19F-magnetic resonance imaging. Two types of fluorinated derivatives were considered: mono-substituted (containing nine fluorine atoms per molecule) and disubstituted (containing eighteen fluorine atoms). The linker connecting the perfluoro moiety with the curcumin scaffold was evaluated for its impact on binding affinity and water solubility. All mono-substituted compounds and one disubstituted compound exhibited a binding affinity toward Aβ fibrils on the same order of magnitude as reference curcumin. The insertion of a charged carboxylate group into the linker enhanced the water solubility of the probes. Compound Curc-Glu-F9 (with one L-glutamyl moiety and a perfluoro-tert-butyl group), showed the best properties in terms of binding affinity towards Aβ fibrils, water solubility, and intensity of the 19F-NMR signal in the Aβ oligomer bound form.
Collapse
Affiliation(s)
- Sebastiano Micocci
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126, Torino, Italy
| | - Rachele Stefania
- Department of Science and Technological Innovation, University of Eastern Piedmont "Amedeo Avogadro", Viale Teresa Michel 11, 15120, Alessandria, Italy.
| | - Francesca Garello
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126, Torino, Italy
| | - Umberto Fasoglio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126, Torino, Italy
| | - Ivan Hawala
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126, Torino, Italy
| | - Lorenzo Tei
- Department of Science and Technological Innovation, University of Eastern Piedmont "Amedeo Avogadro", Viale Teresa Michel 11, 15120, Alessandria, Italy.
| | - Simonetta Geninatti Crich
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Via Nizza 52, 10126, Torino, Italy
| | - Giuseppe Digilio
- Department of Science and Technological Innovation, University of Eastern Piedmont "Amedeo Avogadro", Viale Teresa Michel 11, 15120, Alessandria, Italy.
| |
Collapse
|
6
|
Polykretis P, D’Andrea C, Banchelli M, Napolitano L, Cascella R, de Angelis M, Matteini P. Exploring the Aβ 1-42 fibrillogenesis timeline by atomic force microscopy and surface enhanced Raman spectroscopy. Front Mol Biosci 2024; 11:1376411. [PMID: 38948077 PMCID: PMC11211275 DOI: 10.3389/fmolb.2024.1376411] [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: 01/25/2024] [Accepted: 05/22/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction: Alzheimer's disease (AD) is a progressive debilitating neurological disorder representing the most common neurodegenerative disease worldwide. Although the exact pathogenic mechanisms of AD remain unresolved, the presence of extracellular amyloid-β peptide 1-42 (Aβ1-42) plaques in the parenchymal and cortical brain is considered one of the hallmarks of the disease. Methods: In this work, we investigated the Aβ1-42 fibrillogenesis timeline up to 48 h of incubation, providing morphological and chemo-structural characterization of the main assemblies formed during the aggregation process of Aβ1-42, by atomic force microscopy (AFM) and surface enhanced Raman spectroscopy (SERS), respectively. Results: AFM topography evidenced the presence of characteristic protofibrils at early-stages of aggregation, which form peculiar macromolecular networks over time. SERS allowed to track the progressive variation in the secondary structure of the aggregation species involved in the fibrillogenesis and to determine when the β-sheet starts to prevail over the random coil conformation in the aggregation process. Discussion: Our research highlights the significance of investigating the early phases of fibrillogenesis to better understand the molecular pathophysiology of AD and identify potential therapeutic targets that may prevent or slow down the aggregation process.
Collapse
Affiliation(s)
- Panagis Polykretis
- Institute of Applied Physics “Nello Carrara”, National Research Council, Sesto Fiorentino, Italy
| | - Cristiano D’Andrea
- Institute of Applied Physics “Nello Carrara”, National Research Council, Sesto Fiorentino, Italy
| | - Martina Banchelli
- Institute of Applied Physics “Nello Carrara”, National Research Council, Sesto Fiorentino, Italy
| | - Liliana Napolitano
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Roberta Cascella
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Marella de Angelis
- Institute of Applied Physics “Nello Carrara”, National Research Council, Sesto Fiorentino, Italy
| | - Paolo Matteini
- Institute of Applied Physics “Nello Carrara”, National Research Council, Sesto Fiorentino, Italy
| |
Collapse
|
7
|
Bhole RP, Chikhale RV, Rathi KM. Current biomarkers and treatment strategies in Alzheimer disease: An overview and future perspectives. IBRO Neurosci Rep 2024; 16:8-42. [PMID: 38169888 PMCID: PMC10758887 DOI: 10.1016/j.ibneur.2023.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 01/05/2024] Open
Abstract
Alzheimer's disease (AD), a progressive degenerative disorder first identified by Alois Alzheimer in 1907, poses a significant public health challenge. Despite its prevalence and impact, there is currently no definitive ante mortem diagnosis for AD pathogenesis. By 2050, the United States may face a staggering 13.8 million AD patients. This review provides a concise summary of current AD biomarkers, available treatments, and potential future therapeutic approaches. The review begins by outlining existing drug targets and mechanisms in AD, along with a discussion of current treatment options. We explore various approaches targeting Amyloid β (Aβ), Tau Protein aggregation, Tau Kinases, Glycogen Synthase kinase-3β, CDK-5 inhibitors, Heat Shock Proteins (HSP), oxidative stress, inflammation, metals, Apolipoprotein E (ApoE) modulators, and Notch signaling. Additionally, we examine the historical use of Estradiol (E2) as an AD therapy, as well as the outcomes of Randomized Controlled Trials (RCTs) that evaluated antioxidants (e.g., vitamin E) and omega-3 polyunsaturated fatty acids as alternative treatment options. Notably, positive effects of docosahexaenoic acid nutriment in older adults with cognitive impairment or AD are highlighted. Furthermore, this review offers insights into ongoing clinical trials and potential therapies, shedding light on the dynamic research landscape in AD treatment.
Collapse
Affiliation(s)
- Ritesh P. Bhole
- Department of Pharmaceutical Chemistry, Dr. D. Y. Patil institute of Pharmaceutical Sciences & Research, Pimpri, Pune, India
- Dr. D. Y. Patil Dental College and Hospital, Dr. D. Y. Patil Vidyapeeth, Pimpri, Pune 411018, India
| | | | - Karishma M. Rathi
- Department of Pharmacy Practice, Dr. D. Y. Patil institute of Pharmaceutical Sciences & Research, Pimpri, Pune, India
| |
Collapse
|
8
|
Acharya NK, Grossman HC, Clifford PM, Levin EC, Light KR, Choi H, Swanson II RL, Kosciuk MC, Venkataraman V, Libon DJ, Matzel LD, Nagele RG. A Chronic Increase in Blood-Brain Barrier Permeability Facilitates Intraneuronal Deposition of Exogenous Bloodborne Amyloid-Beta1-42 Peptide in the Brain and Leads to Alzheimer's Disease-Relevant Cognitive Changes in a Mouse Model. J Alzheimers Dis 2024; 98:163-186. [PMID: 38393907 PMCID: PMC10977376 DOI: 10.3233/jad-231028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2023] [Indexed: 02/25/2024]
Abstract
Background Increased blood-brain barrier (BBB) permeability and amyloid-β (Aβ) peptides (especially Aβ1-42) (Aβ42) have been linked to Alzheimer's disease (AD) pathogenesis, but the nature of their involvement in AD-related neuropathological changes leading to cognitive changes remains poorly understood. Objective To test the hypothesis that chronic extravasation of bloodborne Aβ42 peptide and brain-reactive autoantibodies and their entry into the brain parenchyma via a permeable BBB contribute to AD-related pathological changes and cognitive changes in a mouse model. Methods The BBB was rendered chronically permeable through repeated injections of Pertussis toxin (PT), and soluble monomeric, fluorescein isothiocyanate (FITC)-labeled or unlabeled Aβ42 was injected into the tail-vein of 10-month-old male CD1 mice at designated intervals spanning ∼3 months. Acquisition of learned behaviors and long-term retention were assessed via a battery of cognitive and behavioral tests and linked to neuropathological changes. Results Mice injected with both PT and Aβ42 demonstrated a preferential deficit in the capacity for long-term retention and an increased susceptibility to interference in selective attention compared to mice exposed to PT or saline only. Immunohistochemical analyses revealed increased BBB permeability and entry of bloodborne Aβ42 and immunoglobulin G (IgG) into the brain parenchyma, selective neuronal binding of IgG and neuronal accumulation of Aβ42 in animals injected with both PT and Aβ42 compared to controls. Conclusion Results highlight the potential synergistic role of BBB compromise and the influx of bloodborne Aβ42 into the brain in both the initiation and progression of neuropathologic and cognitive changes associated with AD.
Collapse
Affiliation(s)
- Nimish K. Acharya
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Biomarker Discovery Center, New Jersey Institute for Successful Aging (NJISA), Rowan-Virtua School of Osteopathic Medicine, Stratford, NJ, USA
- Rowan-Virtua Graduate School of Biomedical Sciences, Stratford, NJ, USA
- Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Rowan University, Glassboro, NJ, USA
| | | | - Peter M. Clifford
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- HNL Lab Medicine, Allentown, PA, USA
| | - Eli C. Levin
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Department of Graduate Medical Education, Bayhealth Medical Center, Dover, DE, USA
| | - Kenneth R. Light
- Department of Psychology, Barnard College of Columbia University, New York, NY, USA
| | - Hana Choi
- Rowan-Virtua Graduate School of Biomedical Sciences, Stratford, NJ, USA
| | - Randel L. Swanson II
- Center for Neurotrauma, Neurodegeneration and Restoration, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Rehab Medicine Service, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Physical Medicine and Rehabilitation, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mary C. Kosciuk
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
| | - Venkat Venkataraman
- Department of Cell Biology and Neuroscience, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Department of Academic and Student Affairs, Rowan-Virtua School of Osteopathic Medicine, Stratford, NJ, USA
| | - David J. Libon
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Department of Psychology, Rowan University, Glassboro, NJ, USA
| | - Louis D. Matzel
- Department of Psychology, Rutgers University, Piscataway, NJ, USA
| | - Robert G. Nagele
- Department of Geriatrics and Gerontology, New Jersey Institute for Successful Aging, Rowan-Virtua School of Osteopathic Medicine, Rowan University, Stratford, NJ, USA
- Biomarker Discovery Center, New Jersey Institute for Successful Aging (NJISA), Rowan-Virtua School of Osteopathic Medicine, Stratford, NJ, USA
- Rowan-Virtua Graduate School of Biomedical Sciences, Stratford, NJ, USA
- Rowan-Virtua School of Translational Biomedical Engineering and Sciences, Rowan University, Glassboro, NJ, USA
| |
Collapse
|
9
|
Zhaliazka K, Matveyenka M, Kurouski D. Lipids uniquely alter the secondary structure and toxicity of amyloid beta 1-42 aggregates. FEBS J 2023; 290:3203-3220. [PMID: 36705524 PMCID: PMC10389563 DOI: 10.1111/febs.16738] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/25/2022] [Accepted: 01/25/2023] [Indexed: 01/28/2023]
Abstract
Abrupt aggregation of amyloid β1-42 (Aβ) peptide is a hallmark of Alzheimer's disease (AD), a severe pathology that affects more than 44 million people worldwide. A growing body of evidence suggests that lipids can uniquely alter rates of Aβ1-42 aggregation. However, it remains unclear whether lipids only alter rates of protein aggregation or also uniquely modify the secondary structure and toxicity of Aβ1-42 oligomers and fibrils. In this study, we investigated the effect of phosphatidylcholine (PC), cardiolipin (CL), and cholesterol (Chol) on Aβ1-42 aggregation. We found that PC, CL and Chol strongly accelerated the rate of fibril formation compared to the rate of Aβ1-42 aggregation in the lipid-free environment. Furthermore, anionic CL enabled the strongest acceleration of Aβ1-42 aggregation compared to zwitterionic PC and uncharged Chol. We also found that PC, CL and Chol uniquely altered the secondary structure of early-, middle- and late-stage Aβ1-42 aggregates. Specifically, CL and Chol drastically increased the amount of parallel β-sheet in Aβ1-42 oligomers and fibrils grown in the presence of these lipids. This caused a significant increase in the toxicity of Aβ : CL and Aβ : Chol compared to the toxicity of Aβ : PC and Aβ1-42 aggregates formed in the lipid-free environment. These results demonstrate that toxicity of Aβ aggregates correlates with the amount of their β-sheet content, which, in turn, is determined by the chemical structure of lipids present at the stage of Aβ1-42 aggregation.
Collapse
Affiliation(s)
- Kiryl Zhaliazka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Mikhail Matveyenka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas 77843, United States
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, 77843, United States
| |
Collapse
|
10
|
Singh S, Navale GR, Agrawal S, Singh HK, Singla L, Sarkar D, Sarma M, Choudhury AR, Ghosh K. Design and synthesis of ruthenium complexes and their studies on the inhibition of amyloid β (1-42) peptide aggregation. Int J Biol Macromol 2023; 239:124197. [PMID: 36972817 DOI: 10.1016/j.ijbiomac.2023.124197] [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: 11/10/2022] [Revised: 03/07/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023]
Abstract
Misfolding and protein aggregation have been linked to numerous human neurodegenerative disorders such as Alzheimer's, prions, and Parkinson's. Due to their interesting photophysical properties, ruthenium (Ru) complexes have received considerable attention in studying protein aggregation. In this study, we synthesized the novel Ru complexes ([Ru(p-cymene)Cl(L-1)][PF6](Ru-1), and [Ru(p-cymene)Cl(L-2)][PF6](Ru-2)) and investigated their inhibitory activity against the bovine serum albumin (BSA) aggregation and the Aβ1-42 peptides amyloid formation. Several spectroscopic methods were used to characterize the complexes, and the molecular structure was determined by X-ray crystallography. Amyloid aggregation and inhibition activity were examined using the Thioflavin-T (ThT) assay, and secondary structures were analyzed by circular dichroism (CD) spectroscopy and transmission electron microscopy (TEM). The cell viability assay was carried out on the neuroblastoma cell line, revealing that the Ru-2 complex showed better protective effects against Aβ1-42 peptide toxicity on neuro-2a cells than the Ru-1 complex. Molecular docking studies elucidate binding sites and interactions between the Ru-complexes and the Aβ1-42 fibrils. The experimental studies revealed that these complexes significantly inhibited BSA aggregation and Aβ1-42 amyloid fibril formation at 1:3 and 1:1 equimolar concentrations, respectively. Antioxidant assays demonstrated that these complexes act as antioxidants, protecting from amyloid-induced oxidative stress. Molecular docking studies with the monomeric Aβ1-42 (PDB: 1IYT) show hydrophobic interaction, and both complexes bind preferably in the central region of the peptide and coordinate with two binding sites of the peptide. Hence, we suggest that the Ru-based complexes could be applied as a potential agent in metallopharmaceutical research against Alzheimer's disease.
Collapse
Affiliation(s)
- Sain Singh
- Department of Chemistry, Indian Institute of Technology, Roorkee 247667, India
| | - Govinda R Navale
- Department of Chemistry, Indian Institute of Technology, Roorkee 247667, India
| | - Sonia Agrawal
- Department of Organic Chemistry, CSIR-National Chemical Laboratory, Pune 411 008, India
| | - Haobam Kisan Singh
- Department of Chemistry, Indian Institute of Technology, Guwahati 781039, India
| | - Labhini Singla
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Dhiman Sarkar
- Department of Organic Chemistry, CSIR-National Chemical Laboratory, Pune 411 008, India
| | - Manabendra Sarma
- Department of Chemistry, Indian Institute of Technology, Guwahati 781039, India
| | - Anghuman Roy Choudhury
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Mohali, India
| | - Kaushik Ghosh
- Department of Chemistry, Indian Institute of Technology, Roorkee 247667, India; Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee 247667, India.
| |
Collapse
|
11
|
Matveyenka M, Rizevsky S, Pellois JP, Kurouski D. Lipids uniquely alter rates of insulin aggregation and lower toxicity of amyloid aggregates. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159247. [PMID: 36272517 PMCID: PMC10401553 DOI: 10.1016/j.bbalip.2022.159247] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/14/2022] [Accepted: 10/02/2022] [Indexed: 02/25/2023]
Abstract
Amyloid formation is a hallmark of many medical diseases including diabetes type 2, Alzheimer's and Parkinson diseases. Under these pathological conditions, misfolded proteins self-assemble forming oligomers and fibrils, structurally heterogeneous aggregates that exhibit a large variety of shapes and forms. A growing body of evidence points to drastic changes in the lipid profile in organs affected by amyloidogenic diseases. In this study, we investigated the extent to which individual phospho- and sphingolipids, as well as their mixtures can impact insulin aggregation. Our results show that lipids and their mixtures uniquely alter rates of insulin aggregation simultaneously changing the secondary structure of protein aggregates that are grown in their presence. These structurally different protein-lipid aggregates impact cell viability to different extent while using distinct mechanisms of toxicity. These findings suggest that irreversible changes in lipid profiles of organs may trigger formation of toxic protein species that in turn are responsible for the onset and progression of amyloidogenic diseases.
Collapse
Affiliation(s)
- Mikhail Matveyenka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Stanislav Rizevsky
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States; Department of Biotechnology, Binh Duong University, Thu Dau Mot 820000, Viet Nam
| | - Jean-Philippe Pellois
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, United States; Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, United States.
| |
Collapse
|
12
|
Matveyenka M, Zhaliazka K, Rizevsky S, Kurouski D. Lipids uniquely alter secondary structure and toxicity of lysozyme aggregates. FASEB J 2022; 36:e22543. [PMID: 36094052 PMCID: PMC10427241 DOI: 10.1096/fj.202200841r] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/03/2022] [Accepted: 08/29/2022] [Indexed: 08/17/2023]
Abstract
Abrupt aggregation of misfolded proteins is a hallmark of the large group of amyloid pathologies that include diabetes type 2, Alzheimer and Parkinson's diseases. Protein aggregation yields oligomers and fibrils, β-sheet-rich structures that exert cell toxicity. Microscopic examination of amyloid deposits reveals the presence of lipids membranes, which suggests that lipids can be involved in the process of pathogenic protein assembly. In this study, we show that lipids can uniquely alter the aggregation rates of lysozyme, a protein that is associated with systemic amyloidosis. Specifically, cardiolipin (CL), ceramide (CER), and sphingomyelin (SM) accelerate, phosphatidylcholine (PC) strongly inhibits, whereas phosphatidylserine (PS) has no effect on the rate of protein aggregation. Furthermore, lipids uniquely alter the secondary structure of lysozyme aggregates. Furthermore, we found that lysozyme aggregates grown in the presence of CL, CER, SM, PS, and CL:PC mixtures exert significantly lower production of reactive oxygen species and mitochondrial dysfunction compared to lysozyme:PC aggregates and lysozyme fibrils grown in the lipid-free environment. These findings suggest that a change in the lipid composition of cell membranes, which is taken place upon neurodegeneration, may trigger the formation of toxic protein species that otherwise would not be formed.
Collapse
Affiliation(s)
- Mikhail Matveyenka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Kiryl Zhaliazka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Stanislav Rizevsky
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
- Department of Biotechnology, Binh Duong University, Thu Dau Mot, Vietnam
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
13
|
Near-Infrared Photothermally Enhanced Photo-Oxygenation for Inhibition of Amyloid-β Aggregation Based on RVG-Conjugated Porphyrinic Metal-Organic Framework and Indocyanine Green Nanoplatform. Int J Mol Sci 2022; 23:ijms231810885. [PMID: 36142796 PMCID: PMC9505608 DOI: 10.3390/ijms231810885] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Amyloid aggregation is associated with many neurodegenerative diseases such as Alzheimer's disease (AD). The current technologies using phototherapy for amyloid inhibition are usually photodynamic approaches based on evidence that reactive oxygen species can inhibit Aβ aggregation. Herein, we report a novel combinational photothermally assisted photo-oxygenation treatment based on a nano-platform of the brain-targeting peptide RVG conjugated with the 2D porphyrinic PCN-222 metal-organic framework and indocyanine green (PCN-222@ICG@RVG) with enhanced photo-inhibition in Alzheimer's Aβ aggregation. A photothermally assisted photo-oxygenation treatment based on PCN@ICG could largely enhance the photo-inhibition effect on Aβ42 aggregation and lead to much lower neurotoxicity upon near-infrared (NIR) irradiation at 808 nm compared with a single modality of photo-treatment in both cell-free and in vitro experiments. Generally, local photothermal heat increases the instability of Aβ aggregates and keeps Aβ in the status of monomers, which facilitates the photo-oxygenation process of generating oxidized Aβ monomers with low aggregation capability. In addition, combined with the brain-targeting peptide RVG, the PCN-222@ICG@RVG nanoprobe shows high permeability of the human blood-brain barrier (BBB) on a human brain-on-a-chip platform. The ex vivo study also demonstrates that NIR-activated PCN-222@ICG@RVG could efficiently dissemble Aβ plaques. Our work suggests that the combination of photothermal treatment with photo-oxygenation can synergistically enhance the inhibition of Aβ aggregation, which may boost NIR-based combinational phototherapy of AD in the future.
Collapse
|
14
|
Enhancing the Amyloid-β Anti-Aggregation Properties of Curcumin via Arene-Ruthenium(II) Derivatization. Int J Mol Sci 2022; 23:ijms23158710. [PMID: 35955845 PMCID: PMC9369426 DOI: 10.3390/ijms23158710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 11/23/2022] Open
Abstract
Alzheimer’s disease (AD) is a fatal neurodegenerative disorder associated with severe dementia, progressive cognitive decline, and irreversible memory loss. Although its etiopathogenesis is still unclear, the aggregation of amyloid-β (Aβ) peptides into supramolecular structures and their accumulation in the central nervous system play a critical role in the onset and progression of the disease. On such a premise, the inhibition of the early stages of Aβ aggregation is a potential prevention strategy for the treatment of AD. Since several natural occurring compounds, as well as metal-based molecules, showed promising inhibitory activities toward Aβ aggregation, we herein characterized the interaction of an organoruthenium derivative of curcumin with Aβ(1–40) and Aβ(1–42) peptides, and we evaluated its ability to inhibit the oligomerization/fibrillogenesis processes by combining in silico and in vitro methods. In general, besides being less toxic to neuronal cells, the derivative preserved the amyloid binding ability of the parent compound in terms of equilibrium dissociation constants but (most notably) was more effective both in retarding the formation and limiting the size of amyloid aggregates by virtue of a higher hindering effect on the amyloid–amyloid elongation surface. Additionally, the complex protected neuronal cells from amyloid toxicity.
Collapse
|
15
|
Davani L, Fu X, De Simone A, Li P, Montanari S, Lämmerhofer M, Andrisano V. Aß1-42 peptide toxicity on neuronal cells: a lipidomic study. J Pharm Biomed Anal 2022; 219:114876. [DOI: 10.1016/j.jpba.2022.114876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/01/2022] [Accepted: 06/02/2022] [Indexed: 10/18/2022]
|
16
|
Matveyenka M, Rizevsky S, Kurouski D. Unsaturation in the Fatty Acids of Phospholipids Drastically Alters the Structure and Toxicity of Insulin Aggregates Grown in Their Presence. J Phys Chem Lett 2022; 13:4563-4569. [PMID: 35580189 PMCID: PMC9170185 DOI: 10.1021/acs.jpclett.2c00559] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Lipid bilayers play an important role in the pathological assembly of amyloidogenic proteins and peptides. This assembly yields oligomers and fibrils, which are highly toxic protein aggregates. In this study, we investigated the role of saturation in fatty acids of two phospholipids that are present in cell membranes. We found that unsaturated cardiolipin (CL) drastically shortened the lag phase of insulin aggregation. Furthermore, structurally and morphologically different aggregates were formed in the presence of unsaturated CL vs saturated CL. These aggregates exerted drastically different cell toxicity. Both saturated and unsaturated phosphatidylcholine (PC) were able to inhibit insulin aggregation equally efficiently. Similar to CL, structurally different aggregates were formed in the presence of saturated and unsaturated PC. These aggregates exerted different cell toxicities. These results show that unsaturated phospholipids catalyze the formation of more toxic amyloid aggregates comparing to those formed in the presence of saturated lipids.
Collapse
Affiliation(s)
| | - Stanislav Rizevsky
- Department of Biotechnology, Binh Duong University, Thu Dau Mot 820000, Vietnam
| | | |
Collapse
|
17
|
Mansour AM, Khaled RM, Khaled E, Ahmed SK, Ismael OS, Zeinhom A, Magdy H, Ibrahim SS, Abdelfatah M. Ruthenium(II) carbon monoxide releasing molecules: Structural perspective, antimicrobial and anti-inflammatory properties. Biochem Pharmacol 2022; 199:114991. [DOI: 10.1016/j.bcp.2022.114991] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 01/12/2023]
|
18
|
Peng YB, Tao C, Tan CP, Zhao P. Inhibition of Aβ peptide aggregation by ruthenium(II) polypyridyl complexes through copper chelation. J Inorg Biochem 2021; 224:111591. [PMID: 34450410 DOI: 10.1016/j.jinorgbio.2021.111591] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/18/2021] [Accepted: 08/18/2021] [Indexed: 10/20/2022]
Abstract
Alzheimer's disease (AD) is known as a complex multifactorial syndrome and both metal chelators and amyloid β peptide (Aβ) inhibitors show promise against AD. Herein, four small hybrid compounds have been designed and synthesized utilizing 8-hydroxyquinoline, pyridine or imidazole as chelators and benzimidazole as the recognition moiety for AD treatment. These conjugates can capture Cu2+ from Aβ and become dimers upon Cu2+ coordination and show high efficiency for both Cu2+ elimination and Aβ assembly inhibition. Besides, these designed complexes can inhibit the production of Aβ-induced reactive oxygen species (ROS), protect mitochondria from damage, and improve the survival rate of neuron cells. Our work provides a new strategy to combine hydrophobic interaction and metal ion chelation to design amyloid inhibitors.
Collapse
Affiliation(s)
- Yan-Bo Peng
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Education Mega Centre, No. 280, Waihuandong Road, Guangzhou 510006, PR China
| | - Can Tao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Education Mega Centre, No. 280, Waihuandong Road, Guangzhou 510006, PR China
| | - Cai-Ping Tan
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, School of Chemistry, Sun Yat-Sen University, Guangzhou 510275, PR China.
| | - Ping Zhao
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Education Mega Centre, No. 280, Waihuandong Road, Guangzhou 510006, PR China.
| |
Collapse
|
19
|
Chauhan PS, Yadav D, Koul B, Mohanta YK, Jin JO. Recent Advances in Nanotechnology: A Novel Therapeutic System for the Treatment of Alzheimer's Disease. Curr Drug Metab 2020; 21:1144-1151. [PMID: 33234100 DOI: 10.2174/1389200221666201124140518] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/14/2020] [Accepted: 10/15/2020] [Indexed: 11/22/2022]
Abstract
A amyloid-β (Aβ) plaque formation in the brain is known to be the root cause of Alzheimer's disease (AD), which affects the behavior, memory, and cognitive ability in humans. The brain starts undergoing changes several years before the actual appearance of the symptoms. Nanotechnology could prove to be an alternative strategy for treating the disease effectively. It encompasses the diagnosis as well as the therapeutic aspect using validated biomarkers and nano-based drug delivery systems, respectively. A nano-based therapy may provide an alternate strategy, wherein one targets the protofibrillar amyloid-β (Aβ) structures, and this is followed by their disaggregation as random coils. Conventional/routine drug therapies are inefficient in crossing the blood-brain barrier; however, this hurdle can be overcome with the aid of nanoparticles. The present review highlights the various challenges in the diagnosis and treatment of AD. Meticulous and collaborative research using nanotherapeutic systems could provide remarkable breakthroughs in the early-stage diagnosis and therapy of AD.
Collapse
Affiliation(s)
- Pallavi Singh Chauhan
- Amity Institute of Biotechnology, Amity University Madhya Pradesh, Gwalior, 474005, India
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea
| | - Bhupendra Koul
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara-144411, Punjab, India
| | - Yugal Kishore Mohanta
- Biochemistry Laboratory, Department of Botany, North Orissa University Baripada- 757003, India
| | - Jun-O Jin
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea
| |
Collapse
|
20
|
Modulation of Amyloidogenic Peptide Aggregation by Photoactivatable CO-Releasing Ruthenium(II) Complexes. Pharmaceuticals (Basel) 2020; 13:ph13080171. [PMID: 32751396 PMCID: PMC7464691 DOI: 10.3390/ph13080171] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/23/2020] [Accepted: 07/28/2020] [Indexed: 01/19/2023] Open
Abstract
Three Ru(II)-based CO-releasing molecules featuring bidentate benzimidazole and terpyridine derivatives as ligands were investigated for their ability to modulate the aggregation process of the second helix of the C-terminal domain of nucleophosmin 1, namely nucleophosmin 1 (NPM1)264-277, a model amyloidogenic system, before and after irradiation at 365 nm. Thioflavin T (ThT) binding assays and UV/Vis absorption spectra indicate that binding of the compounds to the peptide inhibits its aggregation and that the inhibitory effect increases upon irradiation (half maximal effective concentration (EC50) values in the high micromolar range). Electrospray ionization mass spectrometry data of the peptide in the presence of one of these compounds confirm that the modulation of amyloid aggregation relies on the formation of adducts obtained when the Ru compounds react with the peptide upon releasing of labile ligands, like chloride and carbon monoxide. This mechanism of action explains the subtle different behavior of the three compounds observed in ThT experiments. Overall, data support the hypothesis that metal-based CO releasing molecules can be used to develop metal-based drugs with potential application as anti-amyloidogenic agents.
Collapse
|
21
|
De Simone A, Naldi M, Tedesco D, Bartolini M, Davani L, Andrisano V. Advanced analytical methodologies in Alzheimer’s disease drug discovery. J Pharm Biomed Anal 2020; 178:112899. [DOI: 10.1016/j.jpba.2019.112899] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 12/13/2022]
|