1
|
Chen S, Li R, Liu Y, Zhang Z, Fang M, Huang S, Li Y, Geng L. Multifunctional Nitrogen-Doped Carbon Dots to Inhibit the Aggregation of Aβ Peptide and Depolymerize the Aβ Fibrils by Modulating Reactive Oxygen Species. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:26018-26025. [PMID: 39602498 DOI: 10.1021/acs.langmuir.4c03454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Multifunctional nitrogen-doped carbon dots (N-CDs) were synthesized, and the morphology, composition, and spectral properties of N-CDs were characterized by multiple characterization techniques. The inhibition of β-amyloid (Aβ) peptide aggregation and the destruction of the Aβ fibril structure by N-CDs were also studied. The conformational transition and morphology of Aβ42 in the presence of N-CDs were monitored by far-UV circular dichroism (CD) spectroscopy and transmission electron microscopy (TEM). The results demonstrated that the prepared N-CDs could effectively inhibit Aβ42 peptide aggregation and depolymerize Aβ fibrils. Furthermore, the inhibition and disaggregation mechanism of existing Aβ42 fibrils by N-CDs was studied by electron paramagnetic resonance spectroscopy (EPR). The results showed that the modulation of reactive oxygen species (ROS) by N-CDs and multiple interactions between N-CDs and Aβ42 fibrils played a crucial part in restraining and reducing the aggregation of Aβ42. Our work demonstrates the therapeutic potential of N-CDs in suppressing Aβ42 peptide aggregation and destroying existing Aβ42 fibrils, which provides a new perspective strategy in the treatment of Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Shenna Chen
- Hebei Technology Innovation Center for Energy Conversion Materials and Devices, College of Chemistry and Material Science, Hebei Normal University, Shijiazhuang 050024, P. R. China
| | - Ronghui Li
- Hebei Technology Innovation Center for Energy Conversion Materials and Devices, College of Chemistry and Material Science, Hebei Normal University, Shijiazhuang 050024, P. R. China
| | - Yanxu Liu
- Hebei Technology Innovation Center for Energy Conversion Materials and Devices, College of Chemistry and Material Science, Hebei Normal University, Shijiazhuang 050024, P. R. China
| | - Ziyan Zhang
- Hebei Technology Innovation Center for Energy Conversion Materials and Devices, College of Chemistry and Material Science, Hebei Normal University, Shijiazhuang 050024, P. R. China
| | - Mei Fang
- Hebei Technology Innovation Center for Energy Conversion Materials and Devices, College of Chemistry and Material Science, Hebei Normal University, Shijiazhuang 050024, P. R. China
| | - Sihang Huang
- Hebei Technology Innovation Center for Energy Conversion Materials and Devices, College of Chemistry and Material Science, Hebei Normal University, Shijiazhuang 050024, P. R. China
| | - Yayong Li
- Department of Rehabilitation Medicine, Shijiazhuang People's Hospital, Shijiazhuang 050000, P. R. China
| | - Lina Geng
- Hebei Technology Innovation Center for Energy Conversion Materials and Devices, College of Chemistry and Material Science, Hebei Normal University, Shijiazhuang 050024, P. R. China
| |
Collapse
|
2
|
Mandal S, Suseela YV, Samanta S, Vileno B, Faller P, Govindaraju T. Fluorescent Peptides Sequester Redox Copper to Mitigate Oxidative Stress, Amyloid Toxicity, and Neuroinflammation. ACS Med Chem Lett 2024; 15:1376-1385. [PMID: 39140073 PMCID: PMC11318102 DOI: 10.1021/acsmedchemlett.4c00283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/15/2024] [Accepted: 07/22/2024] [Indexed: 08/15/2024] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder that significantly contributes to dementia. The lack of effective therapeutic interventions presents a significant challenge to global health. We have developed a set of short peptides (PNGln) conjugated with a dual-functional fluorophoric amino acid (NGln). The lead peptide, P2NGln, displays a high affinity for Cu2+, maintaining the metal ion in a redox-inactive state. This mitigates the cytotoxicity generated by reactive oxygen species (ROS), which are produced by Cu2+ under the reductive conditions of Asc and Aβ16 or Aβ42. Furthermore, P2NGln inhibits both Cu-dependent and -independent fibrillation of Aβ42, along with the subsequent toxicity induced by Aβ42. In addition, P2NGln exhibits inhibitory effects on the production of lipopolysaccharide (LPS)-induced ROS and reactive nitrogen species (RNS) in microglial cells. In vitro and cellular studies indicate that P2NGln could significantly reduce Aβ-Cu2+-induced ROS production, amyloid toxicity, and neuroinflammation, offering an innovative strategy against Alzheimer's disease.
Collapse
Affiliation(s)
- Sabyasachi Mandal
- Bioorganic
Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, Karnataka 560064, India
| | - Yelisetty Venkata Suseela
- Institut
de Chimie (UMR 7177), Université de Strasbourg, CNRS, 4 Rue Blaise Pascal, 67000 Strasbourg, France
| | - Sourav Samanta
- Bioorganic
Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, Karnataka 560064, India
| | - Bertrand Vileno
- Institut
de Chimie (UMR 7177), Université de Strasbourg, CNRS, 4 Rue Blaise Pascal, 67000 Strasbourg, France
| | - Peter Faller
- Institut
de Chimie (UMR 7177), Université de Strasbourg, CNRS, 4 Rue Blaise Pascal, 67000 Strasbourg, France
| | - Thimmaiah Govindaraju
- Bioorganic
Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, Karnataka 560064, India
| |
Collapse
|
3
|
Singh K, Kaur A, Goyal B, Goyal D. Harnessing the Therapeutic Potential of Peptides for Synergistic Treatment of Alzheimer's Disease by Targeting Aβ Aggregation, Metal-Mediated Aβ Aggregation, Cholinesterase, Tau Degradation, and Oxidative Stress. ACS Chem Neurosci 2024; 15:2545-2564. [PMID: 38979773 DOI: 10.1021/acschemneuro.4c00246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive multifaceted neurodegenerative disease and remains a formidable global health challenge. The current medication for AD gives symptomatic relief and, thus, urges us to look for alternative disease-modifying therapies based on a multitarget directed approach. Looking at the remarkable progress made in peptide drug development in the last decade and the benefits associated with peptides, they offer valuable chemotypes [multitarget directed ligands (MTDLs)] as AD therapeutics. This review recapitulates the current developments made in harnessing peptides as MTDLs in combating AD by targeting multiple key pathways involved in the disease's progression. The peptides hold immense potential and represent a convincing avenue in the pursuit of novel AD therapeutics. While hurdles remain, ongoing research offers hope that peptides may eventually provide a multifaceted approach to combat AD.
Collapse
Affiliation(s)
- Kamaljot Singh
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib, 140406 Punjab, India
| | - Anupamjeet Kaur
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib, 140406 Punjab, India
| | - Bhupesh Goyal
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering & Technology, Patiala, 147004 Punjab, India
| | - Deepti Goyal
- Department of Chemistry, DAV College, Sector 10, Chandigarh 160011, India
| |
Collapse
|
4
|
Gharai PK, Khan J, Pradhan K, Mallesh R, Garg S, Arshi MU, Barman S, Ghosh S. Power of Dopamine: Multifunctional Compound Assisted Conversion of the Most Risk Factor into Therapeutics of Alzheimer's Disease. ACS Chem Neurosci 2024; 15:2470-2483. [PMID: 38874606 DOI: 10.1021/acschemneuro.3c00777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024] Open
Abstract
In Alzheimer's disease (AD), reactive oxygen species (ROS) plays a crucial role, which is produced from molecular oxygen with extracellular deposited amyloid-β (Aβ) aggregates through the reduction of a Cu2+ ion. In the presence of a small amount of redox-active Cu2+ ion, ROS is produced by the Aβ-Cu2+ complex as Aβ peptide alone is unable to generate excess ROS. Therefore, Cu2+ ion chelators are considered promising therapeutics against AD. Here, we have designed and synthesized a series of Schiff base derivatives (SB) based on 2-hydroxy aromatic aldehyde derivatives and dopamine. These SB compounds contain one copper chelating core, which captures the Cu2+ ions from the Aβ-Cu2+ complex. Thereby, it inhibits copper-induced amyloid aggregation as well as amyloid self-aggregation. It also inhibits copper-catalyzed ROS production through sequestering of Cu2+ ions. The uniqueness of our designed ligands has the dual property of dopamine, which not only acts as a ROS scavenger but also chelates the copper ion. The crystallographic analysis proves the power of the dopamine unit. Therefore, dual exploration of dopamine core can be considered as potential therapeutics for future AD treatment.
Collapse
Affiliation(s)
- Prabir Kumar Gharai
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR─Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar 342037, Rajasthan, India
| | - Juhee Khan
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR─Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar 342037, Rajasthan, India
| | - Krishnangsu Pradhan
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR─Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India
| | - Rathnam Mallesh
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR─Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar 342037, Rajasthan, India
- National Institute of Pharmaceutical Education and Research, Kolkata, Chunilal Bhawan 168, Maniktala Main Road, Kolkata 700054, India
| | - Shubham Garg
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar 342037, Rajasthan, India
| | - Mohammad Umar Arshi
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar 342037, Rajasthan, India
| | - Surajit Barman
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR─Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India
| | - Surajit Ghosh
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR─Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar 342037, Rajasthan, India
- National Institute of Pharmaceutical Education and Research, Kolkata, Chunilal Bhawan 168, Maniktala Main Road, Kolkata 700054, India
| |
Collapse
|
5
|
Padhi D, Baruah P, Ramesh M, Moorthy H, Govindaraju T. Hybrid molecules synergistically mitigate ferroptosis and amyloid-associated toxicities in Alzheimer's disease. Redox Biol 2024; 71:103119. [PMID: 38507972 PMCID: PMC10963859 DOI: 10.1016/j.redox.2024.103119] [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: 01/21/2024] [Revised: 02/22/2024] [Accepted: 03/07/2024] [Indexed: 03/22/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the build-up of extracellular amyloid β (Aβ) plaques and intracellular neurofibrillary tangles (NFTs). Ferroptosis, an iron (Fe)-dependent form of cell death plays a significant role in the multifaceted AD pathogenesis through generation of reactive oxygen species (ROS), mitochondrial damage, lipid peroxidation, and reduction in glutathione peroxidase 4 (GPX4) enzyme activity and levels. Aberrant liquid-liquid phase separation (LLPS) of tau drives the growth and maturation of NFTs contributing to AD pathogenesis. In this study, we strategically combined the structural and functional properties of gallic acid (GA) and cyclic dipeptides (CDPs) to synthesize hybrid molecules that effectively target both ferroptosis and amyloid toxicity in AD. This innovative approach marks a paradigm shift from conventional therapeutic strategies. This is the first report of a synthetic small molecule (GCTR) that effectively combats ferroptosis, simultaneously restoring enzymatic activity and enhancing cellular levels of its master regulator, GPX4. Further, GCTR disrupts Fe3+-induced LLPS of tau, and aids in attenuation of abnormal tau fibrillization. The synergistic action of GCTR in combating both ferroptosis and amyloid toxicity, bolstered by GPX4 enhancement and modulation of Fe3+-induced tau LLPS, holds promise for the development of small molecule-based novel therapeutics for AD.
Collapse
Affiliation(s)
- Dikshaa Padhi
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, Karnataka, 560064, India
| | - Prayasee Baruah
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, Karnataka, 560064, India
| | - Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, Karnataka, 560064, India
| | - Hariharan Moorthy
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, Karnataka, 560064, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bengaluru, Karnataka, 560064, India.
| |
Collapse
|
6
|
Kumar S, Mahajan A, Ambatwar R, Khatik GL. Recent Advancements in the Treatment of Alzheimer's Disease: A Multitarget-directed Ligand Approach. Curr Med Chem 2024; 31:6032-6062. [PMID: 37861025 DOI: 10.2174/0109298673264076230921065945] [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: 05/25/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 10/21/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease and one of the leading causes of progressive dementia, affecting 50 million people worldwide. Many pathogenic processes, including amyloid β aggregation, tau hyperphosphorylation, oxidative stress, neuronal death, and deterioration of the function of cholinergic neurons, are associated with its progression. The one-compound-one-target treatment paradigm was unsuccessful in treating AD due to the multifaceted nature of Alzheimer's disease. The recent development of multitarget-directed ligand research has been explored to target the complementary pathways associated with the disease. We aimed to find the key role and progress of MTDLs in treating AD; thus, we searched for the past ten years of literature on "Pub- Med", "ScienceDirect", "ACS" and "Bentham Science" using the keywords neurodegenerative diseases, Alzheimer's disease, and multitarget-directed ligands. The literature was further filtered based on the quality of work and relevance to AD. Thus, this review highlights the current advancement and advantages of multitarget-directed ligands over traditional single-targeted drugs and recent progress in their development to treat AD.
Collapse
Affiliation(s)
- Sumit Kumar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Amol Mahajan
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Ramesh Ambatwar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| | - Gopal L Khatik
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research-Raebareli, Uttar Pradesh, 226002, India
| |
Collapse
|
7
|
Baruah P, Moorthy H, Ramesh M, Padhi D, Govindaraju T. A natural polyphenol activates and enhances GPX4 to mitigate amyloid-β induced ferroptosis in Alzheimer's disease. Chem Sci 2023; 14:9427-9438. [PMID: 37712018 PMCID: PMC10498722 DOI: 10.1039/d3sc02350h] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 08/19/2023] [Indexed: 09/16/2023] Open
Abstract
Ferroptosis, an iron-dependent cell death, plays a crucial role in the pathology of Alzheimer's disease (AD). Several characteristics of AD, including excessive iron accumulation, elevated lipid peroxide and reactive oxygen species (ROS) levels, and decreased glutathione peroxidase 4 (GPX4) levels, align with the features of ferroptosis. While traditional methods of inhibiting ferroptosis have centered on chelating Fe and trapping radicals, therapeutic strategies that modulate the GPX4 axis to mitigate ferroptosis in AD are yet to be explored. This report introduces naturally occurring polyphenols (PPs) as dual-acting therapeutic agents to synergistically alleviate ferroptosis and AD. The mechanisms of action encompass modulation of amyloid and tau cascade, reduction of oxidative stress, mitochondrial rescue, and inhibition of ferroptosis. For the first time, we show that a single multifunctional molecule, tannic acid (TA) binds at the activator site of GPX4, augmenting both its activity and cellular levels, providing a conceptually innovative and integrated approach for treating AD via the GPX4-ferroptosis axis. The ability of TA to enhance GPX4 levels under conditions of AD pathology opens up newer promising therapeutic avenues for combating the crosstalk between ferroptosis and AD.
Collapse
Affiliation(s)
- Prayasee Baruah
- Bioorganic Chemistry Laboratory, New Chemistry Unit, The School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Bengaluru 560064 Karnataka India
| | - Hariharan Moorthy
- Bioorganic Chemistry Laboratory, New Chemistry Unit, The School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Bengaluru 560064 Karnataka India
| | - Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, The School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Bengaluru 560064 Karnataka India
| | - Dikshaa Padhi
- Bioorganic Chemistry Laboratory, New Chemistry Unit, The School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Bengaluru 560064 Karnataka India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, The School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) Bengaluru 560064 Karnataka India
| |
Collapse
|
8
|
García-García A, Rojas S, Rodríguez-Diéguez A. Therapy and diagnosis of Alzheimer's disease: from discrete metal complexes to metal-organic frameworks. J Mater Chem B 2023; 11:7024-7040. [PMID: 37435638 DOI: 10.1039/d3tb00427a] [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: 07/13/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder affecting 44 million people worldwide. Although many issues (pathogenesis, genetics, clinical features, and pathological aspects) are still unknown, this disease is characterized by noticeable hallmarks such as the formation of β-amyloid plaques, hyperphosphorylation of tau proteins, the overproduction of reactive oxygen species, and the reduction of acetylcholine levels. There is still no cure for AD and the current treatments are aimed at regulating the cholinesterase levels, attenuating symptoms temporarily rather than preventing the AD progression. In this context, coordination compounds are regarded as a promissing tool in AD treatment and/or diagnosis. Coordination compounds (discrete or polymeric) possess several features that make them an interesting option for developing new drugs for AD (good biocompatibility, porosity, synergetic effects of ligand-metal, fluorescence, particle size, homogeneity, monodispersity, etc.). This review discusses the recent progress in the development of novel discrete metal complexes and metal-organic frameworks (MOFs) for the treatment, diagnosis and theragnosis of AD. These advanced therapies for AD treatment are organized according to the target: Aβ peptides, hyperphosphorylated tau proteins, synaptic dysfunction, and mitochondrial failure with subsequent oxidative stress.
Collapse
Affiliation(s)
- Amalia García-García
- Department of Inorganic Chemistry, Faculty of Science, University of Granada, Av. Fuentenueva S/N, 18071 Granada, Spain.
- Centro de Química del Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, 18 sur & Av. San Claudio, Col. San Manuel, 72570 Puebla, Mexico
| | - Sara Rojas
- Department of Inorganic Chemistry, Faculty of Science, University of Granada, Av. Fuentenueva S/N, 18071 Granada, Spain.
| | - Antonio Rodríguez-Diéguez
- Department of Inorganic Chemistry, Faculty of Science, University of Granada, Av. Fuentenueva S/N, 18071 Granada, Spain.
| |
Collapse
|
9
|
Mandal S, Jana D, Dolai J, Sarkar AK, Ghorai BK, Jana NR. Biodegradable Poly(trehalose) Nanoparticle for Preventing Amyloid Beta Aggregation and Related Neurotoxicity. ACS APPLIED BIO MATERIALS 2023. [PMID: 37167565 DOI: 10.1021/acsabm.2c00771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Trehalose is a disaccharide that is capable of inhibiting protein aggregation and activating cellular autophagy. It has been shown that a polymer or nanoparticle form, terminated with multiple trehalose units, can significantly enhance the anti-amyloidogenic performance and is suitable for the treatment of neurodegenerative diseases. Here, we report a trehalose-conjugated polycarbonate-co-lactide polymer and formulation of its nanoparticles having multiple numbers of trehalose exposed on the surface. The resultant poly(trehalose) nanoparticle inhibits the aggregation of amyloid beta peptides and disintegrates matured amyloid fibrils into smaller fragments. Moreover, the poly(trehalose) nanoparticle lowers extracellular amyloid β oligomer-driven cellular stress and enhances cell viability. The presence of biodegradable polycarbonate components in the poly(trehalose) nanoparticle would enhance their application potential as an anti-amyloidogenic material.
Collapse
Affiliation(s)
- Suman Mandal
- School of Materials Science, Indian Association for the Cultivation of Science, Kolkata 700032, India
- Department of Chemistry, University of Houston, Houston, Texas 77204-5003, United States
| | - Debabrata Jana
- Department of Chemistry, Ramakrishna Mission Vivekananda Centenary College, Rahara, West Bengal 700118, India
- Department of Chemistry, Indian Institute of Engineering Science and Technology, Shibpur, Howrah 711103, India
| | - Jayanta Dolai
- School of Materials Science, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Ankan Kumar Sarkar
- School of Materials Science, Indian Association for the Cultivation of Science, Kolkata 700032, India
| | - Binay K Ghorai
- Department of Chemistry, Indian Institute of Engineering Science and Technology, Shibpur, Howrah 711103, India
| | - Nikhil R Jana
- School of Materials Science, Indian Association for the Cultivation of Science, Kolkata 700032, India
| |
Collapse
|
10
|
Zheng R, Shi S, Zhang Q, Yuan S, Guo T, Guo J, Jiang P. Molecular mechanisms of Huanglian Jiedu decoction in treating Alzheimer’s disease by regulating microbiome via network pharmacology and molecular docking analysis. Front Cell Infect Microbiol 2023; 13:1140945. [PMID: 37009506 PMCID: PMC10060893 DOI: 10.3389/fcimb.2023.1140945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023] Open
Abstract
BackgroundHuanglian Jiedu decoction (HLJDD) is a famous traditional Chinese medicine prescription, which is widely used in the treatment of Alzheimer’s disease (AD). However, the interaction between bioactive substances in HLJDD and AD-related targets has not been well elucidated.AimA network pharmacology-based approach combined with molecular docking was performed to determine the bioactives, key targets, and potential pharmacological mechanism of HLJDD against AD, through the regulation of microbial flora.Materials and methodsBioactives and potential targets of HLJDD, as well as AD-related targets, were retrieved from Traditional Chinese Medicine Systems Pharmacology Analysis Database (TCMSP). Key bioactive components, potential targets, and signaling pathways were obtained through bioinformatics analysis, including protein-protein interaction (PPI), Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Subsequently, molecular docking was performed to predict the binding of active compounds with core targets.Results102 bioactive ingredients of HLJDD and 76 HLJDD-AD-related targets were screened. Bioinformatics analysis revealed that kaempferol, wogonin, beta-sitosterol, baicalein, acacetin, isocorypalmine, (S)-canadine, (R)-canadine may be potential candidate agents. AKT1, TNF, TP53, VEGFA, FOS, PTGS2, MMP9 and CASP3 could become potential therapeutic targets. 15 important signaling pathways including the cancer pathway, VEGF signaling pathway, and NF-κB signaling pathway might play an important role in HLJDD against AD. Moreover, molecular docking analysis suggested that kaempferol, wogonin, beta-sitosterol, baicalein, acacetin, isocorypalmine, (S)-canadine, and (R)-canadine combined well with AKT1, TNF, TP53, VEGFA, FOS, PTGS2, MMP9, CASP3, respectively.ConclusionOur results comprehensively illustrated the bioactives, potential targets, and possible molecular mechanisms of HLJDD against AD. HLJDD may regulate the microbiota flora homeostasis to treat AD through multiple targets and multiple pathways. It also provided a promising strategy for the use of traditional Chinese medicine in treating human diseases.
Collapse
Affiliation(s)
- Renyuan Zheng
- Sichuan Key Laboratory of Noncoding RNA and Drugs, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Shenggan Shi
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Qin Zhang
- Sichuan Key Laboratory of Noncoding RNA and Drugs, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Shuqin Yuan
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Tong Guo
- Sichuan Key Laboratory of Noncoding RNA and Drugs, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, China
| | - Jinlin Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Peidu Jiang, ; Jinlin Guo,
| | - Peidu Jiang
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- *Correspondence: Peidu Jiang, ; Jinlin Guo,
| |
Collapse
|
11
|
Gharai PK, Khan J, Mallesh R, Garg S, Saha A, Ghosh S, Ghosh S. Vanillin Benzothiazole Derivative Reduces Cellular Reactive Oxygen Species and Detects Amyloid Fibrillar Aggregates in Alzheimer's Disease Brain. ACS Chem Neurosci 2023; 14:773-786. [PMID: 36728363 DOI: 10.1021/acschemneuro.2c00771] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The misfolding of amyloid beta (Aβ) peptides into Aβ fibrillary aggregates is a major hallmark of Alzheimer's disease (AD), which responsible for the excess production of hydrogen peroxide (H2O2), a prominent reactive oxygen species (ROS) from the molecular oxygen (O2) by the reduction of the Aβ-Cu(I) complex. The excessive production of H2O2 causes oxidative stress and inflammation in the AD brain. Here, we have designed and developed a dual functionalized molecule VBD by using π-conjugation (C═C) in the backbone structure. In the presence of H2O2, the VBD can turn into fluorescent probe VBD-1 by cleaving of the selective boronate ester group. The fluorescent probe VBD-1 can undergo intramolecular charge transfer transition (ICT) by a π-conjugative system, and as a result, its emission increases from the yellow (532 nm) to red (590 nm) region. The fluorescence intensity of VBD-1 increases by 3.5-fold upon binding with Aβ fibrillary aggregates with a high affinity (Kd = 143 ± 12 nM). Finally, the VBD reduces the cellular toxic H2O2 as proven by the CCA assay and DCFDA assay and the binding affinity of VBD-1 was confirmed by using in vitro histological staining in 8- and 18-month-old triple transgenic AD (3xTg-AD) mice brain slices.
Collapse
Affiliation(s)
- Prabir Kumar Gharai
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India.,Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar, Rajasthan 342037, India
| | - Juhee Khan
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India.,Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar, Rajasthan 342037, India
| | - Rathnam Mallesh
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India.,Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar, Rajasthan 342037, India.,National Institute of Pharmaceutical Education and Research, Kolkata, Chunilal Bhawan 168, Maniktala Main Road, Kolkata 700054, India
| | - Shubham Garg
- Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar, Rajasthan 342037, India
| | - Abhijit Saha
- Department of Chemistry, SRM Institute of Science and Technology, Kattankulathur, Chennai 603203, India
| | - Subhajit Ghosh
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India
| | - Surajit Ghosh
- Organic and Medicinal Chemistry and Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S. C. Mullick Road, Jadavpur, Kolkata 700 032, WB, India.,Department of Bioscience & Bioengineering, Indian Institute of Technology, Jodhpur, NH 65, Surpura Bypass Road, Karwar, Rajasthan 342037, India.,National Institute of Pharmaceutical Education and Research, Kolkata, Chunilal Bhawan 168, Maniktala Main Road, Kolkata 700054, India
| |
Collapse
|
12
|
Chen LL, Fan YG, Zhao LX, Zhang Q, Wang ZY. The metal ion hypothesis of Alzheimer's disease and the anti-neuroinflammatory effect of metal chelators. Bioorg Chem 2023; 131:106301. [PMID: 36455485 DOI: 10.1016/j.bioorg.2022.106301] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/13/2022] [Accepted: 11/22/2022] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD), characterized by the β-amyloid protein (Aβ) deposition and tau hyperphosphorylation, is the most common dementia with uncertain etiology. The clinical trials of Aβ monoclonal antibody drugs have almost failed, giving rise to great attention on the other etiologic hypothesis regarding AD such as metal ions dysmetabolism and chronic neuroinflammation. Mounting evidence revealed that the metal ions (iron, copper, and zinc) were dysregulated in the susceptible brain regions of AD patients, which was highly associated with Aβ deposition, tau hyperphosphorylation, neuronal loss, as well as neuroinflammation. Further studies uncovered that iron, copper and zinc could not only enhance the production of Aβ but also directly bind to Aβ and tau to promote their aggregations. In addition, the accumulation of iron and copper could respectively promote ferroptosis and cuproptosis. Therefore, the metal ion chelators were recognized as promising agents for treating AD. This review comprehensively summarized the effects of metal ions on the Aβ dynamics and tau phosphorylation in the progression of AD. Furthermore, taking chronic neuroinflammation contributes to the progression of AD, we also provided a summary of the mechanisms concerning metal ions on neuroinflammation and highlighted the metal ion chelators may be potential agents to alleviate neuroinflammation under the condition of AD. Nevertheless, more investigations regarding metal ions on neuroinflammation should be taken into practice, and the effects of metal ion chelators on neuroinflammation should gain more attention. Running title: Metal chelators against neuroinflammation.
Collapse
Affiliation(s)
- Li-Lin Chen
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Yong-Gang Fan
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Ling-Xiao Zhao
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Qi Zhang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China
| | - Zhan-You Wang
- Key Laboratory of Medical Cell Biology of Ministry of Education, Key Laboratory of Major Chronic Diseases of Nervous System of Liaoning Province, Health Sciences Institute of China Medical University, Shenyang 110122, China.
| |
Collapse
|
13
|
Roy R, Paul S. Illustrating the Effect of Small Molecules Derived from Natural Resources on Amyloid Peptides. J Phys Chem B 2023; 127:600-615. [PMID: 36638829 DOI: 10.1021/acs.jpcb.2c07607] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The onset of amyloidogenic diseases is associated with the misfolding and aggregation of proteins. Despite extensive research, no effective therapeutics are yet available to treat these chronic degenerative diseases. Targeting the aggregation of disease-specific proteins is regarded as a promising new approach to treat these diseases. In the past few years, rapid progress in this field has been made in vitro, in vivo, and in silico to generate potential drug candidates, ranging from small molecules to polymers to nanoparticles. Small molecular probes, mostly those derived from natural sources, have been of particular interest among amyloid inhibitors. Here, we summarize some of the most important natural small molecular probes which can inhibit the aggregation of Aβ, hIAPP, and α-syn peptides and discuss how their binding efficacy and preference for the peptides vary with their structure and conformation. This provides a comprehensive idea of the crucial factors which should be incorporated into the future design of novel drug candidates useful for the treatment of amyloid diseases.
Collapse
Affiliation(s)
- Rituparna Roy
- Department of Chemistry, Indian Institute of Technology, Guwahati Assam-781039, India
| | - Sandip Paul
- Department of Chemistry, Indian Institute of Technology, Guwahati Assam-781039, India
| |
Collapse
|
14
|
Rehman MU, Sehar N, Dar NJ, Khan A, Arafah A, Rashid S, Rashid SM, Ganaie MA. Mitochondrial dysfunctions, oxidative stress and neuroinflammation as therapeutic targets for neurodegenerative diseases: An update on current advances and impediments. Neurosci Biobehav Rev 2023; 144:104961. [PMID: 36395982 DOI: 10.1016/j.neubiorev.2022.104961] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 11/11/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
Neurodegenerative diseases (NDs) such as Alzheimer disease (AD), Parkinson disease (PD), and Huntington disease (HD) represent a major socio-economic challenge in view of their high prevalence yet poor treatment outcomes affecting quality of life. The major challenge in drug development for these NDs is insufficient clarity about the mechanisms involved in pathogenesis and pathophysiology. Mitochondrial dysfunction, oxidative stress and inflammation are common pathways that are linked to neuronal abnormalities and initiation of these diseases. Thus, elucidating the shared initial molecular and cellular mechanisms is crucial for recognizing novel remedial targets, and developing therapeutics to impede or stop disease progression. In this context, use of multifunctional compounds at early stages of disease development unclogs new avenues as it acts on act on multiple targets in comparison to single target concept. In this review, we summarize overview of the major findings and advancements in recent years focusing on shared mechanisms for better understanding might become beneficial in searching more potent pharmacological interventions thereby reducing the onset or severity of various NDs.
Collapse
Affiliation(s)
- Muneeb U Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
| | - Nouroz Sehar
- Centre for Translational and Clinical Research, School of Chemical & Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Nawab John Dar
- School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78992 USA
| | - Andleeb Khan
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Azher Arafah
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Shahzada Mudasir Rashid
- Division of Veterinary Biochemistry, Faculty of Veterinary Science and Animal Husbandry, SKUAST-Kashmir, Srinagar, Jammu and Kashmir, India
| | - Majid Ahmad Ganaie
- Department of Pharmacology & Toxicology, College of Dentistry and Pharmacy, Buraydah Colleges, Buraydah, Saudi Arabia
| |
Collapse
|
15
|
Ramesh M, Govindaraju T. Multipronged diagnostic and therapeutic strategies for Alzheimer's disease. Chem Sci 2022; 13:13657-13689. [PMID: 36544728 PMCID: PMC9710308 DOI: 10.1039/d2sc03932j] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/13/2022] [Indexed: 12/24/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and a major contributor to dementia cases worldwide. AD is clinically characterized by learning, memory, and cognitive deficits. The accumulation of extracellular amyloid β (Aβ) plaques and neurofibrillary tangles (NFTs) of tau are the pathological hallmarks of AD and are explored as targets for clinical diagnosis and therapy. AD pathology is poorly understood and there are no fully approved diagnosis and treatments. Notwithstanding the gap, decades of research in understanding disease mechanisms have revealed the multifactorial nature of AD. As a result, multipronged and holistic approaches are pertinent to targeting multiple biomarkers and targets for developing effective diagnosis and therapeutics. In this perspective, recent developments in Aβ and tau targeted diagnostic and therapeutic tools are discussed. Novel indirect, combination, and circulating biomarkers as potential diagnostic targets are highlighted. We underline the importance of multiplexing and multimodal detection of multiple biomarkers to generate biomarker fingerprints as a reliable diagnostic strategy. The classical therapeutics targeting Aβ and tau aggregation pathways are described with bottlenecks in the strategy. Drug discovery efforts targeting multifaceted toxicity involving protein aggregation, metal toxicity, oxidative stress, mitochondrial damage, and neuroinflammation are highlighted. Recent efforts focused on multipronged strategies to rationally design multifunctional modulators targeting multiple pathological factors are presented as future drug development strategies to discover potential therapeutics for AD.
Collapse
Affiliation(s)
- Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru Karnataka 560064 India
| |
Collapse
|
16
|
Ramesh M, Balachandra C, Andhare P, Govindaraju T. Rationally Designed Molecules Synergistically Modulate Multifaceted Aβ Toxicity, Microglial Activation, and Neuroinflammation. ACS Chem Neurosci 2022; 13:2209-2221. [PMID: 35759686 DOI: 10.1021/acschemneuro.2c00276] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Synergistic modulation of multifaceted toxicity is the key to tackle multifactorial Alzheimer's disease (AD). The etiology of AD includes amyloid β (Aβ) amyloidosis, metal ion dyshomeostasis, reactive oxygen species (ROS), oxidative stress, mitochondrial damage, and neuroinflammation. We rationally designed multifunctional modulators by integrating pharmacophores for metal chelation, antioxidant and anti-inflammatory properties, and modulation of Aβ42 aggregation on the naphthalene monoimide (NMI) scaffold. The in vitro and cellular studies of NMIs revealed that M3 synergistically modulates metal-independent and -dependent amyloid toxicity, scavenges ROS, alleviates oxidative stress, and emulates Nrf2-mediated stress response in neuronal cells. M3 effectively reduced structural and functional damage of mitochondria, reduced Cyt c levels, and rescued cells from apoptosis. The biological atomic force microscopy and Western blot analysis revealed the ability of M3 to suppress microglial activation and neuroinflammation through inhibition of the NF-κβ pathway. The synergistic action of M3 is in agreement with our design strategy to develop a multifunctional therapeutic candidate by integrating multiple pharmacophores with distinct structural and functional elements to ameliorate the multifaceted toxicity of AD.
Collapse
Affiliation(s)
- Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru, Karnataka 560064, India
| | - Chenikkayala Balachandra
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru, Karnataka 560064, India
| | - Pradhnesh Andhare
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru, Karnataka 560064, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru, Karnataka 560064, India
| |
Collapse
|
17
|
Padhi D, Govindaraju T. Mechanistic Insights for Drug Repurposing and the Design of Hybrid Drugs for Alzheimer's Disease. J Med Chem 2022; 65:7088-7105. [PMID: 35559617 DOI: 10.1021/acs.jmedchem.2c00335] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The heterogeneity and complex nature of Alzheimer's disease (AD) is attributed to several genetic risk factors and molecular culprits. The slow pace and increasing failure rate of conventional drug discovery has led to the exploration of complementary strategies based on repurposing approved drugs to treat AD. Drug repurposing (DR) is a cost-effective, low-risk, and efficient approach for identifying novel therapeutic candidates for AD treatment. Similarly, hybrid drug design through the integration of distinct pharmacophores from known or failed drugs and natural products is an interesting strategy to target the multifactorial nature of AD. In this Perspective, we discuss the potential of DR and highlight promising drug candidates that can be advanced for clinical trials, backed by a detailed discussion on their plausible mechanisms of action. Our article fosters research on the hidden potential of DR and hybrid drug design with the goal of unravelling new drugs and targets to tackle AD.
Collapse
Affiliation(s)
- Dikshaa Padhi
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur P.O., Bengaluru, Karnataka 560064, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur P.O., Bengaluru, Karnataka 560064, India
| |
Collapse
|
18
|
Pradhan LK, Sahoo PK, Chauhan S, Das SK. Recent Advances Towards Diagnosis and Therapeutic Fingerprinting for Alzheimer's Disease. J Mol Neurosci 2022; 72:1143-1165. [PMID: 35553375 DOI: 10.1007/s12031-022-02009-7] [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: 01/13/2022] [Accepted: 04/02/2022] [Indexed: 12/12/2022]
Abstract
Since the report of "a peculiar severe disease process of the cerebral cortex" by Alois Alzheimer in 1906, it was considered to be a rare condition characterized by loss of cognition, memory impairment, and pathological markers such as senile plaques or neurofibrillary tangles (NFTs). Later on, the report was published in the textbook "Psychiatrie" and the disease was named as Alzheimer's disease (AD) and was known to be the consequences of aging; however, owing to its complex etiology, there is no cure for the progressive neurodegenerative disorder. Our current understanding of the mechanisms involved in the pathogenesis of AD is still at the mechanistic level. The treatment strategies applied currently only alleviate the symptoms and co-morbidities. For instance, the available treatments such as the usage of acetylcholinesterase inhibitors and N-methyl D-aspartate antagonists have minimal impact on the disease progression and target the later aspects of the disease. The recent advancements in the last two decades have made us more clearly understand the pathophysiology of the disease which has led to the development of novel therapeutic strategies. This review gives a brief idea about the various facets of AD pathophysiology and its management through modern investigational therapies to give a new direction for development of targeted therapeutic measures.
Collapse
Affiliation(s)
- Lilesh Kumar Pradhan
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Kalinga Nagar, Bhubaneswar-751003, India
| | - Pradyumna Kumar Sahoo
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Kalinga Nagar, Bhubaneswar-751003, India
| | - Santosh Chauhan
- Autophagy Laboratory, Infectious Disease Biology Division, Institute of Life Sciences, Bhubaneswar-751023, India.
| | - Saroj Kumar Das
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to Be University), Kalinga Nagar, Bhubaneswar-751003, India.
| |
Collapse
|
19
|
Sciacca MF, Naletova I, Giuffrida ML, Attanasio F. Semax, a Synthetic Regulatory Peptide, Affects Copper-Induced Abeta Aggregation and Amyloid Formation in Artificial Membrane Models. ACS Chem Neurosci 2022; 13:486-496. [PMID: 35080861 PMCID: PMC8855339 DOI: 10.1021/acschemneuro.1c00707] [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] [Indexed: 12/02/2022] Open
Abstract
![]()
Alzheimer’s
disease, the most common form of dementia, is
characterized by the aggregation of amyloid beta protein (Aβ).
The aggregation and toxicity of Aβ are strongly modulated by
metal ions and phospholipidic membranes. In particular, Cu2+ ions play a pivotal role in modulating Aβ aggregation. Although
in the last decades several natural or synthetic compounds were evaluated
as candidate drugs, to date, no treatments are available for the pathology.
Multifunctional compounds able to both inhibit fibrillogenesis, and
in particular the formation of oligomeric species, and prevent the
formation of the Aβ:Cu2+ complex are of particular
interest. Here we tested the anti-aggregating properties of a heptapeptide,
Semax, an ACTH-like peptide, which is known to form a stable complex
with Cu2+ ions and has been proven to have neuroprotective
and nootropic effects. We demonstrated through a combination of spectrofluorometric,
calorimetric, and MTT assays that Semax not only is able to prevent
the formation of Aβ:Cu2+ complexes but also has anti-aggregating
and protective properties especially in the presence of Cu2+. The results suggest that Semax inhibits fiber formation by interfering
with the fibrillogenesis of Aβ:Cu2+ complexes.
Collapse
Affiliation(s)
- Michele F.M. Sciacca
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Irina Naletova
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Maria Laura Giuffrida
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| | - Francesco Attanasio
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Via Paolo Gaifami, 18, Catania 95126, Italy
| |
Collapse
|
20
|
Konar M, Ghosh D, Samanta S, Govindaraju T. Combating amyloid-induced cellular toxicity and stiffness by designer peptidomimetics. RSC Chem Biol 2022; 3:220-226. [PMID: 35360886 PMCID: PMC8827053 DOI: 10.1039/d1cb00235j] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 12/21/2021] [Indexed: 12/24/2022] Open
Abstract
Amyloid beta (Aβ) aggregation species-associated cellular stress instigates cytotoxicity and adverse cellular stiffness in neuronal cells. The study and modulation of these adverse effects demand immediate attention to tackle Alzheimer's disease (AD). We present a de novo design, synthesis and evaluation of Aβ14-23 peptidomimetics with cyclic dipeptide (CDP) units at defined positions. Our study identified AkdNMC with CDP units at the middle, N- and C-termini as a potent candidate to understand and ameliorate Aβ aggregation-induced cellular toxicity and adverse stiffness. Aβ14-23 peptidomimetics incorporated with cyclic dipeptide-based unnatural amino acid at defined positions serve as potential candidates to understand and ameliorate amyloid-induced cellular toxicity and physio-mechanical anomalies.![]()
Collapse
Affiliation(s)
- Mouli Konar
- Bioorganic Chemistry Laboratory, New Chemistry Unit and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru 560064, Karnataka, India
| | - Debasis Ghosh
- Bioorganic Chemistry Laboratory, New Chemistry Unit and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru 560064, Karnataka, India
| | - Sourav Samanta
- Bioorganic Chemistry Laboratory, New Chemistry Unit and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru 560064, Karnataka, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit and School of Advanced Materials (SAMat), Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru 560064, Karnataka, India
| |
Collapse
|
21
|
Yokoya M, Nakai K, Kawashima M, Kurakado S, Sirimangkalakitti N, Kino Y, Sugita T, Kimura S, Yamanaka M, Saito N. Inhibition of BACE1 and amyloid β aggregation by polyketide from Streptomyces sp. Chem Biol Drug Des 2021; 99:264-276. [PMID: 34757664 DOI: 10.1111/cbdd.13980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 10/12/2021] [Accepted: 11/06/2021] [Indexed: 11/27/2022]
Abstract
Alzheimer's disease (AD) causes cognitive impairment in the elderly and is a severe problem worldwide. One of the major reasons for the pathogenesis of AD is thought to be due to the accumulation of amyloid beta (Aβ) peptides that result in neuronal cell death in the brain. In this study, bioassay-guided fractionation was performed to develop seed compounds for anti-AD drugs that can act as dual inhibitors of BACE1 and Aβ aggregation from secondary metabolites produced by Streptomyces sp. To improve the solubility, the crude extracts were methylated with trimethylsilyl (TMS) diazomethane and then purified to yield polyketides 1-5, including the new compound 1. We synthesized the compounds 6 and 7 (original compounds 2 and 3, respectively), and their activities were evaluated. KS-619-1, the demethylated form of 4 and 5, was isolated and evaluated for its inhibitory activity. The IC50 values for BACE1 and Aβ aggregation were found to be 0.48 and 1.1 μM, respectively, indicating that KS-619-1 could be a lead compound for the development of therapeutic agents for AD.
Collapse
Affiliation(s)
- Masashi Yokoya
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, Kiyose, Japan
| | - Keiyo Nakai
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, Kiyose, Japan
| | - Miki Kawashima
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, Kiyose, Japan
| | - Sanae Kurakado
- Department of Microbiology, Meiji Pharmaceutical University, Kiyose, Japan
| | - Natchanun Sirimangkalakitti
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, Kiyose, Japan.,Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Kiyose, Japan
| | - Yoshihiro Kino
- Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Kiyose, Japan
| | - Takashi Sugita
- Department of Microbiology, Meiji Pharmaceutical University, Kiyose, Japan
| | - Shinya Kimura
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, Kiyose, Japan
| | - Masamichi Yamanaka
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, Kiyose, Japan
| | - Naoki Saito
- Department of Pharmaceutical Chemistry, Meiji Pharmaceutical University, Kiyose, Japan
| |
Collapse
|
22
|
Demasi M, Augusto O, Bechara EJH, Bicev RN, Cerqueira FM, da Cunha FM, Denicola A, Gomes F, Miyamoto S, Netto LES, Randall LM, Stevani CV, Thomson L. Oxidative Modification of Proteins: From Damage to Catalysis, Signaling, and Beyond. Antioxid Redox Signal 2021; 35:1016-1080. [PMID: 33726509 DOI: 10.1089/ars.2020.8176] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The systematic investigation of oxidative modification of proteins by reactive oxygen species started in 1980. Later, it was shown that reactive nitrogen species could also modify proteins. Some protein oxidative modifications promote loss of protein function, cleavage or aggregation, and some result in proteo-toxicity and cellular homeostasis disruption. Recent Advances: Previously, protein oxidation was associated exclusively to damage. However, not all oxidative modifications are necessarily associated with damage, as with Met and Cys protein residue oxidation. In these cases, redox state changes can alter protein structure, catalytic function, and signaling processes in response to metabolic and/or environmental alterations. This review aims to integrate the present knowledge on redox modifications of proteins with their fate and role in redox signaling and human pathological conditions. Critical Issues: It is hypothesized that protein oxidation participates in the development and progression of many pathological conditions. However, no quantitative data have been correlated with specific oxidized proteins or the progression or severity of pathological conditions. Hence, the comprehension of the mechanisms underlying these modifications, their importance in human pathologies, and the fate of the modified proteins is of clinical relevance. Future Directions: We discuss new tools to cope with protein oxidation and suggest new approaches for integrating knowledge about protein oxidation and redox processes with human pathophysiological conditions. Antioxid. Redox Signal. 35, 1016-1080.
Collapse
Affiliation(s)
- Marilene Demasi
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, Brazil
| | - Ohara Augusto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Etelvino J H Bechara
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Renata N Bicev
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda M Cerqueira
- CENTD, Centre of Excellence in New Target Discovery, Instituto Butantan, São Paulo, Brazil
| | - Fernanda M da Cunha
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana Denicola
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Fernando Gomes
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Luis E S Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Lía M Randall
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Cassius V Stevani
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Leonor Thomson
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
23
|
Wu J, Kou X, Ju H, Zhang H, Yang A, Shen R. Design, synthesis and biological evaluation of naringenin carbamate derivatives as potential multifunctional agents for the treatment of Alzheimer's disease. Bioorg Med Chem Lett 2021; 49:128316. [PMID: 34391893 DOI: 10.1016/j.bmcl.2021.128316] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/04/2021] [Accepted: 08/07/2021] [Indexed: 01/14/2023]
Abstract
A series of naringenin derivatives were designed and synthesized as multifunctional anti-Alzheimer's disease (AD) agents. The results showed that these derivatives displayed moderate-to-good acetylcholinesterase (AChE) and butyrylcholinesterase (BuChE) inhibitory activities at the micromolar range (IC50, 12.91 ~ 62.52 μM for AChE and 0.094 ~ 13.72 μM for BuChE). Specifically, compound 1 showed the highest inhibitory activity against BuChE with the IC50 value of (0.094 ± 0.0054) μM. A Lineweaver-Burk plot and molecular docking studies demonstrated that 1 targeted both the catalytically active site (CAS) and the peripheral anion site (PAS) of BuChE. Besides, all derivatives showed excellent hydroxyl free radicals (·OH) scavenging ability than vitamin C and cyclic voltammetry results displayed that 1 could effectively scavenge superoxide anion radical (·O2-). In addition, compound 1 displayed good metal chelating properties and had anti-Aβ aggregation activities. Therefore, compound 1 might be the potential anti-AD agent for further developments.
Collapse
Affiliation(s)
- Jiarui Wu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Xiaodi Kou
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Hui Ju
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Hongwei Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Aihong Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| | - Rui Shen
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
24
|
Ramesh M, Acharya A, Murugan NA, Ila H, Govindaraju T. Thiophene-Based Dual Modulators of Aβ and Tau Aggregation. Chembiochem 2021; 22:3348-3357. [PMID: 34546619 DOI: 10.1002/cbic.202100383] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/21/2021] [Indexed: 01/29/2023]
Abstract
Alzheimer's disease is characterized by the accumulation of amyloid beta (Aβ) and Tau aggregates in the brain, which induces various pathological events resulting in neurodegeneration. There have been continuous efforts to develop modulators of the Aβ and Tau aggregation process to halt or modify disease progression. A few small-molecule-based inhibitors that target both Aβ and Tau pathology have been reported. Here, we report the screening of a targeted library of small molecules to modulate Aβ and Tau aggregation together with their in vitro, in silico and cellular studies. In vitro ThT fluorescence assay, dot blot assay, gel electrophoresis and transmission electron microscopy (TEM) results have shown that thiophene-based lead molecules effectively modulate Aβ aggregation and inhibit Tau aggregation. In silico studies performed by employing molecular docking, molecular dynamics and binding-free energy calculations have helped in understanding the mechanism of interaction of the lead thiophene compounds with Aβ and Tau fibril targets. In cellulo studies revealed that the lead candidate is biocompatible and effectively ameliorates neuronal cells from Aβ and Tau-mediated amyloid toxicity.
Collapse
Affiliation(s)
- Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru, 560064, Karnataka, India
| | - Anand Acharya
- New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru, 560064, Karnataka, India
| | - N Arul Murugan
- Department of Computer Science, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, 10044, Stockholm, Sweden
| | - Hiriyakkanavar Ila
- New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru, 560064, Karnataka, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru, 560064, Karnataka, India
| |
Collapse
|
25
|
Zhao W, Jiang L, Wang W, Sang J, Sun Q, Dong Q, Li L, Lu F, Liu F. Design of carboxylated single-walled carbon nanotubes as highly efficient inhibitors against Aβ40 fibrillation based on the HyBER mechanism. J Mater Chem B 2021; 9:6902-6914. [PMID: 34612337 DOI: 10.1039/d1tb00920f] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Misfolding and the subsequent self-assembly of amyloid-β protein (Aβ) is very important in the occurrence of Alzheimer's disease (AD). Thus, inhibition of Aβ aggregation is currently an effective method to alleviate and treat AD. Herein, a carboxylated single-walled carbon nanotube (SWCNT-COOH) was rationally designed based on the hydrophobic binding-electrostatic repulsion (HyBER) mechanism. The inhibitory effect of SWCNT-COOH on Aβ fibrillogenesis was first studied. Based on the results of thioflavin T fluorescence and atomic force microscopy imaging assays, it was shown that SWCNT-COOH can not only effectively inhibit Aβ aggregation, but also depolymerize the mature fibrils of Aβ. In addition, its inhibitory action will be affected by the content of carboxyl groups. Moreover, the influence of SWCNT-COOH on cytotoxicity induced by Aβ was investigated by the MTT method. It was found that SWCNT-COOH can produce an anti-Aβ neuroprotective effect in vitro. Molecular dynamics simulations showed that SWCNT-COOH significantly destroyed the overall and internal structural stability of an Aβ40 trimer. Moreover, SWCNT-COOH interacted strongly with the N-terminal region, turn region and C-terminal region of the Aβ40 trimer via hydrogen bonds, salt bridges and π-π interactions, which triggered a large structural disturbance of the Aβ40 trimer, reduced the β-sheet content of the Aβ40 trimer and led to more disorder in these regions. All the above data not only reveal the suppressive effect of SWCNT-COOH on Aβ aggregation, but also reveal its inhibitory mechanism, which provides a useful clue to exploit anti-Aβ drugs in the future.
Collapse
Affiliation(s)
- Wenping Zhao
- Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education, Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science & Technology, Tianjin 300457, P. R. China.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Emodin derivatives with multi-factor anti-AD activities: AChE inhibitor, anti-oxidant and metal chelator. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.130459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
27
|
Phadke AV, Tayade AA, Khambete MP. Therapeutic potential of ferulic acid and its derivatives in Alzheimer's disease-A systematic review. Chem Biol Drug Des 2021; 98:713-721. [PMID: 34240555 DOI: 10.1111/cbdd.13922] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/05/2021] [Accepted: 06/20/2021] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder primarily caused by accumulation of amyloid-beta (Aβ) peptide extracellularly and neurofibrillary tangles intracellularly. Recently, it has been shown that oxidative stress and mitochondrial dysregulation play an important role in pathology of AD. Therefore, modulating various targets such as Aβ aggregation, neuro-inflammation, and oxidative stress, genetic factors such as Apolipoprotein E gene (ApoE) are some of the ways to manage AD. Studying the natural products which can act as multifunctional agents could be key toward discovering new therapeutics. Ferulic acid (FA) represents one such natural product, which has exhibited great potential in this regard. Found in the plant cell walls, FA is an antioxidant, free radical scavenger with anti-inflammatory activity. Taking this into consideration, over the years, various derivatives have been reported as anti-AD molecules based on structure of FA. The present review explores the role of FA and its derivatives as therapeutic agents in AD.
Collapse
Affiliation(s)
- Apoorva V Phadke
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Apurva A Tayade
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | | |
Collapse
|
28
|
Savran T, Nihan Karuk Elmas S, Akin Geyik G, Bostanci A, Aydin D, Nur Arslan F, Sadi G, Yilmaz I. “Turn‐on” Fluorescence Chemosensor Based Probing of Cu
2+
with Excellent Sensitivity: Experimental Study, DFT Calculations and Application in Living Cells and Natural Waters. ChemistrySelect 2021. [DOI: 10.1002/slct.202101060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Tahir Savran
- Karamanoglu Mehmetbey University, Kamil Ozdag Science Faculty, Department of Chemistry 70100 Karaman Turkey
| | - Sukriye Nihan Karuk Elmas
- Karamanoglu Mehmetbey University, Kamil Ozdag Science Faculty, Department of Chemistry 70100 Karaman Turkey
| | - Gonul Akin Geyik
- Karamanoglu Mehmetbey University, Kamil Ozdag Science Faculty, Department of Chemistry 70100 Karaman Turkey
| | - Aykut Bostanci
- Karamanoglu Mehmetbey University, Kamil Ozdag Science Faculty, Department of Chemistry 70100 Karaman Turkey
| | - Duygu Aydin
- Karamanoglu Mehmetbey University, Kamil Ozdag Science Faculty, Department of Chemistry 70100 Karaman Turkey
| | - Fatma Nur Arslan
- Karamanoglu Mehmetbey University, Kamil Ozdag Science Faculty, Department of Chemistry 70100 Karaman Turkey
| | - Gökhan Sadi
- Karamanoglu Mehmetbey University, Kamil Ozdag Science Faculty, Department of Chemistry 70100 Karaman Turkey
| | - Ibrahim Yilmaz
- Karamanoglu Mehmetbey University, Kamil Ozdag Science Faculty, Department of Chemistry 70100 Karaman Turkey
| |
Collapse
|
29
|
Wang K, Wang L, Chen L, Peng C, Luo B, Mo J, Chen W. Intranasal administration of dauricine loaded on graphene oxide: multi-target therapy for Alzheimer's disease. Drug Deliv 2021; 28:580-593. [PMID: 33729067 PMCID: PMC7971267 DOI: 10.1080/10717544.2021.1895909] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is a degenerative disease of the central nervous system characterized by progressive cognitive and memory-related impairment. However, current therapeutic treatments have not proved sufficiently effective, mainly due to the complicated pathogenesis of the disease. In this study, a nano-formulation of graphene oxide (GO) loaded with dauricine (Dau) was investigated in terms of the combined anti-inflammatory and anti-oxidative stress effects of Dau and the inhibition of misfolding and aggregation of the amyloid-β (Aβ) protein by GO. Both in vivo and in vitro models were induced using Aβ1-42, and the formulation was administered nasally in mice. The results showed that GO loaded with Dau greatly reduced oxidative stress through increasing superoxide dismutase levels and decreasing reactive oxygen species and malondialdehyde levels in vitro; it also alleviated the cognitive memory deficits and brain glial cell activation in mice with Aβ1-42-induced AD. This proved that GO loaded with Dau could protect against Aβ1-42-induced oxidative damage and apoptosis in both in vitro and in vivo AD models; therefore, GO loaded with Dau has the potential to be an effective and agent for the rapid treatment of AD.
Collapse
Affiliation(s)
- Kaixuan Wang
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Lingfeng Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Zhejiang, China
| | - Ling Chen
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Chiwei Peng
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Beijiao Luo
- School of Pharmacy, Guilin Medical University, Guilin, China
| | - Jingxin Mo
- Department of Pharmacy, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Wei Chen
- Department of Pharmacy, Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
30
|
Maity D, Howarth M, Vogel MC, Magzoub M, Hamilton AD. Peptidomimetic-Based Vesicles Inhibit Amyloid-β Fibrillation and Attenuate Cytotoxicity. J Am Chem Soc 2021; 143:3086-3093. [PMID: 33600171 DOI: 10.1021/jacs.0c09967] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
An interruption in Aβ homeostasis leads to the deposit of neurotoxic amyloid plaques and is associated with Alzheimer's disease. A supramolecular strategy based on the assembly of peptidomimetic agents into functional vesicles has been conceived for the simultaneous inhibition of Aβ42 fibrillation and expedited clearance of Aβ42 aggregates. Tris-pyrrolamide peptidomimetic, ADH-353, contains one hydrophobic N-butyl and two hydrophilic N-propylamine side chains and readily forms vesicles under physiological conditions. These vesicles completely rescue both mouse neuroblastoma N2a and human neuroblastoma SH-SY5Y cells from the cytotoxicity that follows from Aβ42 misfolding likely in mitochondria. Biophysical studies, including confocal imaging, demonstrate the biocompatibility and selectivity of the approach toward this aberrant protein assembly in cellular milieu.
Collapse
Affiliation(s)
- Debabrata Maity
- Department of Chemistry, New York University, New York, New York 10003, United States
| | - Madeline Howarth
- Biology Program, New York University Abu Dhabi, P.O. Box 129188, Saadiyat Island Campus, Abu Dhabi, United Arab Emirates
| | - Maria C Vogel
- Biology Program, New York University Abu Dhabi, P.O. Box 129188, Saadiyat Island Campus, Abu Dhabi, United Arab Emirates
| | - Mazin Magzoub
- Biology Program, New York University Abu Dhabi, P.O. Box 129188, Saadiyat Island Campus, Abu Dhabi, United Arab Emirates
| | - Andrew D Hamilton
- Department of Chemistry, New York University, New York, New York 10003, United States
| |
Collapse
|
31
|
Samanta S, Rajasekhar K, Ramesh M, Murugan NA, Alam S, Shah D, Clement JP, Govindaraju T. Naphthalene Monoimide Derivative Ameliorates Amyloid Burden and Cognitive Decline in a Transgenic Mouse Model of Alzheimer's Disease. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000225] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Sourav Samanta
- Bioorganic Chemistry Laboratory New Chemistry Unit Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru 560064 India
| | - Kolla Rajasekhar
- Bioorganic Chemistry Laboratory New Chemistry Unit Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru 560064 India
| | - Madhu Ramesh
- Bioorganic Chemistry Laboratory New Chemistry Unit Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru 560064 India
| | - Natarajan Arul Murugan
- Department of Theoretical Chemistry and Biology School of Chemistry Biotechnology and Health KTH Royal Institute of Technology S‐106 91 Stockholm Sweden
| | - Shadab Alam
- Bioorganic Chemistry Laboratory New Chemistry Unit Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru 560064 India
| | - Devanshi Shah
- Neuroscience Unit Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru 560064 India
| | - James Premdas Clement
- Neuroscience Unit Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru 560064 India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory New Chemistry Unit Jawaharlal Nehru Centre for Advanced Scientific Research Jakkur P.O. Bengaluru 560064 India
| |
Collapse
|
32
|
Xu S, Wang W, Dong X, Sun Y. Molecular Insight into Cu 2+-Induced Conformational Transitions of Amyloid β-Protein from Fast Kinetic Analysis and Molecular Dynamics Simulations. ACS Chem Neurosci 2021; 12:300-310. [PMID: 33401892 DOI: 10.1021/acschemneuro.0c00502] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cu2+-mediated amyloid β-protein (Aβ) aggregation is implicated in the pathogenesis of Alzheimer's disease, so it is of significance to understand Cu2+-mediated conformational transitions of Aβ. Herein, four Aβ mutants were created by using the environment-sensitive cyanophenylalanine to respectively substitute F4, Y10, F19, and F20 residues of Aβ40. By using stopped-flow fluorescence spectroscopy and molecular dynamics (MD) simulations, the early stage conformational transitions of the mutants mediated by Cu2+ binding were investigated. The fast kinetics unveils that Cu2+ has more significant influence on the conformational changes of N-terminal (F4 and Y10) than on the central hydrophobic core (CHC, F19, and F20) under different pH conditions (pH 6.6-8.0), especially Y10. Interestingly, lag periods of the conformational transitions are observed for the F19 and F20 mutants at pH 8.0, indicating the slow response of the two mutation sites on the conformational transitions. More importantly, significantly longer lag periods for F20 than for F19 indicate the conduction of the transition from F19 to F20. The conduction time (difference in lag period) decreases from 4.5 s at Cu2+ = 0 to undetectable (<1 ms) at Cu2+ = 10 μM. The significant difference in the response time of F19 and F20 and the fast local conformational changes of Y10 imply that the conformational transitions of Aβ start around Y10. MD simulations support the observation of hydrophobicity increase at N-terminal during the conformational transitions of Aβ-Cu2+. It also reveals that Y10 is immediately approached by Cu2+, supporting the speculation that the starting point of conformational transitions of Aβ is near Y10. The work has provided molecular insight into the early stage conformational transitions of Aβ40 mediated by Cu2+.
Collapse
Affiliation(s)
- Shaoying Xu
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Wenjuan Wang
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Xiaoyan Dong
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| | - Yan Sun
- Department of Biochemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- Key Laboratory of Systems Bioengineering and Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300350, China
| |
Collapse
|
33
|
Xie YQ, Zhang YM, Li ZH, Qi XN, Yao H, Shi BB, Qu WJ, Wei TB, Lin Q. A novel highly sensitive dual-channel chemical sensor for sequential recognition of Cu 2+ and CN − in aqueous media and its bioimaging applications in living cells. NEW J CHEM 2021. [DOI: 10.1039/d1nj03548g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A simple and unique dual-channel chemical probe (DH) was designed and synthesized, which not only realized sequential recognition of Cu2+ and CN− by colorimetric and fluorometric methods, but also realized fluorescence detection of CN−.
Collapse
Affiliation(s)
- Yong-Qiang Xie
- Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, Gansu, 730070, P. R. China
- Experimental & Training Teaching Centers, Gansu University of Chinese Medicine Lanzhou, Gansu, 730000, P. R. China
| | - You-Ming Zhang
- Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, Gansu, 730070, P. R. China
- Gansu Natural Energy Research Institute, Lanzhou, 730046, P. R. China
| | - Zhao-Hui Li
- Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, Gansu, 730070, P. R. China
| | - Xiao-Ni Qi
- Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, Gansu, 730070, P. R. China
| | - Hong Yao
- Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, Gansu, 730070, P. R. China
| | - Bing-Bing Shi
- Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, Gansu, 730070, P. R. China
| | - Wen-Juan Qu
- Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, Gansu, 730070, P. R. China
| | - Tai-Bao Wei
- Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, Gansu, 730070, P. R. China
| | - Qi Lin
- Key Laboratory of Eco-Environment-Related Polymer Materials, Ministry of Education of China, Key Laboratory of Polymer Materials of Gansu Province, College of Chemistry and Chemical Engineering, Northwest Normal University, Lanzhou, Gansu, 730070, P. R. China
| |
Collapse
|
34
|
Kaur A, Goyal D, Goyal B. An α-helix mimetic oligopyridylamide, ADH-31, modulates Aβ 42 monomer aggregation and destabilizes protofibril structures: insights from molecular dynamics simulations. Phys Chem Chem Phys 2020; 22:28055-28073. [PMID: 33289734 DOI: 10.1039/d0cp04672h] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD), an epidemic growing worldwide due to no effective medical aid available in the market, is a neurological disorder. AD is known to be directly associated with the toxicity of amyloid-β (Aβ) aggregates. In search of potent inhibitors of Aβ aggregation, Hamilton and co-workers reported an α-helix mimetic, ADH-31, which acts as a powerful antagonist of Aβ42 aggregation. To identify the key interactions between protein-ligand complexes and to gain insights into the inhibitory mechanism of ADH-31 against Aβ42 aggregation, molecular dynamics (MD) simulations were performed in the present study. The MD simulations highlighted that ADH-31 showed distinct binding capabilities with residues spanning from the N-terminal to the central hydrophobic core (CHC) region of Aβ42 and restricted the conformational transition of the helix-rich structure of Aβ42 into another form of secondary structures (coil/turn/β-sheet). Hydrophobic contacts, hydrogen bonding and π-π interaction contribute to the strong binding between ADH-31 and Aβ42 monomer. The Dictionary of Secondary Structure of Proteins (DSSP) analysis highlighted that the probability of helical content increases from 38.5% to 50.2% and the turn content reduces from 14.7% to 6.2% with almost complete loss of the β-sheet structure (4.5% to 0%) in the Aβ42 monomer + ADH-31 complex. The per-residue binding free energy analysis demonstrated that Arg5, Tyr10, His14, Gln15, Lys16, Val18, Phe19 and Lys28 residues of Aβ42 are responsible for the favourable binding free energy in Aβ42 monomer + ADH-31 complex, which is consistent with the 2D HSQC NMR of the Aβ42 monomer that depicted a change in the chemical shift of residues spanning from Glu11 to Phe20 in the presence of ADH-31. The MD simulations highlighted the prevention of sampling of amyloidogenic β-strand conformations in Aβ42 trimer in the presence of ADH-31 as well as the ability of ADH-31 to destabilize Aβ42 trimer and protofibril structures. The lower binding affinity between Aβ42 trimer chains in the presence of ADH-31 highlights the destabilization of the Aβ42 trimer structure. Overall, MD results highlighted that ADH-31 inhibited Aβ42 aggregation by constraining Aβ peptides into helical conformation and destabilized Aβ42 trimer as well as protofibril structures. The present study provides a theoretical insight into the atomic level details of the inhibitory mechanism of ADH-31 against Aβ42 aggregation as well as protofibril destabilization and could be implemented in the structure-based drug design of potent therapeutic agents for AD.
Collapse
Affiliation(s)
- Anupamjeet Kaur
- Department of Chemistry, Faculty of Basic and Applied Sciences, Sri Guru Granth Sahib World University, Fatehgarh Sahib-140406, Punjab, India.
| | | | | |
Collapse
|
35
|
Onça LO, de Souza JCP, Dos Santos IGN, Santos EDS, Soares SM, Diniz PHGD. A new highly selective colorimetric Schiff base chemosensor for determining the copper content in artisanal cachaças. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2020; 243:118783. [PMID: 32818693 DOI: 10.1016/j.saa.2020.118783] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/18/2020] [Accepted: 07/19/2020] [Indexed: 06/11/2023]
Abstract
This work demonstrated the feasibility of applying the Schiff base 5-bromo-2-salicyl-beta-alanine as a colorimetric chemosensor for the spectrophotometric quantification of the copper content in artisanal cachaças. For this, the experimental conditions were optimized to obtain an efficient, sensitive, reversible, and highly selective chemosensor to Cu2+ ions. The complex stoichiometry was 1:1, with a formation constant of 5.82 × 102 L mol-1 and molar absorptivity of 5.82 × 103 mol L-1 cm-1. Then, a spectrophotometric analytical method was developed and validated according to the Brazilian legislation. The linearity of the analytical curve was demonstrated by ANOVA, at a confidence level of 95%. The limits of detection and quantification were 0.0659 and 0.200 mg L-1, respectively. The coefficients of variation for both the intra- and inter-day precisions were lower than 3.83%, and the accuracy presented a mean recovery of 100.55 ± 2.87%. The absence of a matrix effect was confirmed by the standard addition method, and the copper content in three artisanal cachaças from different geographical origins was estimated as lower than 2.93 mg L-1. This result was in accordance with the Brazilian legislation but reinforces the need to carry out stricter quality control to achieve exportation standards. Therefore, the proposed method can be considered a simple, selective, linear, precise, and accurate tool that involves only a simple complexation reaction through the addition of the chemosensor solution in a buffered medium. As a consequence, the simplicity, practicality, rapidity, and low cost of synthesis of the proposed Schiff base chemosensor are highlighted.
Collapse
Affiliation(s)
- Larissa Oliveira Onça
- Programa de Pós-Graduação em Química Pura e Aplicada (POSQUIPA), Centro das Ciências Exatas e das Tecnologias (CCET), Universidade Federal do Oeste da Bahia (UFOB), 47.810-059 Barreiras, BA, Brazil
| | - Joseana Caroline Palmeira de Souza
- Undergraduate Course of Chemistry, Centro das Ciências Exatas e das Tecnologias (CCET), Universidade Federal do Oeste da Bahia (UFOB), 47.810-059 Barreiras, BA, Brazil
| | - Izabela Gessyane Nogueira Dos Santos
- Undergraduate Course of Chemistry, Centro das Ciências Exatas e das Tecnologias (CCET), Universidade Federal do Oeste da Bahia (UFOB), 47.810-059 Barreiras, BA, Brazil
| | - Emerson de Sousa Santos
- Undergraduate Course of Chemistry, Centro das Ciências Exatas e das Tecnologias (CCET), Universidade Federal do Oeste da Bahia (UFOB), 47.810-059 Barreiras, BA, Brazil
| | - Sérgio Macêdo Soares
- Programa de Pós-Graduação em Química Pura e Aplicada (POSQUIPA), Centro das Ciências Exatas e das Tecnologias (CCET), Universidade Federal do Oeste da Bahia (UFOB), 47.810-059 Barreiras, BA, Brazil
| | - Paulo Henrique Gonçalves Dias Diniz
- Programa de Pós-Graduação em Química Pura e Aplicada (POSQUIPA), Centro das Ciências Exatas e das Tecnologias (CCET), Universidade Federal do Oeste da Bahia (UFOB), 47.810-059 Barreiras, BA, Brazil.
| |
Collapse
|
36
|
Mondal S, Vashi Y, Ghosh P, Roy D, Barthakur M, Kumar S, Iyer PK. Amyloid Targeting "Artificial Chaperone" Impairs Oligomer Mediated Neuronal Damage and Mitochondrial Dysfunction Associated with Alzheimer's Disease. ACS Chem Neurosci 2020; 11:3277-3287. [PMID: 32941009 DOI: 10.1021/acschemneuro.0c00387] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is an irreversible memory disorder associated with multiple neuropathological events including amyloid aggregation that triggers oxidative stress and mitochondrial dysfunction in humans. Herein, a new artificial chaperone, benzimidazole functionalized polyfluorene (PFBZ) is reported to efficiently sequester toxic amyloid beta (Aβ) by binding at their 'amyloidogenic domain' (Aβ16-21) with unprecedented selectivity and prevent amyloid-mediated neuronal damage in a wild-type (WT) mouse model. An accurate dose of PFBZ chaperone successfully attenuated an amyloid triggered internal hemorrhage and pyknosis in the cerebral cortex of WT mice. The structural advantage of the polymer results in an efficient Cu(II) chelation arresting a redox cycle to prevent reactive oxygen species (ROS) generation and protect mitochondria from ROS mediated damage. This was further evidenced by caspase activation and mitochondrial membrane potential (MMP) biomarkers and was complemented by brain histology and electron microscopy data which revealed that the PFBZ chaperone provided a protective coating over the amyloid surface and resists from interacting with cell membrane and prevents inducing toxicity. This conjugated polymer artificial chaperone-based nanodrug showed exceptional properties such as its multipotent and highly biocompatible nature, the first of its kind with specific amyloid (Aβ16-21) targeting behavior, bioimaging, and BBB permeability with a potential to suppress amyloid triggered neurotoxicity implicated in numerous human disorders through a rare synergistic mechanism.
Collapse
Affiliation(s)
- Subrata Mondal
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Yoya Vashi
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Priyam Ghosh
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Dhrubajyoti Roy
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Manash Barthakur
- Department of Zoology, Pub Kamrup College, Baihata Chariali, Kamrup 781381, Assam, India
| | - Sachin Kumar
- Department of Bioscience and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Parameswar Krishnan Iyer
- Department of Chemistry, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
- Center for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| |
Collapse
|
37
|
So H, Lee M, Kim C. A Unique Thiosemicarbazide‐Based Colorimetric Chemosensor for Fe
2+
in Pure Aqueous Solution with the Lowest Detection Limit. ChemistrySelect 2020. [DOI: 10.1002/slct.202002062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Haeri So
- Department of Fine Chemistry Seoul National Univ. of Sci. and Tech. (SNUT) Seoul 01188 Korea
| | - Minji Lee
- Department of Fine Chemistry Seoul National Univ. of Sci. and Tech. (SNUT) Seoul 01188 Korea
| | - Cheal Kim
- Department of Fine Chemistry Seoul National Univ. of Sci. and Tech. (SNUT) Seoul 01188 Korea
| |
Collapse
|
38
|
Zhang H, Sang J, Li L, Jiang L, Lu F, He S, Cui W, Zhang X, Liu F. Molecular basis for the inhibitory effects of 5-hydroxycyclopenicillone on the conformational transition of Aβ 40 monomer. J Biomol Struct Dyn 2020; 39:6440-6451. [PMID: 32723218 DOI: 10.1080/07391102.2020.1799863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Previous studies have indicated that 5-hydroxycyclopenicillone (HCP), an active compound derived from marine sponge, could inhibit oligomerization of amyloid β-protein (Aβ). However, the molecular basis for the interaction between HCP and Aβ remains unclear. Herein, all-atom molecular dynamics (MD) simulations were used to explore the conformational conversion of an Aβ40 monomer at different concentrations (0-40 mM) of HCP at the atomic level. It is confirmed that the conformational transition of the Aβ40 monomer is prevented by HCP in a concentration-dependent manner in silico. In 40 mM HCP solution, the initial α-helix-rich conformation of Aβ40 monomer is kept under the action of HCP. The intra-peptide hydrophobic collapse and D23-K28 salt bridge are prevented by HCP. Moreover, it is indicated that the non-polar binding energy dominates the binding between HCP and Aβ40 monomer as evaluated by molecular mechanics Poisson-Boltzmann surface area method. And, the residues of F4, Y10, V12, L17 and L34 in Aβ40 might contribute to the binding energy in HCP-Aβ40 complex. All these results elucidate the molecular mechanism underlying the inhibitory effects of HCP against the conformational transformation of Aβ40, providing a support that HCP may be developed as a potential anti-Aβ compound for the treatment of Aβ-related diseases.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Huitu Zhang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education; Tianjin Key Laboratory of Industrial Microbiology; College of Biotechnology, Tianjin University of Science & Technology, Tianjin, P. R. China
| | - Jingcheng Sang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education; Tianjin Key Laboratory of Industrial Microbiology; College of Biotechnology, Tianjin University of Science & Technology, Tianjin, P. R. China
| | - Li Li
- College of Marine and Environmental Sciences, Tianjin University of Science & Technology, Tianjin, P. R. China
| | - Luying Jiang
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education; Tianjin Key Laboratory of Industrial Microbiology; College of Biotechnology, Tianjin University of Science & Technology, Tianjin, P. R. China
| | - Fuping Lu
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education; Tianjin Key Laboratory of Industrial Microbiology; College of Biotechnology, Tianjin University of Science & Technology, Tianjin, P. R. China
| | - Shan He
- Li Dak Sum Yip Yio Chin Kenneth Li Marine Biopharmaceutical Research Center, Ningbo University, Ningbo, China
| | - Wei Cui
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Xiaoqing Zhang
- Ningbo Key Laboratory of Behavioral Neuroscience, Zhejiang Provincial Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Fufeng Liu
- State Key Laboratory of Food Nutrition and Safety, Key Laboratory of Industrial Fermentation Microbiology of Ministry of Education; Tianjin Key Laboratory of Industrial Microbiology; College of Biotechnology, Tianjin University of Science & Technology, Tianjin, P. R. China
| |
Collapse
|
39
|
Gabr M, Murugan NA. Discovery of biphenyl pyrazole scaffold for neurodegenerative diseases: A novel class of acetylcholinesterase-centered multitargeted ligands. Bioorg Med Chem Lett 2020; 30:127370. [PMID: 32738978 DOI: 10.1016/j.bmcl.2020.127370] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/19/2022]
Abstract
Multitargeted ligands have demonstrated remarkable efficiency as potential therapeutics for neurodegenerative diseases as they target multiple pathways involved in the progression of these diseases. Herein, we report first-in-class dual inhibitor of acetylcholinesterase (AChE) and tau aggregation as a novel class of multitargeted ligands for neurodegenerative diseases. The reported biphenyl pyrazole scaffold binds monomeric tau with submicromolar affinity and impedes the formation of tau oligomers at early stages. Additionally, the lead compound inhibited AChE activity with an IC50 value of 0.35 ± 0.02 μM. Remarkably, the neuroprotective effect of this lead in induced cytotoxicity model of SH-SY5Y neuroblastoma cells is superior to single-targeted AChE and tau-aggregation inhibitors. This scaffold would enable development of new generation of multitargeted ligands for neurodegenerative diseases that function through dual targeting of AChE and monomeric tau.
Collapse
Affiliation(s)
- Moustafa Gabr
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, United States.
| | - Natarajan Arul Murugan
- Department of Theoretical Chemistry and Biology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, S-106 91 Stockholm, Sweden
| |
Collapse
|
40
|
Rajasekhar K, Samanta S, Bagoband V, Murugan NA, Govindaraju T. Antioxidant Berberine-Derivative Inhibits Multifaceted Amyloid Toxicity. iScience 2020; 23:101005. [PMID: 32272441 PMCID: PMC7138924 DOI: 10.1016/j.isci.2020.101005] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/22/2020] [Accepted: 03/18/2020] [Indexed: 12/21/2022] Open
Abstract
Multiple lines of evidence indicate that amyloid beta (Aβ) peptide is responsible for the pathological devastation caused in Alzheimer's disease (AD). Aβ aggregation species predominantly contribute to multifaceted toxicity observed in neuronal cells including generation of reactive oxygen species (ROS), mitochondrial dysfunction, interfering with synaptic signaling, and activation of premature apoptosis. Herein, we report a natural product berberine-derived (Ber-D) multifunctional inhibitor to ameliorate in cellulo multifaceted toxicity of AD. The structural attributes of polyphenolic Ber-D have contributed to its efficient Cu chelation and arresting the redox cycle to prevent the generation of ROS and rescue biomacromolecules from oxidative damage. Ber-D inhibits metal-dependent and -independent Aβ aggregation, which is supported by in silico studies. Ber-D treatment averts mitochondrial dysfunction and corresponding neuronal toxicity contributing to premature apoptosis. These key multifunctional attributes make Ber-D a potential therapeutic candidate to ameliorate multifaceted Aβ toxicity in AD.
Collapse
Affiliation(s)
- Kolla Rajasekhar
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, 560064 Karnataka, India
| | - Sourav Samanta
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, 560064 Karnataka, India
| | - Vardhaman Bagoband
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, 560064 Karnataka, India
| | - N Arul Murugan
- Department of Theoretical Chemistry and Biology, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, S-106 91 Stockholm, Sweden
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru, 560064 Karnataka, India.
| |
Collapse
|
41
|
Wang W, Liu W, Xu S, Dong X, Sun Y. Design of Multifunctional Agent Based on Basified Serum Albumin for Efficient In Vivo β-Amyloid Inhibition and Imaging. ACS APPLIED BIO MATERIALS 2020; 3:3365-3377. [DOI: 10.1021/acsabm.0c00295] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Wenjuan Wang
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
| | - Wei Liu
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
| | - Shaoying Xu
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
| | - Xiaoyan Dong
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
| | - Yan Sun
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300354, China
| |
Collapse
|
42
|
Gao L, Zhou F, Wang KX, Zhou YZ, Du GH, Qin XM. Baicalein protects PC12 cells from Aβ 25-35-induced cytotoxicity via inhibition of apoptosis and metabolic disorders. Life Sci 2020; 248:117471. [PMID: 32112868 DOI: 10.1016/j.lfs.2020.117471] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 02/16/2020] [Accepted: 02/24/2020] [Indexed: 12/18/2022]
Abstract
AIMS This study aimed to explore the protective effects and possible mechanisms of baicalein on Aβ25-35-induced toxicity. MAIN METHODS Thioflavin-T (Th-T) dye was used to determine the effects of baicalein on Aβ25-35 aggregation in vitro. PC12 cells were stimulated with Aβ25-35, then the effects of baicalein on apoptosis, mitochondrial membrane potential (MMP), adenosine triphosphate (ATP), mitochondrial respiratory complex I, reactive oxygen species (ROS) and nitric oxide (NO) levels were determined. Moreover, LC-MS metabolomics approach was used to detect metabolic changes induced by baicalein in Aβ25-35-injured PC12 cells. KEY FINDINGS The results showed that baicalein could inhibit the aggregation of Aβ25-35 in vitro. Furthermore, pretreatment with baicalein significantly prevented Aβ25-35-induced cell apoptosis, as manifested by increasing the levels of MMP, ATP and mitochondrial respiratory complex I, decreasing the contents of ROS and NO. LC-MS metabolomics revealed that baicalein can regulate 5 metabolites, mainly involving two metabolic pathways, arginine and proline metabolism, nicotinate and nicotinamide metabolism. SIGNIFICANCE Our study revealed that baicalein has a protective effect on Aβ25-35-induced neurotoxicity in PC12 cells, which may be related to inhibition of apoptosis and metabolic disorders.
Collapse
Affiliation(s)
- Li Gao
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, PR China.
| | - Feng Zhou
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, PR China; College of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, PR China
| | - Ke-Xin Wang
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, PR China; College of Chemistry and Chemical Engineering, Shanxi University, Taiyuan 030006, PR China
| | - Yu-Zhi Zhou
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, PR China
| | - Guan-Hua Du
- Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, PR China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, PR China.
| |
Collapse
|
43
|
Ramesh M, Gopinath P, Govindaraju T. Role of Post-translational Modifications in Alzheimer's Disease. Chembiochem 2020; 21:1052-1079. [PMID: 31863723 DOI: 10.1002/cbic.201900573] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/19/2019] [Indexed: 12/22/2022]
Abstract
The global burden of Alzheimer's disease (AD) is growing. Valiant efforts to develop clinical candidates for treatment have continuously met with failure. Currently available palliative treatments are temporary and there is a constant need to search for reliable disease pathways, biomarkers and drug targets for developing diagnostic and therapeutic tools to address the unmet medical needs of AD. Challenges in drug-discovery efforts raise further questions about the strategies of current conventional diagnosis; drug design; and understanding of disease pathways, biomarkers and targets. In this context, post-translational modifications (PTMs) regulate protein trafficking, function and degradation, and their in-depth study plays a significant role in the identification of novel biomarkers and drug targets. Aberrant PTMs of disease-relevant proteins could trigger pathological pathways, leading to disease progression. Advancements in proteomics enable the generation of patterns or signatures of such modifications, and thus, provide a versatile platform to develop biomarkers based on PTMs. In addition, understanding and targeting the aberrant PTMs of various proteins provide viable avenues for addressing AD drug-discovery challenges. This review highlights numerous PTMs of proteins relevant to AD and provides an overview of their adverse effects on the protein structure, function and aggregation propensity that contribute to the disease pathology. A critical discussion offers suggestions of methods to develop PTM signatures and interfere with aberrant PTMs to develop viable diagnostic and therapeutic interventions in AD.
Collapse
Affiliation(s)
- Madhu Ramesh
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur P.O., Bengaluru, 560064, Karnataka, India
| | - Pushparathinam Gopinath
- Department of Chemistry, SRM-Institute of Science and Technology, Kattankulathur, 603203, Chennai, Tamilnadu, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR), Jakkur P.O., Bengaluru, 560064, Karnataka, India
| |
Collapse
|
44
|
Samanta S, Govindaraju T. Unambiguous Detection of Elevated Levels of Hypochlorous Acid in Double Transgenic AD Mouse Brain. ACS Chem Neurosci 2019; 10:4847-4853. [PMID: 31790189 DOI: 10.1021/acschemneuro.9b00554] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Alzheimer's disease (AD) is one of the most prevalent forms of dementia. The current diagnosis methods based on the behavior and cognitive decline or imaging of core biomarkers, namely, amyloid-β (Aβ) plaques and neurofibrillary tangles (NFTs), in the brain offer poor to moderate success. Detection and imaging of biomarkers that cause additional traits of pathophysiological aberrations in the brain are invaluable to monitor early disease onset and progression of AD pathology. The pathological hallmark of AD is associated with generation of excessive reactive oxygen species (ROS) in the brain, which aggravate oxidative stress and inflammation. ROS production involves elevated levels of hypochlorous acid (HOCl) and can serve as one of the potential biomarkers for the diagnosis of AD. We report the design, synthesis, and characterization of switchable coumarin-morpholine (CM) conjugates as off-on fluorescence probes for the specific detection of HOCl produced and proximally localized with amyloid plaques. The nonfluorescent thioamide probe CM2 undergoes regioselective transformation to fluorescent amide probe CM1 in the presence of HOCl (∼90-fold fluorescence enhancement and 0.32 quantum yield) with high selectivity and sensitivity (detection limit: 0.17 μM). The excellent cellular uptake and blood-brain barrier (BBB) crossing ability of CM2 allowed unambiguous and differential detection, imaging, and quantification of HOCl in cellular milieu and in the wild type (WT) and AD mouse brains. This study demonstrates the elevated level of HOCl in the AD mouse brain and the potential to expand the repertoire of biomarkers for the diagnosis of AD.
Collapse
Affiliation(s)
- Sourav Samanta
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru 560064, Karnataka, India
| | - Thimmaiah Govindaraju
- Bioorganic Chemistry Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur P.O., Bengaluru 560064, Karnataka, India
| |
Collapse
|