1
|
Kim TK, Hong JM, Kim J, Kim KH, Han SJ, Kim IC, Oh H, Jo DG, Yim JH. Therapeutic Potential of Ramalin Derivatives with Enhanced Stability in the Treatment of Alzheimer's Disease. Molecules 2024; 29:5223. [PMID: 39598614 PMCID: PMC11597085 DOI: 10.3390/molecules29225223] [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: 09/30/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Abstract
Alzheimer's disease (AD) remains a significant public health challenge with limited effective treatment options. Ramalin, a compound derived from Antarctic lichens, has shown potential in the treatment of AD because of its strong antioxidant and anti-inflammatory properties. However, its instability and toxicity have hindered the development of Ramalin as a viable therapeutic agent. The primary objective of this study was to synthesize and evaluate novel Ramalin derivatives with enhanced stabilities and reduced toxic profiles, with the aim of retaining or improving their therapeutic potential against AD. The antioxidant, anti-inflammatory, anti-BACE-1, and anti-tau activities of four synthesized Ramalin derivatives (i.e., RA-Hyd-Me, RA-Hyd-Me-Tol, RA-Sali, and RA-Benzo) were evaluated. These derivatives demonstrated significantly improved stabilities compared to the parent compound, with RA-Sali giving the most promising results. More specifically, RA-Sali exhibited a potent BACE-1 inhibitory activity and effectively reduced tau phosphorylation, a critical factor in AD pathology. Despite exhibiting reduced antioxidant activities compared to the parent compound, these derivatives represent a potential multi-targeted approach for AD treatment, marking a significant step forward in the development of stable and effective AD therapeutics.
Collapse
Affiliation(s)
- Tai Kyoung Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.-M.H.); (J.K.); (K.H.K.); (S.J.H.); (I.-C.K.)
| | - Ju-Mi Hong
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.-M.H.); (J.K.); (K.H.K.); (S.J.H.); (I.-C.K.)
| | - Jaewon Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.-M.H.); (J.K.); (K.H.K.); (S.J.H.); (I.-C.K.)
- Department of Plant Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Kyung Hee Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.-M.H.); (J.K.); (K.H.K.); (S.J.H.); (I.-C.K.)
- Department of Chemistry, Hanseo University, Seosan 31962, Republic of Korea
| | - Se Jong Han
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.-M.H.); (J.K.); (K.H.K.); (S.J.H.); (I.-C.K.)
| | - Il-Chan Kim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.-M.H.); (J.K.); (K.H.K.); (S.J.H.); (I.-C.K.)
| | - Hyuncheol Oh
- College of Pharmacy, Wonkwang University, Iksan 54538, Republic of Korea;
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Joung Han Yim
- Division of Polar Life Sciences, Korea Polar Research Institute, Incheon 21990, Republic of Korea; (T.K.K.); (J.-M.H.); (J.K.); (K.H.K.); (S.J.H.); (I.-C.K.)
| |
Collapse
|
2
|
Di Lorenzo D. Tau Protein and Tauopathies: Exploring Tau Protein-Protein and Microtubule Interactions, Cross-Interactions and Therapeutic Strategies. ChemMedChem 2024; 19:e202400180. [PMID: 39031682 DOI: 10.1002/cmdc.202400180] [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: 03/07/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/22/2024]
Abstract
Tau, a microtubule-associated protein (MAP), is essential to maintaining neuronal stability and function in the healthy brain. However, aberrant modifications and pathological aggregations of Tau are implicated in various neurodegenerative disorders, collectively known as tauopathies. The most common Tauopathy is Alzheimer's Disease (AD) counting nowadays more than 60 million patients worldwide. This comprehensive review delves into the multifaceted realm of Tau protein, puzzling out its intricate involvement in both physiological and pathological roles. Emphasis is put on Tau Protein-Protein Interactions (PPIs), depicting its interaction with tubulin, microtubules and its cross-interaction with other proteins such as Aβ1-42, α-synuclein, and the chaperone machinery. In the realm of therapeutic strategies, an overview of diverse possibilities is presented with their relative clinical progresses. The focus is mostly addressed to Tau protein aggregation inhibitors including recent small molecules, short peptides and peptidomimetics with specific focus on compounds that showed a double anti aggregative activity on both Tau protein and Aβ amyloid peptide. This review amalgamates current knowledge on Tau protein and evolving therapeutic strategies, providing a comprehensive resource for researchers seeking to deepen their understanding of the Tau protein and for scientists involved in the development of new peptide-based anti-aggregative Tau compounds.
Collapse
Affiliation(s)
- Davide Di Lorenzo
- Department of Chemistry, Organic and Bioorganic Chemistry, Bielefeld University, Universitätsstraße 25, D-33615, Bielefeld, Germany
| |
Collapse
|
3
|
James EI, Baggett DW, Chang E, Schachter J, Nixey T, Choi K, Guttman M, Nath A. Tryptanthrin Analogs Substoichiometrically Inhibit Seeded and Unseeded Tau4RD Aggregation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578649. [PMID: 38352474 PMCID: PMC10862865 DOI: 10.1101/2024.02.02.578649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Microtubule-associated protein tau is an intrinsically disordered protein (IDP) that forms characteristic fibrillar aggregates in several diseases, the most well-known of which is Alzheimer's disease (AD). Despite keen interest in disrupting or inhibiting tau aggregation to treat AD and related dementias, there are currently no FDA-approved tau-targeting drugs. This is due, in part, to the fact that tau and other IDPs do not exhibit a single well-defined conformation but instead populate a fluctuating conformational ensemble that precludes finding a stable "druggable" pocket. Despite this challenge, we previously reported the discovery of two novel families of tau ligands, including a class of aggregation inhibitors, identified through a protocol that combines molecular dynamics, structural analysis, and machine learning. Here we extend our exploration of tau druggability with the identification of tryptanthrin and its analogs as potent, substoichiometric aggregation inhibitors, with the best compounds showing potencies in the low nanomolar range even at a ~100-fold molar excess of tau4RD. Moreover, conservative changes in small molecule structure can have large impacts on inhibitory potency, demonstrating that similar structure-activity relationship (SAR) principles as used for traditional drug development also apply to tau and potentially to other IDPs.
Collapse
Affiliation(s)
- Ellie I. James
- Department of Medicinal Chemistry, University of Washington, Seattle, WA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA
| | - David W. Baggett
- Department of Medicinal Chemistry, University of Washington, Seattle, WA
- Current address: Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN
| | - Edcon Chang
- Takeda Development Center Americas, San Diego, CA
| | | | - Thomas Nixey
- Takeda Development Center Americas, San Diego, CA
| | | | - Miklos Guttman
- Department of Medicinal Chemistry, University of Washington, Seattle, WA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA
| | - Abhinav Nath
- Department of Medicinal Chemistry, University of Washington, Seattle, WA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA
| |
Collapse
|
4
|
Shi H, Zhao Y. Modulation of Tau Pathology in Alzheimer's Disease by Dietary Bioactive Compounds. Int J Mol Sci 2024; 25:831. [PMID: 38255905 PMCID: PMC10815728 DOI: 10.3390/ijms25020831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/02/2024] [Accepted: 01/02/2024] [Indexed: 01/24/2024] Open
Abstract
Tau is a microtubule-associated protein essential for microtubule assembly and stability in neurons. The abnormal intracellular accumulation of tau aggregates is a major characteristic of brains from patients with Alzheimer's disease (AD) and other tauopathies. In AD, the presence of neurofibrillary tangles (NFTs), which is composed of hyperphosphorylated tau protein, is positively correlated with the severity of the cognitive decline. Evidence suggests that the accumulation and aggregation of tau cause synaptic dysfunction and neuronal degeneration. Thus, the prevention of abnormal tau phosphorylation and elimination of tau aggregates have been proposed as therapeutic strategies for AD. However, currently tau-targeting therapies for AD and other tauopathies are limited. A number of dietary bioactive compounds have been found to modulate the posttranslational modifications of tau, including phosphorylation, small ubiquitin-like modifier (SUMO) mediated modification (SUMOylation) and acetylation, as well as inhibit tau aggregation and/or promote tau degradation. The advantages of using these dietary components over synthetic substances in AD prevention and intervention are their safety and accessibility. This review summarizes the mechanisms leading to tau pathology in AD and highlights the effects of bioactive compounds on the hyperphosphorylation, aggregation and clearance of tau protein. The potential of using these bioactive compounds for AD prevention and intervention is also discussed.
Collapse
Affiliation(s)
- Huahua Shi
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, China;
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Yan Zhao
- Department of Bioengineering, Harbin Institute of Technology, Weihai 264209, China;
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| |
Collapse
|
5
|
Dassamiour S, Bensaad MS, Ghebache W. Utility of phenolic acids in neurological disorders. ADVANCEMENT OF PHENOLIC ACIDS IN DRUG DISCOVERY 2024:295-344. [DOI: 10.1016/b978-0-443-18538-0.00015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
6
|
Kamboj S, Sharma P, Kamboj R, Kamboj S, Hariom, Girija, Guarve K, Dutt R, Verma I, Dua K, Rani N. Exploring the Therapeutic Potential of Phytoconstituents for Addressing Neurodegenerative Disorders. Cent Nerv Syst Agents Med Chem 2024; 24:129-144. [PMID: 38265386 DOI: 10.2174/0118715249273015231225091339] [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: 07/19/2023] [Revised: 10/03/2023] [Accepted: 11/20/2023] [Indexed: 01/25/2024]
Abstract
Neurodegenerative disorder is a serious condition that is caused by abnormal or no neurological function. Neurodegenerative disease is a major growing cause of mortality and morbidity worldwide, especially in the elderly. After World War Ⅱ, eugenics term was exterminated from medicines. Neurodegenerative disease is a genetically inherited disease. Lifestyle changes, environmental factors, and genetic modification, together or alone, are involved in the occurrence of this disorder. The major examples of neurodegenerative disorders are Alzheimer's and Parkinson's disease, in which apoptosis and necrosis are the two major death pathways for neurons. It has been determined from various studies that the etiology of the neurodegenerative disease involves the role of oxidative stress and anti-oxidant defence system, which are prime factors associated with the activation of signal transduction pathway that is responsible for the formation of synuclein in the brain and manifestation of toxic reactions in the form of functional abnormality, which ultimately leads to the dysfunction of neuronal pathway or cell. There has not been much success in the discovery of effective therapy to treat neurodegenerative diseases because the main cause of abnormal functioning or death of neurons is not well known. However, the use of natural products that are derived from plants has effective therapeutic potential against neurodegenerative disease. The natural compounds with medicinal properties to prevent neurological dysfunction are curcumin, wolfberry, ginseng, and Withania somnifera. The selection and use of natural compounds are based on their strong anti-inflammatory and anti-oxidant properties against neurodegenerative disease. Herbal products have active constituents that play an important role in the prevention of communication errors between neurons and neurotransmitters and their respective receptors in the brain, which influence their function. Considering this, natural products have great potential against neurodegenerative diseases. This article reviews the natural compounds used to treat neurodegenerative diseases and their mechanisms of action.
Collapse
Affiliation(s)
- Sweta Kamboj
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar, India
| | - Prerna Sharma
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar, India
| | - Rohit Kamboj
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar, India
| | - Shikha Kamboj
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar, India
| | - Hariom
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar, India
| | - Girija
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar, India
| | - Kumar Guarve
- Guru Gobind Singh College of Pharmacy, Yamuna Nagar, India
| | - Rohit Dutt
- Gandhi Memorial National College, Ambala, India
| | - Inderjeet Verma
- MM College of Pharmacy, MM (Deemed to be University), Mullana, Ambala, India
| | - Kamal Dua
- Discipline of Pharmacy Graduate of Technology, Sydney, Australia
| | - Nidhi Rani
- Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| |
Collapse
|
7
|
Arar S, Haque MA, Kayed R. Protein aggregation and neurodegenerative disease: Structural outlook for the novel therapeutics. Proteins 2023:10.1002/prot.26561. [PMID: 37530227 PMCID: PMC10834863 DOI: 10.1002/prot.26561] [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: 06/08/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 08/03/2023]
Abstract
Before the controversial approval of humanized monoclonal antibody lecanemab, which binds to the soluble amyloid-β protofibrils, all the treatments available earlier, for Alzheimer's disease (AD) were symptomatic. The researchers are still struggling to find a breakthrough in AD therapeutic medicine, which is partially attributable to lack in understanding of the structural information associated with the intrinsically disordered proteins and amyloids. One of the major challenges in this area of research is to understand the structural diversity of intrinsically disordered proteins under in vitro conditions. Therefore, in this review, we have summarized the in vitro applications of biophysical methods, which are aimed to shed some light on the heterogeneity, pathogenicity, structures and mechanisms of the intrinsically disordered protein aggregates associated with proteinopathies including AD. This review will also rationalize some of the strategies in modulating disease-relevant pathogenic protein entities by small molecules using structural biology approaches and biophysical characterization. We have also highlighted tools and techniques to simulate the in vivo conditions for native and cytotoxic tau/amyloids assemblies, urge new chemical approaches to replicate tau/amyloids assemblies similar to those in vivo conditions, in addition to designing novel potential drugs.
Collapse
Affiliation(s)
- Sharif Arar
- Mitchell Center for Neurodegenerative Diseases
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
- Department of Chemistry, School of Science, The University of Jordan, Amman 11942, Jordan
| | - Md Anzarul Haque
- Mitchell Center for Neurodegenerative Diseases
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases
- Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, 77555, USA
| |
Collapse
|
8
|
Khan S, Hassan MI, Shahid M, Islam A. Nature's Toolbox Against Tau Aggregation: An Updated Review of Current Research. Ageing Res Rev 2023; 87:101924. [PMID: 37004844 DOI: 10.1016/j.arr.2023.101924] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/21/2023] [Accepted: 03/30/2023] [Indexed: 04/03/2023]
Abstract
Tau aggregation is a hallmark of several neurodegenerative disorders, such as Alzheimer's disease (AD), frontotemporal dementia, and progressive supranuclear palsy. Hyperphosphorylated tau is believed to contribute to the degeneration of neurons and the development of these complex diseases. Therefore, one potential treatment for these illnesses is to prevent or counteract tau aggregation. In recent years, interest has been increasing in developing nature-derived tau aggregation inhibitors as a potential treatment for neurodegenerative disorders. Researchers have become increasingly interested in natural compounds with multifunctional features, such as flavonoids, alkaloids, resveratrol, and curcumin, since these molecules can interact simultaneously with the various targets of AD. Recent studies have demonstrated that several natural compounds can inhibit tau aggregation and promote the disassembly of pre-formed tau aggregates. Nature-derived tau aggregation inhibitors hold promise as a potential treatment for neurodegenerative disorders. However, it is important to note that more research is needed to fully understand the mechanisms by which these compounds exert their effects and their safety and efficacy in preclinical and clinical studies. Nature-derived inhibitors of tau aggregation are a promising new direction in the research of neurodegenerative complexities. This review focuses on the natural products that have proven to be a rich supply for inhibitors in tau aggregation and their uses in neurodegenerative complexities, including AD.
Collapse
|
9
|
Dubey T, Sonawane SK, Mannava MKC, Nangia AK, Chandrashekar M, Chinnathambi S. The inhibitory effect of Curcumin-Artemisinin co-amorphous on Tau aggregation and Tau phosphorylation. Colloids Surf B Biointerfaces 2023; 221:112970. [DOI: 10.1016/j.colsurfb.2022.112970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 11/08/2022]
|
10
|
Choudhir G, Sharma S, Hariprasad P. A combinatorial approach to screen structurally diverse acetylcholinesterase inhibitory plant secondary metabolites targeting Alzheimer's disease. J Biomol Struct Dyn 2022; 40:11705-11718. [PMID: 34351840 DOI: 10.1080/07391102.2021.1962408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) is a form of Dementia known to diminish the brain's function by perturbating its structural and functional components. Though cholinesterase inhibitors are widely used to treat AD, they are limited by numbers and side effects. Hence, present study aims to identify structurally diverse Acetylcholinesterase (AChE) inhibitory plant secondary metabolites (PSM) by employing high throughput screening and computational studies. AChE inhibitory activity was performed using 390 crude extracts from 63 plant parts belongs to 58 plants. The lowest IC50 value was recorded by acetone extract of Cyperus rotundus rhizome at 0.5 mg/ml, followed by methanol extract of Terminalia arjuna bark (0.95 mg/ml) and water extract Acacia catechu stem (0.95 mg/ml). A virtual library containing 487 PSM belongs to 18 plants found positive for AChE inhibition (IC50≤5 mg/ml) was prepared. Through ADMET analysis, 78 PSM fulfilling selected drug-likeness parameters were selected for further analysis. Molecular docking studies of selected PSM against AChE recorded a wide range of binding energy from -3.40 to -10.90 Kcal/mol. Further molecular dynamics simulation studies also recorded stabilized interactions of AChE-ligand complexes in the term of RMSD, RMSF, Rg, SASA, and hydrogen bond interaction. MMPBSA analysis revealed the binding energy of selected PSM ranging from -123.757 to -261.697 kJ/mol. Our study demonstrated the potential of 12 PSM (Sugiol, Margolone, 7-Hydroxy-3',4'-(Methylenedioxy) flavan, Beta-cyprone, Ethenone, Isomargolonone, Serpentine, Cryptolepine, Rotundone, Strictamin, Rotundenol and Nootkatone) as AChE inhibitors. Further in vitro and in vivo experimental evaluations with pure PSM could be beneficial for therapeutic uses.
Collapse
Affiliation(s)
- Gourav Choudhir
- Centre for Rural Development and Technology, Indian Institute of Technology Delhi, New Delhi, India
| | - Satyawati Sharma
- Centre for Rural Development and Technology, Indian Institute of Technology Delhi, New Delhi, India
| | - P Hariprasad
- Centre for Rural Development and Technology, Indian Institute of Technology Delhi, New Delhi, India
| |
Collapse
|
11
|
A study from structural insight to the antiamyloidogenic and antioxidant activities of flavonoids: scaffold for future therapeutics of Alzheimer’s disease. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02990-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
12
|
Rawat P, Sehar U, Bisht J, Selman A, Culberson J, Reddy PH. Phosphorylated Tau in Alzheimer's Disease and Other Tauopathies. Int J Mol Sci 2022; 23:12841. [PMID: 36361631 PMCID: PMC9654278 DOI: 10.3390/ijms232112841] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 07/29/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in elderly people. Amyloid beta (Aβ) deposits and neurofibrillary tangles are the major pathological features in an Alzheimer's brain. These proteins are highly expressed in nerve cells and found in most tissues. Tau primarily provides stabilization to microtubules in the part of axons and dendrites. However, tau in a pathological state becomes hyperphosphorylated, causing tau dysfunction and leading to synaptic impairment and degeneration of neurons. This article presents a summary of the role of tau, phosphorylated tau (p-tau) in AD, and other tauopathies. Tauopathies, including Pick's disease, frontotemporal dementia, corticobasal degeneration, Alzheimer's disease, argyrophilic grain disease, progressive supranuclear palsy, and Huntington's disease, are the result of misprocessing and accumulation of tau within the neuronal and glial cells. This article also focuses on current research on the post-translational modifications and genetics of tau, tau pathology, the role of tau in tauopathies and the development of new drugs targeting p-tau, and the therapeutics for treating and possibly preventing tauopathies.
Collapse
Affiliation(s)
- Priyanka Rawat
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Ujala Sehar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Jasbir Bisht
- Department of Pediatrics, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Ashley Selman
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - John Culberson
- Department of Family Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Nutritional Sciences Department, College Human Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
13
|
Bhat BA, Almilaibary A, Mir RA, Aljarallah BM, Mir WR, Ahmad F, Mir MA. Natural Therapeutics in Aid of Treating Alzheimer's Disease: A Green Gateway Toward Ending Quest for Treating Neurological Disorders. Front Neurosci 2022; 16:884345. [PMID: 35651632 PMCID: PMC9149276 DOI: 10.3389/fnins.2022.884345] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
The current scientific community is facing a daunting challenge to unravel reliable natural compounds with realistic potential to treat neurological disorders such as Alzheimer's disease (AD). The reported compounds/drugs mostly synthetic deemed the reliability and therapeutic potential largely due to their complexity and off-target issues. The natural products from nutraceutical compounds emerge as viable preventive therapeutics to fill the huge gap in treating neurological disorders. Considering that Alzheimer's disease is a multifactorial disease, natural compounds offer the advantage of a multitarget approach, tagging different molecular sites in the human brain, as compared with the single-target activity of most of the drugs so far used to treat Alzheimer's disease. A wide range of plant extracts and phytochemicals reported to possess the therapeutic potential to Alzheimer's disease includes curcumin, resveratrol, epigallocatechin-3-gallate, morin, delphinidins, quercetin, luteolin, oleocanthal, and other phytochemicals such as huperzine A, limonoids, and azaphilones. Reported targets of these natural compounds include inhibition of acetylcholinesterase, amyloid senile plaques, oxidation products, inflammatory pathways, specific brain receptors, etc. We tenaciously aimed to review the in-depth potential of natural products and their therapeutic applications against Alzheimer's disease, with a special focus on a diversity of medicinal plants and phytocompounds and their mechanism of action against Alzheimer's disease pathologies. We strongly believe that the medicinal plants and phytoconstituents alone or in combination with other compounds would be effective treatments against Alzheimer's disease with lesser side effects as compared to currently available treatments.
Collapse
Affiliation(s)
- Basharat Ahmad Bhat
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Abdullah Almilaibary
- Department of Family and Community Medicine, Faculty of Medicine, Albaha University Alaqiq, Alaqiq, Saudi Arabia
| | - Rakeeb Ahmad Mir
- Department of Biotechnology, Baba Ghulam Shah Badshah University, Rajouri, India
| | - Badr M. Aljarallah
- Department of Gastroenterology and Hepatology, Qassim University, Buraydah, Saudi Arabia
| | - Wajahat R. Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| | - Fuzail Ahmad
- College of Applied Medical Science, Majmaah University, Al Majma’ah, Saudi Arabia
| | - Manzoor Ahmad Mir
- Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar, India
| |
Collapse
|
14
|
Recent trends of natural based therapeutics for mitochondria targeting in Alzheimer’s disease. Mitochondrion 2022; 64:112-124. [DOI: 10.1016/j.mito.2022.03.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 12/13/2022]
|
15
|
Ogbodo JO, Agbo CP, Njoku UO, Ogugofor MO, Egba SI, Ihim SA, Echezona AC, Brendan KC, Upaganlawar AB, Upasani CD. Alzheimer's Disease: Pathogenesis and Therapeutic Interventions. Curr Aging Sci 2022; 15:2-25. [PMID: 33653258 DOI: 10.2174/1874609814666210302085232] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/04/2020] [Accepted: 11/20/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Alzheimer's Disease (AD) is the most common cause of dementia. Genetics, excessive exposure to environmental pollutants, as well as unhealthy lifestyle practices are often linked to the development of AD. No therapeutic approach has achieved complete success in treating AD; however, early detection and management with appropriate drugs are key to improving prognosis. INTERVENTIONS The pathogenesis of AD was extensively discussed in order to understand the reasons for the interventions suggested. The interventions reviewed include the use of different therapeutic agents and approaches, gene therapy, adherence to healthy dietary plans (Mediterranean diet, Okinawan diet and MIND diet), as well as the use of medicinal plants. The potential of nanotechnology as a multidisciplinary and interdisciplinary approach in the design of nano-formulations of AD drugs and the use of Superparamagnetic Iron Oxide Nanoparticles (SPIONs) as theranostic tools for early detection of Alzheimer's disease were also discussed.
Collapse
Affiliation(s)
- John O Ogbodo
- Department of Science Laboratory Technology, University of Nigeria, Nsukka, Nigeria
| | - Chinazom P Agbo
- Department of Pharmaceutics, University of Nigeria, Nsukka, Nigeria
| | - Ugochi O Njoku
- Department of Biochemistry, University of Nigeria, Nsukka, Nigeria
| | | | - Simeon I Egba
- Department of Biochemistry, Michael Okpara University of Agriculture, Umudike, Nigeria
| | - Stella A Ihim
- Department of Pharmacology and Toxicology, University of Nigeria, Nsukka, Nigeria
| | | | | | - Aman B Upaganlawar
- Department of Pharmacology, Sureshdada Shriman\'s College of Pharmacy, New Dehli, India
| | | |
Collapse
|
16
|
Saravanan K, Karthikeyan S, Sugarthi S, Stephen AD. Binding studies of known molecules with acetylcholinesterase and bovine serum albumin: A comparative view. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2021; 259:119856. [PMID: 33979725 DOI: 10.1016/j.saa.2021.119856] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 03/25/2021] [Accepted: 04/15/2021] [Indexed: 06/12/2023]
Abstract
The interactions between selected molecules (piperine, tacrine, curcumin and silibinin) and proteins (acetylcholinesterase and bovine serum albumin) were investigated by Fluorescence spectroscopy, molecular docking, molecular dynamics, free energy calculation and non-covalent interaction analysis. These binding characteristics are of huge interest for understanding pharmacokinetic mechanism of the target molecules. The steady-state emission spectrum results showed that presence of static quenching mode for piperine, tacrine, curcumin, silibinin molecules with BSA and AChE complexes separately and this excitation-emission matrix analysis suggest that formation of ground-state complex between piperine, tacrine, curcumin, silibinin drugs and both BSA, AChE protein molecules. And, the binding model from molecular docking analysis of both BSA and AChE with these molecules clearly displayed non-covalent interactions (hydrogen bonding and hydrophobic interactions) which played a significant role in the binding mechanism. Further, the protein-ligand complexes are subjected to molecular dynamics and binding free energy calculation to confirm the stability of the molecule in the active site of BSA and AChE. The NCI (non-covalent interaction) approach supports to visualize the iso-surface of the reduced density gradient of such interactions between protein and ligands.
Collapse
Affiliation(s)
- Kandasamy Saravanan
- Faculty of Chemistry, University of Warsaw, Ludwika Pasteura 1, Warsaw 02093, Poland; Department of Physics, Periyar University, Salem 636 011, India.
| | - Subramani Karthikeyan
- G. S. Gill Research Institute, Guru Nanak College (Autonomous), Chennai 600 042, India
| | - Srinivasan Sugarthi
- Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | | |
Collapse
|
17
|
Durairajan SSK, Selvarasu K, Bera MR, Rajaram K, Iyaswamy A, Li M. Alzheimer's Disease and other Tauopathies: Exploring Efficacy of Medicinal Plant-Derived Compounds in Alleviating Tau-Mediated Neurodegeneration. Curr Mol Pharmacol 2021; 15:361-379. [PMID: 34488602 DOI: 10.2174/1874467214666210906125318] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/12/2020] [Accepted: 01/27/2021] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD), a major form of dementia, has been reported to affect more than 50 million people worldwide. It is characterized by the presence of amyloid-β (Aβ) plaques and hyperphosphorylated Tau-associated neurofibrillary tangles in the brain. Apart from AD, microtubule (MT)-associated protein Tau is also involved in other neurodegenerative diseases called tauopathies, including Pick's disease, frontotemporal lobar degeneration, progressive supranuclear palsy, and corticobasal degeneration. The recently unsuccessful phase III clinical trials related to Aβ-targeted therapeutic drugs indicated that alternative targets, such as Tau, should be studied to discover more effective and safer drugs. Recent drug discovery approaches to reduce AD-related Tau pathologies are primarily based on blocking Tau aggregation, inhibiting Tau phosphorylation, compensating impaired Tau function with MT-stabilizing agents, and targeting the degradation pathways in neuronal cells to degrade Tau protein aggregates. Owing to several limitations of the currently-available Tau-directed drugs, further studies are required to generate further effective and safer Tau-based disease-modifying drugs. Here, we review the studies that focused on medicinal plant-derived compounds capable of modulating the Tau protein, which is significantly elevated and hyperphosphorylated in AD and other tauopathies. We mainly considered the studies that focused on Tau protein as a therapeutic target. We reviewed several pertinent papers retrieved from PubMed and ScienceDirect using relevant keywords, with a primary focus on the Tau-targeting compounds from medicinal plants. These compounds include indolines, phenolics, flavonoids, coumarins, alkaloids, and iridoids, which have been scientifically proven to be Tau-targeting candidates for the treatment of AD.
Collapse
Affiliation(s)
- Siva Sundara Kumar Durairajan
- Mycobiology and Neurodegenerative Disease Research Lab, Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur. India
| | - Karthikeyan Selvarasu
- Mycobiology and Neurodegenerative Disease Research Lab, Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur. India
| | - Minu Rani Bera
- Mycobiology and Neurodegenerative Disease Research Lab, Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur. India
| | - Kaushik Rajaram
- Mycobiology and Neurodegenerative Disease Research Lab, Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Tiruvarur. India
| | - Ashok Iyaswamy
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR. China
| | - Min Li
- Mr. & Mrs. Ko Chi-Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR. China
| |
Collapse
|
18
|
Pinzi L, Tinivella A, Rastelli G. Chemoinformatics Analyses of Tau Ligands Reveal Key Molecular Requirements for the Identification of Potential Drug Candidates against Tauopathies. Molecules 2021; 26:5039. [PMID: 34443629 PMCID: PMC8400687 DOI: 10.3390/molecules26165039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/08/2021] [Accepted: 08/16/2021] [Indexed: 11/16/2022] Open
Abstract
Tau is a highly soluble protein mainly localized at a cytoplasmic level in the neuronal cells, which plays a crucial role in the regulation of microtubule dynamic stability. Recent studies have demonstrated that several factors, such as hyperphosphorylation or alterations of Tau metabolism, may contribute to the pathological accumulation of protein aggregates, which can result in neuronal death and the onset of a number of neurological disorders called Tauopathies. At present, there are no available therapeutic remedies able to reduce Tau aggregation, nor are there any structural clues or guidelines for the rational identification of compounds preventing the accumulation of protein aggregates. To help identify the structural properties required for anti-Tau aggregation activity, we performed extensive chemoinformatics analyses on a dataset of Tau ligands reported in ChEMBL. The performed analyses allowed us to identify a set of molecular properties that are in common between known active ligands. Moreover, extensive analyses of the fragment composition of reported ligands led to the identification of chemical moieties and fragment combinations prevalent in the more active compounds. Interestingly, many of these fragments were arranged in recurring frameworks, some of which were clearly present in compounds currently under clinical investigation. This work represents the first in-depth chemoinformatics study of the molecular properties, constituting fragments and similarity profiles, of known Tau aggregation inhibitors. The datasets of compounds employed for the analyses, the identified molecular fragments and their combinations are made publicly available as supplementary material.
Collapse
Affiliation(s)
- Luca Pinzi
- Department of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi 103/287, 41125 Modena, Italy; (L.P.); (A.T.)
| | - Annachiara Tinivella
- Department of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi 103/287, 41125 Modena, Italy; (L.P.); (A.T.)
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giulio Rastelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Via G. Campi 103/287, 41125 Modena, Italy; (L.P.); (A.T.)
| |
Collapse
|
19
|
Gartman JA, Tambar UK. Synthetic Studies of the Rubellin Natural Products: Development of a Stereoselective Strategy and Total Synthesis of (+)-Rubellin C. J Org Chem 2021; 86:11237-11262. [PMID: 34288689 DOI: 10.1021/acs.joc.1c00920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This manuscript describes our studies of the class of natural products known as the rubellins, culminating in the total synthesis of (+)-rubellin C. These anthraquinone-based natural products contain a variety of stereochemical and architectural motifs, including a 6-5-6-fused ring system, 5 stereogenic centers, and a central quaternary center. Herein, we report our development of a strategy to target the stereochemically dense core and anthraquinone nucleus, including approaches such as a bifunctional allylboron and vinyl triflate reagent, an anthraquinone benzylic metalation strategy, and a late-stage anthraquinone introduction strategy. Our studies culminate in a successful route to highly functionalized anthraquinone-based natural product scaffolds and a stereoselective total synthesis of (+)-rubellin C. These strategies and outcomes will aid in synthetic planning toward anthraquinone-based natural products of high interest.
Collapse
Affiliation(s)
- Jackson A Gartman
- Department of Biochemistry, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9038, United States
| | - Uttam K Tambar
- Department of Biochemistry, UT Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9038, United States
| |
Collapse
|
20
|
Wang D, Huang X, Yan L, Zhou L, Yan C, Wu J, Su Z, Huang Y. The Structure Biology of Tau and Clue for Aggregation Inhibitor Design. Protein J 2021; 40:656-668. [PMID: 34401998 DOI: 10.1007/s10930-021-10017-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2021] [Indexed: 12/22/2022]
Abstract
Tau is a microtubule-associated protein that is mainly expressed in central and peripheral nerve systems. Tau binds to tubulin and regulates assembly and stabilization of microtubule, thus playing a critical role in neuron morphology, axon development and navigation. Tau is highly stable under normal conditions; however, there are several factors that can induce or promote aggregation of tau, forming neurofibrillary tangles. Neurofibrillary tangles are toxic to neurons, which may be related to a series of neurodegenerative diseases including Alzheimer's disease. Thus, tau is widely accepted as an important therapeutic target for neurodegenerative diseases. While the monomeric structure of tau is highly disordered, the aggregate structure of tau is formed by closed packing of β-stands. Studies on the structure of tau and the structural transition mechanism provide valuable information on the occurrence, development, and therapy of tauopathies. In this review, we summarize recent progress on the structural investigation of tau and based on which we discuss aggregation inhibitor design.
Collapse
Affiliation(s)
- Dan Wang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Xianlong Huang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Lu Yan
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Luoqi Zhou
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Chang Yan
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Jinhu Wu
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Zhengding Su
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China.,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China
| | - Yongqi Huang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, 430068, China. .,Hubei Key Laboratory of Industrial Microbiology, Department of Biological Engineering, Hubei University of Technology, Wuhan, 430068, Hubei, China.
| |
Collapse
|
21
|
Kim Y, Cho SH. Lindera glauca Blume ameliorates amyloid-β 1-42-induced memory impairment in mice with neuroprotection and activation of the CREB-BDNF pathway. Neurochem Int 2021; 147:105071. [PMID: 34000331 DOI: 10.1016/j.neuint.2021.105071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disorder presenting cognitive decline accompanied by deposits of amyloid-β (Aβ) and tau hyperphosphorylation. Without current treatment to AD, many studies suggested diverse approaches, one of which was herbal medicine and its active compounds. Very few studies have examined the effect of Lindera glauca Blume (L. glauca) in models of degenerative disease despite the attention that it received as a novel potential treatment source. We examined the efficacy of L. glauca in a mouse model of AD, which was induced by intrahippocampal injection of Aβ1-42. METHODS Mice were intrahippocampally infused with Aβ1-42 and were orally administered ethanolic extract of L.glauca before and after infusion for 21 days. Y-maze test and Morris water maze was conducted to assess memory impairment. Immunohistochemistry and western blot analysis were performed to assess the effect of L. glauca administration on pathological changes in mice. RESULTS L. glauca exhibited beneficial effects in spatial and reference learning as shown in increased time spent in the target quadrant in Morris water maze and increased spontaneous alternation in Y-maze. At the same time, decline of Aβ burden and phosphorylated tau were observed in the hippocampus of L. glauca-treated mouse under intrahippocampal injection of Aβ1-42. The results corresponded with amelioration of the decreased neuronal marker, neuronal-specific nuclear protein (NeuN) and attenuation of the increased reactive astrocyte marker, glial fibrillary acidic protein (GFAP) levels in hippocampus. Additionally, 21-day treatment with L. glauca inhibited downregulation of phosphorylated cAMP response element-binding protein (p-CREB) and brain-derived neurotrophic factor (BDNF) levels. CONCLUSION L. glauca improves behavioral deficits induced by Aβ1-42 and inhibits both Aβ- and tau-related pathological changes, stimulating neuroprotection mediated by CREB activation. L. glauca can be suggested as a new candidate for treatment of AD.
Collapse
Affiliation(s)
- Yunna Kim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea; Research Group of Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul, Republic of Korea; Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea.
| | - Seung-Hun Cho
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea; Research Group of Neuroscience, East-West Medical Research Institute, WHO Collaborating Center, Kyung Hee University, Seoul, Republic of Korea; Department of Clinical Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
22
|
Dubey T, Chinnathambi S. Photodynamic sensitizers modulate cytoskeleton structural dynamics in neuronal cells. Cytoskeleton (Hoboken) 2021; 78:232-248. [DOI: 10.1002/cm.21655] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 01/10/2023]
Affiliation(s)
- Tushar Dubey
- Neurobiology Group, Division of Biochemical Sciences CSIR‐National Chemical Laboratory Pune India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences CSIR‐National Chemical Laboratory Pune India
- Academy of Scientific and Innovative Research (AcSIR) Ghaziabad India
| |
Collapse
|
23
|
Zeng Y, Yang J, Zhang B, Gao M, Su Z, Huang Y. The structure and phase of tau: from monomer to amyloid filament. Cell Mol Life Sci 2021; 78:1873-1886. [PMID: 33078207 PMCID: PMC11073437 DOI: 10.1007/s00018-020-03681-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/20/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022]
Abstract
Tau is a microtubule-associated protein involved in regulation of assembly and spatial organization of microtubule in neurons. However, in pathological conditions, tau monomers assemble into amyloid filaments characterized by the cross-β structures in a number of neurodegenerative diseases known as tauopathies. In this review, we summarize recent progression on the characterization of structures of tau monomer and filament, as well as the dynamic liquid droplet assembly. Our aim is to reveal how post-translational modifications, amino acid mutations, and interacting molecules modulate the conformational ensemble of tau monomer, and how they accelerate or inhibit tau assembly into aggregates. Structure-based aggregation inhibitor design is also discussed in the context of dynamics and heterogeneity of tau structures.
Collapse
Affiliation(s)
- Yifan Zeng
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, China
- Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Jing Yang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, China
- Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Bailing Zhang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, China
- Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Meng Gao
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, China
- Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Zhengding Su
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, China
- Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China
| | - Yongqi Huang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan, China.
- Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, China.
| |
Collapse
|
24
|
Rai SN, Mishra D, Singh P, Vamanu E, Singh MP. Therapeutic applications of mushrooms and their biomolecules along with a glimpse of in silico approach in neurodegenerative diseases. Biomed Pharmacother 2021; 137:111377. [PMID: 33601145 DOI: 10.1016/j.biopha.2021.111377] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/29/2021] [Accepted: 02/08/2021] [Indexed: 12/19/2022] Open
Abstract
Neurodegenerative diseases (NDs) represent a common neurological pathology that determines a progressive deterioration of the brain or the nervous system. For treating NDs, comprehensive and alternative medicines have attracted scientific researchers' attention recently. Edible mushrooms are essential for preventing several age-based neuronal dysfunctions such as Parkinson's and Alzheimer's diseases. Mushroom such as Grifola frondosa, Lignosus rhinocerotis, Hericium erinaceus, may improve cognitive functions. It has also been reported that edible mushrooms (basidiocarps/mycelia extracts or isolated bioactive compounds) may reduce beta-amyloid-induced neurotoxicity. Medicinal mushrooms are being used for novel and natural compounds that help modulate immune responses and possess anti-cancer, anti-microbial, and anti-oxidant properties. Compounds such as polyphenols, terpenoids, alkaloids, sesquiterpenes, polysaccharides, and metal chelating agents are validated in different ND treatments. This review aims to assess mushrooms' role and their biomolecules utilization for treating different kinds of NDs. The action mechanisms, presented here, including reducing oxidative stress, neuroinflammation, and modulation of acetylcholinesterase activity, protecting neurons or stimulation, and regulating neurotrophins synthesis. We also provide background about neurodegenerative diseases and in-silico techniques of the drug research. High costs associated with experiments and current ethical law imply efficient alternatives with limited cost value. In silico approaches provide an alternative method with low cost that has been successfully implemented to cure ND disorders in recent days. We also describe the applications of computational procedures such as molecular docking, virtual high-throughput screening, molecular dynamic (MD) simulation, quantum-mechanical methods for drug design. They were reported against various targets in NDs.
Collapse
Affiliation(s)
- Sachchida Nand Rai
- Centre of Biotechnology, University of Allahabad, Prayagraj 211002, India.
| | - Divya Mishra
- Centre of Bioinformatics, University of Allahabad, Prayagraj 211002, India.
| | - Payal Singh
- Department of Zoology, MMV, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| | - Emanuel Vamanu
- Faculty of Biotechnology, University of Agronomic Science and Veterinary Medicine, 59 Marasti blvd, 1 district, 011464 Bucharest, Romania.
| | - M P Singh
- Centre of Biotechnology, University of Allahabad, Prayagraj 211002, India.
| |
Collapse
|
25
|
Li W, Qiu J, Li XL, Aday S, Zhang J, Conley G, Xu J, Joseph J, Lan H, Langer R, Mannix R, Karp JM, Joshi N. BBB pathophysiology-independent delivery of siRNA in traumatic brain injury. SCIENCE ADVANCES 2021; 7:7/1/eabd6889. [PMID: 33523853 DOI: 10.1101/2020.06.26.173393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/03/2020] [Indexed: 05/23/2023]
Abstract
Small interfering RNA (siRNA)-based therapeutics can mitigate the long-term sequelae of traumatic brain injury (TBI) but suffer from poor permeability across the blood-brain barrier (BBB). One approach to overcoming this challenge involves treatment administration while BBB is transiently breached after injury. However, it offers a limited window for therapeutic intervention and is applicable to only a subset of injuries with substantially breached BBB. We report a nanoparticle platform for BBB pathophysiology-independent delivery of siRNA in TBI. We achieved this by combined modulation of surface chemistry and coating density on nanoparticles, which maximized their active transport across BBB. Engineered nanoparticles injected within or outside the window of breached BBB in TBI mice showed threefold higher brain accumulation compared to nonengineered PEGylated nanoparticles and 50% gene silencing. Together, our data suggest that this nanoparticle platform is a promising next-generation drug delivery approach for the treatment of TBI.
Collapse
Affiliation(s)
- Wen Li
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Jianhua Qiu
- Harvard Medical School, Boston, MA 02115, USA
- Division of Emergency Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Xiang-Ling Li
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Sezin Aday
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jingdong Zhang
- Division of Emergency Medicine, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Grace Conley
- Division of Emergency Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jun Xu
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - John Joseph
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Haoyue Lan
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Robert Langer
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Rebekah Mannix
- Harvard Medical School, Boston, MA 02115, USA.
- Division of Emergency Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jeffrey M Karp
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
- Harvard Medical School, Boston, MA 02115, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute, Cambridge, MA 02142, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Nitin Joshi
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
26
|
Li W, Qiu J, Li XL, Aday S, Zhang J, Conley G, Xu J, Joseph J, Lan H, Langer R, Mannix R, Karp JM, Joshi N. BBB pathophysiology-independent delivery of siRNA in traumatic brain injury. SCIENCE ADVANCES 2021; 7:eabd6889. [PMID: 33523853 PMCID: PMC7775748 DOI: 10.1126/sciadv.abd6889] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 11/03/2020] [Indexed: 05/02/2023]
Abstract
Small interfering RNA (siRNA)-based therapeutics can mitigate the long-term sequelae of traumatic brain injury (TBI) but suffer from poor permeability across the blood-brain barrier (BBB). One approach to overcoming this challenge involves treatment administration while BBB is transiently breached after injury. However, it offers a limited window for therapeutic intervention and is applicable to only a subset of injuries with substantially breached BBB. We report a nanoparticle platform for BBB pathophysiology-independent delivery of siRNA in TBI. We achieved this by combined modulation of surface chemistry and coating density on nanoparticles, which maximized their active transport across BBB. Engineered nanoparticles injected within or outside the window of breached BBB in TBI mice showed threefold higher brain accumulation compared to nonengineered PEGylated nanoparticles and 50% gene silencing. Together, our data suggest that this nanoparticle platform is a promising next-generation drug delivery approach for the treatment of TBI.
Collapse
Affiliation(s)
- Wen Li
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Jianhua Qiu
- Harvard Medical School, Boston, MA 02115, USA
- Division of Emergency Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Xiang-Ling Li
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Sezin Aday
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jingdong Zhang
- Division of Emergency Medicine, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Grace Conley
- Division of Emergency Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jun Xu
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - John Joseph
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Haoyue Lan
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Robert Langer
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Rebekah Mannix
- Harvard Medical School, Boston, MA 02115, USA.
- Division of Emergency Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Jeffrey M Karp
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
- Harvard Medical School, Boston, MA 02115, USA
- Harvard-Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Broad Institute, Cambridge, MA 02142, USA
- Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Nitin Joshi
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
- Department of Chemical Engineering and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
27
|
Abstract
The rubellins are a family of stereochemically complex anthraquinoid heterodimers containing an unprecedented chemical scaffold. Although the rubellins have been known for over three decades, no total synthesis has been achieved since their discovery. Their topology is characterized by a 6-5-6 fused ring system, five neighboring stereocenters including a quaternary center all in a convoluted core, and an anthraquinone nucleus. The rubellin architecture has been shown to inhibit and reverse the aggregation of tau protein, a therapeutically relevant target for Alzheimer's disease. Herein, we describe the first stereoselective synthesis of a member of the family, (+)-rubellin C, in 16 steps. Strategic disconnections allow expedient construction of stereochemical and topological intricacy in a short sequence of borylative and transition metal-catalyzed steps.
Collapse
Affiliation(s)
- Jackson A Gartman
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9038, United States
| | - Uttam K Tambar
- Department of Biochemistry, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9038, United States
| |
Collapse
|
28
|
Dong K, Fernando WMADB, Durham R, Stockmann R, W. Jayatunga DP, Jayasena V. A role of sea buckthorn on Alzheimer’s disease. Int J Food Sci Technol 2020. [DOI: 10.1111/ijfs.14571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Ke Dong
- School of Science and Health Western Sydney University Locked Bag 1797 Penrith NSW 2751 Australia
| | - Warnakulasuriya M. A. D. Binosha Fernando
- Centre of Excellence for Alzheimer's Disease Research and Care School of Medical and Health Sciences Edith Cowan University Joondalup WA 6027 Australia
- Australian Alzheimer’s Research Foundation Ralph and Patricia Sarich Neuroscience Research Institute 8 Verdun Street Nedlands WA 6009 Australia
| | - Rosalie Durham
- School of Science and Health Western Sydney University Locked Bag 1797 Penrith NSW 2751 Australia
| | - Regine Stockmann
- CSIRO Agriculture and Food 671 Sneydes Rd. Werribee Vic. 3030 Australia
| | - Dona Pamoda W. Jayatunga
- Centre of Excellence for Alzheimer's Disease Research and Care School of Medical and Health Sciences Edith Cowan University Joondalup WA 6027 Australia
- Australian Alzheimer’s Research Foundation Ralph and Patricia Sarich Neuroscience Research Institute 8 Verdun Street Nedlands WA 6009 Australia
| | - Vijay Jayasena
- School of Science and Health Western Sydney University Locked Bag 1797 Penrith NSW 2751 Australia
| |
Collapse
|
29
|
Lo Cascio F, Garcia S, Montalbano M, Puangmalai N, McAllen S, Pace A, Palumbo Piccionello A, Kayed R. Modulating disease-relevant tau oligomeric strains by small molecules. J Biol Chem 2020; 295:14807-14825. [PMID: 32737202 PMCID: PMC7606668 DOI: 10.1074/jbc.ra120.014630] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
The pathological aggregation of tau plays an important role in Alzheimer's disease and many other related neurodegenerative diseases, collectively referred to as tauopathies. Recent evidence has demonstrated that tau oligomers, small and soluble prefibrillar aggregates, are highly toxic due to their strong ability to seed tau misfolding and propagate the pathology seen across different neurodegenerative diseases. We previously showed that novel curcumin derivatives affect preformed tau oligomer aggregation pathways by promoting the formation of more aggregated and nontoxic tau aggregates. To further investigate their therapeutic potential, we have extended our studies o disease-relevant brain-derived tau oligomers (BDTOs). Herein, using well-characterized BDTOs, isolated from brain tissues of different tauopathies, including Alzheimer's disease, progressive supranuclear palsy, and dementia with Lewy bodies, we found that curcumin derivatives modulate the aggregation state of BDTOs by reshaping them and rescue neurons from BDTO-associated toxicity. Interestingly, compound CL3 showed an effect on the aggregation pattern of BDTOs from different tauopathies, resulting in the formation of less neurotoxic larger tau aggregates with decreased hydrophobicity and seeding propensity. Our results lay the groundwork for potential investigations of the efficacy and beneficial effects of CL3 and other promising compounds for the treatment of tauopathies. Furthermore, CL3 may aid in the development of tau imaging agent for the detection of tau oligomeric strains and differential diagnosis of the tauopathies, thus enabling earlier interventions.
Collapse
Affiliation(s)
- Filippa Lo Cascio
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Stephanie Garcia
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Mauro Montalbano
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Nicha Puangmalai
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Salome McAllen
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Andrea Pace
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies-STEBICEF, University of Palermo, Palermo, Italy
| | - Antonio Palumbo Piccionello
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies-STEBICEF, University of Palermo, Palermo, Italy
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, Texas, USA.
| |
Collapse
|
30
|
Sonawane SK, Chidambaram H, Boral D, Gorantla NV, Balmik AA, Dangi A, Ramasamy S, Marelli UK, Chinnathambi S. EGCG impedes human Tau aggregation and interacts with Tau. Sci Rep 2020; 10:12579. [PMID: 32724104 PMCID: PMC7387440 DOI: 10.1038/s41598-020-69429-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 06/02/2020] [Indexed: 12/13/2022] Open
Abstract
Tau aggregation and accumulation is a key event in the pathogenesis of Alzheimer’s disease. Inhibition of Tau aggregation is therefore a potential therapeutic strategy to ameliorate the disease. Phytochemicals are being highlighted as potential aggregation inhibitors. Epigallocatechin-3-gallate (EGCG) is an active phytochemical of green tea that has shown its potency against various diseases including aggregation inhibition of repeat Tau. The potency of EGCG in altering the PHF assembly of full-length human Tau has not been fully explored. By various biophysical and biochemical analyses like ThS fluorescence assay, MALDI-TOF analysis and Isothermal Titration Calorimetry, we demonstrate dual effect of EGCG on aggregation inhibition and disassembly of full-length Tau and their binding affinity. The IC50 for Tau aggregation by EGCG was found to be 64.2 μM.
Collapse
Affiliation(s)
- Shweta Kishor Sonawane
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, 411008, India
| | - Hariharakrishnan Chidambaram
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, 411008, India
| | - Debjyoti Boral
- Structural Biology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, 411008, India
| | - Nalini Vijay Gorantla
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, 411008, India
| | - Abhishek Ankur Balmik
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, 411008, India
| | - Abha Dangi
- Central NMR Facility and Division of Organic Chemistry, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, 411008, India
| | - Sureshkumar Ramasamy
- Structural Biology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, 411008, India
| | - Udaya Kiran Marelli
- Central NMR Facility and Division of Organic Chemistry, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, 411008, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune, 411008, India. .,Academy of Scientific and Innovative Research (AcSIR), Pune, 411008, India.
| |
Collapse
|
31
|
Dubey T, Gorantla NV, Chandrashekara KT, Chinnathambi S. Photodynamic exposure of Rose-Bengal inhibits Tau aggregation and modulates cytoskeletal network in neuronal cells. Sci Rep 2020; 10:12380. [PMID: 32704015 PMCID: PMC7378248 DOI: 10.1038/s41598-020-69403-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/29/2020] [Indexed: 01/24/2023] Open
Abstract
The intracellular Tau aggregates are known to be associated with Alzheimer’s disease. The inhibition of Tau aggregation is an important strategy for screening of therapeutic molecules in Alzheimer's disease. Several classes of dyes possess a unique property of photo-excitation, which is applied as a therapeutic measure against numerous neurological dysfunctions. Rose Bengal is a Xanthene dye, which has been widely used as a photosensitizer in photodynamic therapy. The aim of this work was to study the protective role of Rose Bengal against Tau aggregation and cytoskeleton modulations. The aggregation inhibition and disaggregation potency of Rose Bengal and photo-excited Rose Bengal were observed by in-vitro fluorescence, circular dichroism, and electron microscopy. Rose Bengal and photo-excited Rose Bengal induce minimal cytotoxicity in neuronal cells. In our studies, we observed that Rose Bengal and photo-excited Rose Bengal modulate the cytoskeleton network of actin and tubulin. The immunofluorescence studies showed the increased filopodia structures after photo-excited Rose Bengal treatment. Furthermore, Rose Bengal treatment increases the connections between the cells. Rose Bengal and photo-excited Rose Bengal treatment-induced actin-rich podosome-like structures associated with cell membranes. The in-vivo studies on UAS E-14 Tau mutant Drosophila suggested that exposure to Rose Bengal and photo-excited Rose Bengal efficiency rescues the behavioural and memory deficit in flies. Thus, the overall results suggest that Rose Bengal could have a therapeutic potency against Tau aggregation.
Collapse
Affiliation(s)
- Tushar Dubey
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India
| | - Nalini Vijay Gorantla
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India
| | | | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory (CSIR-NCL), Dr. Homi Bhabha Road, Pune, 411008, India. .,Academy of Scientific and Innovative Research (AcSIR), New Delhi, 110025, India.
| |
Collapse
|
32
|
Viswanathan GK, Shwartz D, Losev Y, Arad E, Shemesh C, Pichinuk E, Engel H, Raveh A, Jelinek R, Cooper I, Gosselet F, Gazit E, Segal D. Purpurin modulates Tau-derived VQIVYK fibrillization and ameliorates Alzheimer's disease-like symptoms in animal model. Cell Mol Life Sci 2020; 77:2795-2813. [PMID: 31562564 PMCID: PMC11104911 DOI: 10.1007/s00018-019-03312-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 08/11/2019] [Accepted: 09/19/2019] [Indexed: 01/20/2023]
Abstract
Neurofibrillary tangles of the Tau protein and plaques of the amyloid β peptide are hallmarks of Alzheimer's disease (AD), which is characterized by the conversion of monomeric proteins/peptides into misfolded β-sheet rich fibrils. Halting the fibrillation process and disrupting the existing aggregates are key challenges for AD drug development. Previously, we performed in vitro high-throughput screening for the identification of potent inhibitors of Tau aggregation using a proxy model, a highly aggregation-prone hexapeptide fragment 306VQIVYK311 (termed PHF6) derived from Tau. Here we have characterized a hit molecule from that screen as a modulator of Tau aggregation using in vitro, in silico, and in vivo techniques. This molecule, an anthraquinone derivative named Purpurin, inhibited ~ 50% of PHF6 fibrillization in vitro at equimolar concentration and disassembled pre-formed PHF6 fibrils. In silico studies showed that Purpurin interacted with key residues of PHF6, which are responsible for maintaining its β-sheets conformation. Isothermal titration calorimetry and surface plasmon resonance experiments with PHF6 and full-length Tau (FL-Tau), respectively, indicated that Purpurin interacted with PHF6 predominantly via hydrophobic contacts and displayed a dose-dependent complexation with FL-Tau. Purpurin was non-toxic when fed to Drosophila and it significantly ameliorated the AD-related neurotoxic symptoms of transgenic flies expressing WT-FL human Tau (hTau) plausibly by inhibiting Tau accumulation and reducing Tau phosphorylation. Purpurin also reduced hTau accumulation in cell culture overexpressing hTau. Importantly, Purpurin efficiently crossed an in vitro human blood-brain barrier model. Our findings suggest that Purpurin could be a potential lead molecule for AD therapeutics.
Collapse
Affiliation(s)
- Guru Krishnakumar Viswanathan
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel-Aviv University, 69978, Tel Aviv, Israel
| | - Dana Shwartz
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel-Aviv University, 69978, Tel Aviv, Israel
| | - Yelena Losev
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel-Aviv University, 69978, Tel Aviv, Israel
| | - Elad Arad
- Ilse Katz Institute (IKI) for Nanoscale Science and Technology, Ben Gurion University of the Negev, 8410501, Beer Sheva, Israel
- Department of Chemistry, Ben Gurion University of the Negev, 8410501, Beer Sheva, Israel
| | - Chen Shemesh
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, 52621, Ramat Gan, Israel
| | - Edward Pichinuk
- Blavatnik Center for Drug Discovery, Tel-Aviv University, 69978, Tel Aviv, Israel
| | - Hamutal Engel
- Blavatnik Center for Drug Discovery, Tel-Aviv University, 69978, Tel Aviv, Israel
| | - Avi Raveh
- Blavatnik Center for Drug Discovery, Tel-Aviv University, 69978, Tel Aviv, Israel
| | - Raz Jelinek
- Ilse Katz Institute (IKI) for Nanoscale Science and Technology, Ben Gurion University of the Negev, 8410501, Beer Sheva, Israel
- Department of Chemistry, Ben Gurion University of the Negev, 8410501, Beer Sheva, Israel
| | - Itzik Cooper
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, 52621, Ramat Gan, Israel
- Interdisciplinary Center Herzliya, Herzliya, Israel
| | - Fabien Gosselet
- Blood-Brain Barrier Laboratory (LBHE), Université d'Artois, Lens, France
| | - Ehud Gazit
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel-Aviv University, 69978, Tel Aviv, Israel
- Blavatnik Center for Drug Discovery, Tel-Aviv University, 69978, Tel Aviv, Israel
| | - Daniel Segal
- Department of Molecular Microbiology and Biotechnology, School of Molecular Cell Biology and Biotechnology, Tel-Aviv University, 69978, Tel Aviv, Israel.
- The Interdisciplinary Sagol School of Neurosciences, Tel-Aviv University, 69978, Tel Aviv, Israel.
| |
Collapse
|
33
|
Sayad-Fathi S, Zaminy A, Babaei P, Yousefbeyk F, Azizi N, Nasiri E. The methanolic extract of Cinnamomum zeylanicum bark improves formaldehyde-induced neurotoxicity through reduction of phospho-tau (Thr231), inflammation, and apoptosis. EXCLI JOURNAL 2020; 19:671-686. [PMID: 32536837 PMCID: PMC7290100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/20/2020] [Indexed: 11/08/2022]
Abstract
Accumulation of formaldehyde (FA) in the brain is linked to age-related neurodegenerative disorders, as it accelerates memory impairment through tau protein aggregation, inflammation, and nuclear damage. This study aimed to assess the possible effects of methanolic cinnamon extract (CE) on FA-induced neurotoxicity in rats. The animals were treated with CE (100, 200, and 400 mg/kg, P.O.) for 30 days following FA administration (60 mg/kg, I.P.) for 30 days. Briefly, spatial and inhibitory memory were examined by Morris water maze (MWM) and passive avoidance (PA) tasks, respectively. The Nissl, Hoechst, and Bielschowsky silver staining methods were also used to assess apoptosis and neurofibrillary tangles (NFTs) in the hippocampal CA1 region, respectively. Brain tissues were probed with an anti-phospho-tau (Thr231) monoclonal antibody to assess tau hyperphosphorylation. Inflammatory cytokines (IL-1β, IL-6, and TNF-α) were also measured by ELISA assay. Western blotting was performed to quantify the amount of phospho-tau (Thr231), caspase-8, and caspase-9. The results showed that FA injection significantly caused tau hyperphosphorylation at Thr231 residue, which in turn disturbed the MWM performance. The ratio of apoptotic to intact neurons increased following FA treatment. The results of Western blotting indicated that the hippocampal levels of phospho-tau (Thr231) and caspase-8 were significantly higher in the FA group compared to the control group. The hippocampal levels of IL-1β, IL-6, and TNF-α in the FA group were also higher than the control group. Administration of 200 mg/kg of CE significantly improved the rats' MWM performance, decreased the levels of phospho-tau (Thr231), caspase-8, IL-6, and TNF-α, and reduced the ratio of apoptotic to intact neurons. Overall, cinnamon improved cognitive performance in FA-treated rats by eliminating tau hyperphosphorylation, inflammatory cytokines, and nuclear damage.
Collapse
Affiliation(s)
- Sara Sayad-Fathi
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran,Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Arash Zaminy
- Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Parvin Babaei
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran,Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran,Department of Physiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Fatemeh Yousefbeyk
- Department of Pharmacognosy, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Nasibeh Azizi
- Department of Medicinal Chemistry, School of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Ebrahim Nasiri
- Cellular and Molecular Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran,Neuroscience Research Center, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran,*To whom correspondence should be addressed: Ebrahim Nasiri, Cellular and Molecular Research Center, School of Medicine, Guilan University Campus, P.O. Box: 3363, Rasht, Iran; Cell: +989113357783, Tel: +98 1333690884, Fax: +98 1333690036, E-mail:
| |
Collapse
|
34
|
Son NT. A Mini-review of the Tropical Plant Cratoxylum fomosum ssp. pruniflorum: Phytochemical and Pharmacological Aspects. LETT ORG CHEM 2020. [DOI: 10.2174/1570178616666190902111630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The tropical plant C. formosum ssp. pruniflorum belongs to family Clusiaceae, which is
native to Southeast Asia countries. Phytochemical investigations on this plant showed interesting secondary
metabolites, comprising the main classes of xanthones, anthraquinones, flavonoids, phenolics,
and triterpenoids. Biological assessments established the wide spectrum of properties, either the extracts
or isolated compounds have been becoming valuable resources, constituents from C. formosum
ssp. pruniflorum were used for anti-bacteria, anti-inflammation, anti-cancer, or neuroprotective and
vascular protective activities. The long history of traditional application has confirmed the prospect in
use, this herbal plant was consumed as a combination tea or to treat skin wound healing, fever, cough,
ulcer, diarrhea, internal bleeding, stomachic and diuretic effects, and food poisoning.
Collapse
Affiliation(s)
- Ninh The Son
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Caugiay, Hanoi, Vietnam
| |
Collapse
|
35
|
Gorantla NV, Balaraman E, Chinnathambi S. Cobalt-based metal complexes prevent Repeat Tau aggregation and nontoxic to neuronal cells. Int J Biol Macromol 2020; 152:171-179. [PMID: 32105696 DOI: 10.1016/j.ijbiomac.2020.02.278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/14/2020] [Accepted: 02/24/2020] [Indexed: 01/11/2023]
Abstract
Alzheimer's disease (AD) is a fatal neurodegenerative disorder with an alarming increase in the death rate every year. AD is characterised by an aberrant accumulation of proteins in the form of aggregates. The axonal microtubule-associated protein Tau and amyloid-β undergo structural transition to β-sheet rich structure and form aggregates in neuronal soma as well as in the extracellular region. The loss of Tau from microtubules leads to the disintegration of axon and causing neuronal degeneration. This led to the development of effective drugs against AD, to prevent Tau aggregation. Here, we synthesized and screen metal-based complexes to prevent Tau protein aggregation. ThS fluorescence and TEM suggested the role of synthetic cobalt complexes in inhibiting Tau aggregation. CD spectroscopy showed that these complexes prevented conformational changes in Tau to β-sheet. CBMCs were not toxic at lower concentrations and formed non-toxic Tau species. L1 and L2 prevented membrane leakage; whereas, higher concentrations of L3 caused membrane leakage as observed by LDH release assay. The overall results indicate the synthetic cobalt complexes to be a promising molecule against AD.
Collapse
Affiliation(s)
- Nalini V Gorantla
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India; Academy of Scientific and Innovative Research (AcSIR), 411008 Pune, India.
| | - Ekambaram Balaraman
- Catalysis Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India; Academy of Scientific and Innovative Research (AcSIR), 411008 Pune, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India; Academy of Scientific and Innovative Research (AcSIR), 411008 Pune, India.
| |
Collapse
|
36
|
Lo Cascio F, Puangmalai N, Ellsworth A, Bucchieri F, Pace A, Palumbo Piccionello A, Kayed R. Toxic Tau Oligomers Modulated by Novel Curcumin Derivatives. Sci Rep 2019; 9:19011. [PMID: 31831807 PMCID: PMC6908736 DOI: 10.1038/s41598-019-55419-w] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/22/2019] [Indexed: 01/05/2023] Open
Abstract
The pathological aggregation and accumulation of tau, a microtubule-associated protein, is a common feature amongst more than 18 different neurodegenerative diseases that are collectively known as tauopathies. Recently, it has been demonstrated that the soluble and hydrophobic tau oligomers are highly toxic in vitro due to their capacity towards seeding tau misfolding, thereby propagating the tau pathology seen across different neurodegenerative diseases. Modulating the aggregation state of tau oligomers through the use of small molecules could be a useful therapeutic strategy to target their toxicity, regardless of other factors involved in their formation. In this study, we screened and tested a small library of newly synthesized curcumin derivatives against preformed recombinant tau oligomers. Our results show that the curcumin derivatives affect and modulate the tau oligomer aggregation pathways, converting to a more aggregated non-toxic state as assessed in the human neuroblastoma SH-SY5Y cell line and primary cortical neuron cultures. These results provide insight into tau aggregation and may become a basis for the discovery of new therapeutic agents, as well as advance the diagnostic field for the detection of toxic tau oligomers.
Collapse
Affiliation(s)
- Filippa Lo Cascio
- 0000 0001 1547 9964grid.176731.5Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555 USA ,0000 0001 1547 9964grid.176731.5Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Nicha Puangmalai
- 0000 0001 1547 9964grid.176731.5Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555 USA ,0000 0001 1547 9964grid.176731.5Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Anna Ellsworth
- 0000 0001 1547 9964grid.176731.5Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX 77555 USA ,0000 0001 1547 9964grid.176731.5Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX 77555 USA
| | - Fabio Bucchieri
- 0000 0004 1762 5517grid.10776.37Department of Biomedicine, Neurosciences and Advanced Diagnostic (BiND), University of Palermo, Palermo, 90127 Italy
| | - Andrea Pace
- 0000 0004 1762 5517grid.10776.37Department of Biological, Chemical and Pharmaceutical Sciences and Technologies - STEBICEF, University of Palermo, Palermo, 90128 Italy
| | - Antonio Palumbo Piccionello
- 0000 0004 1762 5517grid.10776.37Department of Biological, Chemical and Pharmaceutical Sciences and Technologies - STEBICEF, University of Palermo, Palermo, 90128 Italy
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA. .,Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
37
|
Dubey T, Gorantla NV, Chandrashekara KT, Chinnathambi S. Photoexcited Toluidine Blue Inhibits Tau Aggregation in Alzheimer's Disease. ACS OMEGA 2019; 4:18793-18802. [PMID: 31737841 PMCID: PMC6854831 DOI: 10.1021/acsomega.9b02792] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 09/19/2019] [Indexed: 05/12/2023]
Abstract
The aggregates of microtubule-associated protein Tau are considered as a major hallmark of Alzheimer's disease. Tau aggregates accumulate intracellularly leading to neuronal toxicity. Numerous approaches have been targeted against Tau protein aggregation, which include application of synthetic and natural compounds. Toluidine blue is a basic dye of phenothiazine family, which on irradiation with a 630 nm light gets converted into a photoexcited form, leading to generation of singlet oxygen species. Methylene blue is the parent compound of toluidine blue, which has been reported to be potent against tauopathy. In the present work, we studied the potency of toluidine blue and photoexcited toluidine blue against Tau aggregation. Biochemical and biophysical analyses using sodium dodecyl sulfate-polyacrylamide gel electrophoresis, ThS fluorescence, circular dichroism spectroscopy, and electron microscopy suggested that toluidine blue inhibited the aggregation of Tau in vitro. The photoexcited toluidine blue potentially dissolved the matured Tau fibrils, which indicated the disaggregation property of toluidine blue. The cell biology studies including the cytotoxicity assay and reactive oxygen species (ROS) production assay suggested toluidine blue to be a biocompatible dye as it reduced ROS levels and cell death. The photoexcited toluidine blue modulates the cytoskeleton network in cells, which was supported by immunofluorescence studies of neuronal cells. The studies in a UAS Tau E14 transgenic Drosophila model suggested that photoexcited toluidine blue was potent to restore the survival and memory deficits of Drosophila. The overall finding of our studies suggested toluidine blue to be a potent molecule in rescuing the Tau-mediated pathology by inhibiting its aggregation, reducing the cell death, and modulating the tubulin levels and behavioral characteristics of Drosophila. Thus, toluidine blue can be addressed as a potent molecule against Alzheimer's disease.
Collapse
Affiliation(s)
- Tushar Dubey
- Neurobiology
Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India
- Academy
of Scientific and Innovative Research (AcSIR), 411008 Pune, India
| | - Nalini Vijay Gorantla
- Neurobiology
Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India
- Academy
of Scientific and Innovative Research (AcSIR), 411008 Pune, India
| | | | - Subashchandrabose Chinnathambi
- Neurobiology
Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India
- Academy
of Scientific and Innovative Research (AcSIR), 411008 Pune, India
- E-mail: .
Tel: +91-20-25902232. Fax: +91-20-25902648
| |
Collapse
|
38
|
Gorantla N, Landge VG, Nagaraju PG, Priyadarshini CG P, Balaraman E, Chinnathambi S. Molecular Cobalt(II) Complexes for Tau Polymerization in Alzheimer's Disease. ACS OMEGA 2019; 4:16702-16714. [PMID: 31646215 PMCID: PMC6796896 DOI: 10.1021/acsomega.9b00692] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 08/01/2019] [Indexed: 05/15/2023]
Abstract
Tau is an axonal protein known to form abnormal aggregates and is the biomarker of Alzheimer's disease. Metal-based therapeutics for inhibition of Tau aggregation is limited and rarely reported in contemporary science. Here, we report the first example of rationally designed molecular cobalt(II)-complexes for effective inhibition of Tau and disaggregation of preformed Tau fibrils. The mechanistic studies reveal that prevention of Tau aggregation by cobalt-based metal complexes (CBMCs) is concentration-dependent and Tau seldom exhibits conformational changes. Interestingly, CBMCs play dual role in causing disassembly of preformed aggregates as well as inhibition of complete Tau aggregation. Furthermore, CBMCs were nontoxic and maintained the tubulin network intact. CBMCs also prevented okadaic acid-induced toxicity in SH-SY5Y cells thus, preventing hyperphosphorylation of Tau. We believe that this unprecedented finding by the newly developed molecular complexes has a potential toward metal-based therapeutics for Alzheimer's disease.
Collapse
Affiliation(s)
- Nalini
Vijay Gorantla
- Neurobiology
Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India
- Academy
of Scientific and Innovative
Research (AcSIR), 411008 Pune, India
| | - Vinod G. Landge
- Department
of Chemistry, Indian Institute of Science
Education and Research (IISER) Tirupati, 517507 Tirupati, India
| | - Pramod Gudigenahally Nagaraju
- Academy
of Scientific and Innovative
Research (AcSIR), 411008 Pune, India
- Department
of Molecular Nutrition, CSIR-CFTRI, 570020 Mysore, India
| | - Poornima Priyadarshini CG
- Academy
of Scientific and Innovative
Research (AcSIR), 411008 Pune, India
- Department
of Molecular Nutrition, CSIR-CFTRI, 570020 Mysore, India
| | - Ekambaram Balaraman
- Department
of Chemistry, Indian Institute of Science
Education and Research (IISER) Tirupati, 517507 Tirupati, India
| | - Subashchandrabose Chinnathambi
- Neurobiology
Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India
- Academy
of Scientific and Innovative
Research (AcSIR), 411008 Pune, India
| |
Collapse
|
39
|
Baicalein suppresses Repeat Tau fibrillization by sequestering oligomers. Arch Biochem Biophys 2019; 675:108119. [DOI: 10.1016/j.abb.2019.108119] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/05/2019] [Accepted: 09/24/2019] [Indexed: 12/16/2022]
|
40
|
Liu L, Zhu H, Wu W, Shen Y, Lin X, Wu Y, Liu L, Tang J, Zhou Y, Sun F, Lin HW. Neoantimycin F, a Streptomyces-Derived Natural Product Induces Mitochondria-Related Apoptotic Death in Human Non-Small Cell Lung Cancer Cells. Front Pharmacol 2019; 10:1042. [PMID: 31619992 PMCID: PMC6760012 DOI: 10.3389/fphar.2019.01042] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 08/16/2019] [Indexed: 01/15/2023] Open
Abstract
Streptomyces-derived natural products have been become a major focus of anti-tumor drug discovery studies. Neoantimycin F (NAT-F), was isolated from Streptomyces conglobatus by our group. Here, we examined the anti-cancer activities and its underlying molecular mechanisms implicated in NAT-F-induced apoptosis of non-small cell lung cancer (NSCLC) cells. Our results showed that NAT-F exerted excellent growth-inhibitory activity against PC9 and H1299 cells in a concentration-dependent manner. NAT-F-induced cell cycle arrest at S and G0/G1 phase in PC9 and H1299 cells, respectively. Further investigation revealed that the key proteins (including cyclinD1, cyclinE1, cyclinB1, CDK2, and CDK4) were involved in the cell regulation by NAT-F. Additionally, NAT-F significantly increased the production of reactive oxygen species (ROS), induced DNA damage, nuclear condensation, and cell apoptosis in both cell lines. Moreover, loss of the mitochondrial membrane potential (MMP) was markedly induced by NAT-F. Additional results revealed that NAT-F could up-regulate pro-apoptotic protein Bax and down-regulate anti-apoptotic protein Bcl-2, Mcl-1, and Bcl-xL, resulting in cytochrome c release from mitochondria and sequential activation of caspase-9 and -3, as well as the cleavage of poly (ADP-ribose) polymerase. Meanwhile, c-Jun N-terminal kinase (JNK), p38 MAPK (p38), and extracellular signal-regulated kinase (ERK) signaling pathway were also involved in anti-cancer activity of NAT-F in NSCLC cells. Taken together, these findings indicated that NAT-F possessed anti-proliferative effect and induced apoptosis in NSCLC cells in vitro and may be conducive to promote the development of novel anti-NSCLC agents.
Collapse
Affiliation(s)
- Liyun Liu
- Research Center for Marine Drugs, State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongrui Zhu
- School of Life Sciences and Biopharmaceutical Sciences, Shenyang Pharmaceutical University, Liaoning, China
| | - Wei Wu
- Research Center for Marine Drugs, State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yaoyao Shen
- Research Center for Marine Drugs, State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao Lin
- College of Pharmacy, Jinan University, Guangzhou, China
| | - Ying Wu
- Research Center for Marine Drugs, State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li Liu
- Research Center for Marine Drugs, State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Tang
- Research Center for Marine Drugs, State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yongjun Zhou
- Research Center for Marine Drugs, State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Fan Sun
- Research Center for Marine Drugs, State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hou-Wen Lin
- Research Center for Marine Drugs, State Key Laboratory of Oncogenes and Related Genes, Department of Pharmacy, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
41
|
Saravanan K, Sivanandam M, Hunday G, Pavan MS, Kumaradhas P. Exploring the different environments effect of piperine via combined crystallographic, QM/MM and molecular dynamics simulation study. J Mol Graph Model 2019; 92:280-295. [PMID: 31425905 DOI: 10.1016/j.jmgm.2019.07.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 12/20/2022]
Abstract
Piperine is a pungent alkaloid, largely present in the skin of pepper. It is the most active component of pepper and being used as a medicine in many Asian countries. The effect of piperine on memory impairment and neurodegeneration in Alzheimer's disease model has been investigated. In the present study, we aim to investigate the effect of piperine molecule in different environments (crystal and active site of proteins) from crystallography, molecular docking, QM/MM based charge density analysis and molecular dynamic simulation. The crystal structure of piperine has been used to determine the topological electron density of intermolecular interactions. The O-atoms of piperine is forming C-H⋅⋅⋅O interactions with the neighboring molecules in the crystal, these interactions also confirmed from the Hirshfeld surface. Further, to understand the nature of interactions and the conformational flexibility of piperine in the active site of recombinant human acetylcholinesterase (rhAChE), molecular docking analysis has been performed. The selected docked complex suggests favorable hydrogen bonding and hydrophobic interactions with rhAChE enzyme; notably, the O3 atom of piperine molecule forms strong hydrogen bonding interaction with Glu202 at 1.8 Å. To determine the charge density distribution and the electrostatic properties of piperine molecule in the active site of rhAChE, the piperine-rhAChE complex was minimized at QM/MM energy level; in which, the binding pocket with piperine was considered as QM region. The charge density analysis of piperine and the interacting amino acid groups have been carried out. The topological analysis of O3⋯H-O/Glu202 hydrogen bonding interaction exhibits strong interactions and the electron density ρcp(r): 0.242 eÅ-3 and the Laplacian ∇2ρcp(r): 3.176 eÅ-5 respectively. These results were compared with the corresponding molecule present in the crystal and gas phase environments of piperine. The comparison of active site structure with the corresponding crystal phase and gas phase structures reveal that piperine exhibits large conformational modification in the active site. The molecular dynamics simulation and binding free energy calculations were performed, this gives the stability, binding affinity of the molecule in the active site of rhAChE. The O3⋯H-O/Glu202 interaction shows the high stability (89.2%), this was confirmed from the stability of hydrogen bond analysis. The binding free energy was used to measure the rate of inhibition of enzyme in the presence of ligand molecule. The comparative study allows to understand the nature of piperine molecule in the gas and crystal phases, and amino acids environment.
Collapse
Affiliation(s)
- Kandasamy Saravanan
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, 636 011, India
| | - Magudeeswaran Sivanandam
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, 636 011, India
| | - Govindasamy Hunday
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, 636 011, India
| | - Mysore S Pavan
- Solid State and Structural Chemistry Unit, Indian Institute of Science, Bangalore, 560 012, India
| | - Poomani Kumaradhas
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, 636 011, India.
| |
Collapse
|
42
|
Talwar P, Gupta R, Kushwaha S, Agarwal R, Saso L, Kukreti S, Kukreti R. Viral Induced Oxidative and Inflammatory Response in Alzheimer's Disease Pathogenesis with Identification of Potential Drug Candidates: A Systematic Review using Systems Biology Approach. Curr Neuropharmacol 2019; 17:352-365. [PMID: 29676229 PMCID: PMC6482477 DOI: 10.2174/1570159x16666180419124508] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 03/19/2018] [Accepted: 04/10/2018] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is genetically complex with multifactorial etiology. Here, we aim to identify the potential viral pathogens leading to aberrant inflammatory and oxidative stress response in AD along with potential drug candidates using systems biology approach. We retrieved protein interactions of amyloid precursor protein (APP) and tau protein (MAPT) from NCBI and genes for oxidative stress from NetAge, for inflammation from NetAge and InnateDB databases. Genes implicated in aging were retrieved from GenAge database and two GEO expression datasets. These genes were individually used to create protein-protein interaction network using STRING database (score≥0.7). The interactions of candidate genes with known viruses were mapped using virhostnet v2.0 database. Drug molecules targeting candidate genes were retrieved using the Drug- Gene Interaction Database (DGIdb). Data mining resulted in 2095 APP, 116 MAPT, 214 oxidative stress, 1269 inflammatory genes. After STRING PPIN analysis, 404 APP, 109 MAPT, 204 oxidative stress and 1014 inflammation related high confidence proteins were identified. The overlap among all datasets yielded eight common markers (AKT1, GSK3B, APP, APOE, EGFR, PIN1, CASP8 and SNCA). These genes showed association with hepatitis C virus (HCV), Epstein- Barr virus (EBV), human herpes virus 8 and Human papillomavirus (HPV). Further, screening of drugs targeting candidate genes, and possessing anti-inflammatory property, antiviral activity along with a suggested role in AD pathophysiology yielded 12 potential drug candidates. Our study demonstrated the role of viral etiology in AD pathogenesis by elucidating interaction of oxidative stress and inflammation causing candidate genes with common viruses along with the identification of potential AD drug candidates.
Collapse
Affiliation(s)
- Puneet Talwar
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Delhi, India
| | - Renu Gupta
- Institute of Human Behaviour & Allied Sciences (IHBAS), Dilshad Garden, Delhi 110 095, India
| | - Suman Kushwaha
- Institute of Human Behaviour & Allied Sciences (IHBAS), Dilshad Garden, Delhi 110 095, India
| | - Rachna Agarwal
- Institute of Human Behaviour & Allied Sciences (IHBAS), Dilshad Garden, Delhi 110 095, India
| | - Luciano Saso
- Department of Physiology and Pharmacology, Sapienza University of Rome, Italy
| | | | - Ritushree Kukreti
- Genomics and Molecular Medicine Unit, Institute of Genomics and Integrative Biology (IGIB), Council of Scientific and Industrial Research (CSIR), Delhi, India
| |
Collapse
|
43
|
Ayaz M, Sadiq A, Junaid M, Ullah F, Ovais M, Ullah I, Ahmed J, Shahid M. Flavonoids as Prospective Neuroprotectants and Their Therapeutic Propensity in Aging Associated Neurological Disorders. Front Aging Neurosci 2019; 11:155. [PMID: 31293414 PMCID: PMC6606780 DOI: 10.3389/fnagi.2019.00155] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 06/11/2019] [Indexed: 01/13/2023] Open
Abstract
Modern research has revealed that dietary consumption of flavonoids and flavonoids-rich foods significantly improve cognitive capabilities, inhibit or delay the senescence process and related neurodegenerative disorders including Alzheimer’s disease (AD). The flavonoids rich foods such as green tea, cocoa, blue berry and other foods improve the various states of cognitive dysfunction, AD and dementia-like pathological alterations in different animal models. The mechanisms of flavonoids have been shown to be mediated through the inhibition of cholinesterases including acetylcholinesterase (AChE), and butyrylcholinesterase (BChE), β-secretase (BACE1), free radicals and modulation of signaling pathways, that are implicated in cognitive and neuroprotective functions. Flavonoids interact with various signaling protein pathways like ERK and PI3-kinase/Akt and modulate their actions, thereby leading to beneficial neuroprotective effects. Moreover, they enhance vascular blood flow and instigate neurogenesis particularly in the hippocampus. Flavonoids also hamper the progression of pathological symptoms of neurodegenerative diseases by inhibiting neuronal apoptosis induced by neurotoxic substances including free radicals and β-amyloid proteins (Aβ). All these protective mechanisms contribute to the maintenance of number, quality of neurons and their synaptic connectivity in the brain. Thus flavonoids can thwart the progression of age-related disorders and can be a potential source for the design and development of new drugs effective in cognitive disorders.
Collapse
Affiliation(s)
- Muhammad Ayaz
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Abdul Sadiq
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Muhammad Junaid
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan.,Department of Pharmacy, University of Swabi, Swabi, Pakistan
| | - Farhat Ullah
- Department of Pharmacy, University of Malakand, Chakdara, Pakistan
| | - Muhammad Ovais
- University of Chinese Academy of Sciences, Beijing, China.,Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Ikram Ullah
- Suliman Bin Abdullah Aba-Alkhail Centre for Interdisciplinary Research in Basic Sciences, International Islamic University Islamabad, Islamabad, Pakistan
| | - Jawad Ahmed
- Institute of Basic Medical Sciences (IBMS), Khyber Medical University, Peshawar, Pakistan
| | - Muhammad Shahid
- Department of Pharmacy, Sarhad University of Science and Information Technology (SUIT), Peshawar, Pakistan
| |
Collapse
|
44
|
Zholos AV, Moroz OF, Storozhuk MV. Curcuminoids and Novel Opportunities for the Treatment of Alzheimer's Disease: Which Molecules are Actually Effective? Curr Mol Pharmacol 2019; 12:12-26. [PMID: 30318014 DOI: 10.2174/1874467211666181012150847] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2018] [Revised: 09/30/2018] [Accepted: 10/08/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Millions of people worldwide are suffering from Alzheimer's disease (AD), and there are only symptomatic treatments available for this disease. Thus, there is a great need to identify drugs capable of arresting or reversing AD. Constituents of the spice turmeric, in particular, curcuminoids, seem to be very promising, as evident from in vitro experiments and tests using animal models of AD. However, most of the clinical trials did not reveal any beneficial effects of curcuminoids in the treatment of AD. These controversies, including conflicting results of clinical trials, are thought to be related to bioavailability of curcuminoids, which is low unless it is enhanced by developing a special formulation. However, there is growing evidence suggesting that other reasons may be of even greater importance, but these avenues are less explored. OBJECTIVE Review relevant literature, and analyze potential reasons for the controversial results. METHODOLOGY Recent in vitro and preclinical studies; clinical trials (without a limiting period) were searched in PubMed and Google Scholar. RESULTS While recent in vitro and preclinical studies confirm the therapeutic potential of curcuminoids in the treatment of AD and cognitive dysfunctions, results of corresponding clinical trials remain rather controversial. CONCLUSION The controversial results obtained in the clinical trials may be in part due to particularities of the curcuminoid formulations other than bioavailability. Namely, it seems likely that the various formulations differ in terms of their minor turmeric constituent(s). We hypothesize that these distinctions may be of key importance for efficacy of the particular formulation in clinical trials. A testable approach addressing this hypothesis is suggested.
Collapse
Affiliation(s)
- Alexander V Zholos
- A.A. Bogomoletz Institute of Physiology, National Academy of Science of Ukraine, 4 Bogomoletz Street, Kiev 01024, Ukraine.,Taras Shevchenko National University of Kyiv, Educational and Scientific Centre "Institute of Biology and Medicine", 2 Academician Glushkov Avenue, Kiev 03022, Ukraine
| | - Olesia F Moroz
- Taras Shevchenko National University of Kyiv, Educational and Scientific Centre "Institute of Biology and Medicine", 2 Academician Glushkov Avenue, Kiev 03022, Ukraine
| | - Maksim V Storozhuk
- A.A. Bogomoletz Institute of Physiology, National Academy of Science of Ukraine, 4 Bogomoletz Street, Kiev 01024, Ukraine
| |
Collapse
|
45
|
Gorantla NV, Das R, Mulani FA, Thulasiram HV, Chinnathambi S. Neem Derivatives Inhibits Tau Aggregation. J Alzheimers Dis Rep 2019; 3:169-178. [PMID: 31259310 PMCID: PMC6597962 DOI: 10.3233/adr-190118] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tau is a phosphoprotein with natively unfolded conformation that functions to stabilize microtubules in axons. Alzheimer’s disease pathology triggers several modifications in tau, which causes it to lose its affinity towards microtubule, thus, leading to microtubule disassembly and loss of axonal integrity. This elicit accumulation of tau as paired helical filaments is followed by stable neurofibrillary tangles formation. A large number of small molecules have been isolated from Azadirachta indica with varied medicinal applications. The intermediate and final limonoids, nimbin and salannin respectively, isolated from Azadirachta indica, were screened against tau aggregation. ThS and ANS fluorescence assay showed the role of intermediate and final limonoids in preventing heparin induced cross-β sheet formation and also decreased hydrophobicity, which are characteristic nature of tau aggregation. Transmission electron microscopy studies revealed that limonoids restricted the aggregation of tau to fibrils; in turn, limonoids led to the formation of short and fragile aggregates. Both the limonoids were non-toxic to HEK293T cells thus, substantiating limonoids as a potential lead in overcoming Alzheimer’s disease.
Collapse
Affiliation(s)
- Nalini V Gorantla
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, India
| | - Rashmi Das
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, India
| | - Fayaj A Mulani
- Division of Organic Chemistry, CSIR-National Chemical Laboratory, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, India
| | - Hirekodathakallu V Thulasiram
- Division of Organic Chemistry, CSIR-National Chemical Laboratory, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, India
| | - Subashchandrabose Chinnathambi
- Neurobiology Group, Division of Biochemical Sciences, CSIR-National Chemical Laboratory, Pune, India.,Academy of Scientific and Innovative Research (AcSIR), Pune, India
| |
Collapse
|
46
|
Sivakumar M, Saravanan K, Saravanan V, Sugarthi S, kumar SM, Alhaji Isa M, Rajakumar P, Aravindhan S. Discovery of new potential triplet acting inhibitor for Alzheimer’s disease via X-ray crystallography, molecular docking and molecular dynamics. J Biomol Struct Dyn 2019; 38:1903-1917. [DOI: 10.1080/07391102.2019.1620128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
| | - Kandasamy Saravanan
- X-Ray Crystallography and Computational Molecular Biology Lab, Department of Physics, Periyar University, Salem, India
| | | | - Srinivasan Sugarthi
- Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur, Kancheepuram, Tamil Nadu, India
| | | | - Mustafa Alhaji Isa
- Bioinformatics and Computational Biology Lab, Department of Microbiology, Faculty of Sciences, University of Maiduguri, Maiduguri, Nigeria
| | - Perumal Rajakumar
- Department of Organic Chemistry, University of Madras, Chennai, India
| | | |
Collapse
|
47
|
Saravanan K, Hunday G, Kumaradhas P. Binding and stability of indirubin-3-monoxime in the GSK3β enzyme: a molecular dynamics simulation and binding free energy study. J Biomol Struct Dyn 2019; 38:957-974. [DOI: 10.1080/07391102.2019.1591301] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Kandasamy Saravanan
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, Tamil Nadu, India
| | - Govindasamy Hunday
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, Tamil Nadu, India
| | - Poomani Kumaradhas
- Laboratory of Biocrystallography and Computational Molecular Biology, Department of Physics, Periyar University, Salem, Tamil Nadu, India
| |
Collapse
|
48
|
Kizhakke P. A, Olakkaran S, Antony A, Tilagul K. S, Hunasanahally P. G. Convolvulus pluricaulis (Shankhapushpi) ameliorates human microtubule-associated protein tau (hMAPτ) induced neurotoxicity in Alzheimer’s disease Drosophila model. J Chem Neuroanat 2019; 95:115-122. [DOI: 10.1016/j.jchemneu.2017.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/27/2017] [Accepted: 10/15/2017] [Indexed: 10/18/2022]
|
49
|
Stone J, Mitrofanis J, Johnstone DM, Falsini B, Bisti S, Adam P, Nuevo AB, George-Weinstein M, Mason R, Eells J. Acquired Resilience: An Evolved System of Tissue Protection in Mammals. Dose Response 2018; 16:1559325818803428. [PMID: 30627064 PMCID: PMC6311597 DOI: 10.1177/1559325818803428] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 08/22/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022] Open
Abstract
This review brings together observations on the stress-induced regulation of resilience mechanisms in body tissues. It is argued that the stresses that induce tissue resilience in mammals arise from everyday sources: sunlight, food, lack of food, hypoxia and physical stresses. At low levels, these stresses induce an organised protective response in probably all tissues; and, at some higher level, cause tissue destruction. This pattern of response to stress is well known to toxicologists, who have termed it hormesis. The phenotypes of resilience are diverse and reports of stress-induced resilience are to be found in journals of neuroscience, sports medicine, cancer, healthy ageing, dementia, parkinsonism, ophthalmology and more. This diversity makes the proposing of a general concept of induced resilience a significant task, which this review attempts. We suggest that a system of stress-induced tissue resilience has evolved to enhance the survival of animals. By analogy with acquired immunity, we term this system 'acquired resilience'. Evidence is reviewed that acquired resilience, like acquired immunity, fades with age. This fading is, we suggest, a major component of ageing. Understanding of acquired resilience may, we argue, open pathways for the maintenance of good health in the later decades of human life.
Collapse
Affiliation(s)
- Jonathan Stone
- Discipline of Physiology, Bosch Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - John Mitrofanis
- Discipline of Anatomy and Histology, Bosch Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Daniel M. Johnstone
- Discipline of Physiology, Bosch Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Benedetto Falsini
- Facolta’ di Medicina e Chirurgia, Fondazione Policlinico A. Gemelli, Universita’ Cattolica del S. Cuore, Rome, Italy
| | - Silvia Bisti
- Department of Biotechnical and Applied Clinical Sciences, Università degli Studi dell’Aquila, IIT Istituto Italiano di Tecnologia Genova and INBB Istituto Nazionale Biosistemi e Biostrutture, Rome, Italy
| | - Paul Adam
- School of Biological, Earth and Environmental Science, University of New South Wales, Sydney, New South Wales, Australia
| | - Arturo Bravo Nuevo
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Mindy George-Weinstein
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA, USA
| | - Rebecca Mason
- Discipline of Physiology, Bosch Institute of Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Janis Eells
- College of Health Sciences, University of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
50
|
Nieto CI, Cornago MP, Cabildo MP, Sanz D, Claramunt RM, Torralba MC, Torres MR, Martínez Casanova D, Sánchez-Alegre YR, Escudero E, Lavandera JL. Evaluation of the Antioxidant and Neuroprotectant Activities of New Asymmetrical 1,3-Diketones. Molecules 2018; 23:E1837. [PMID: 30042315 PMCID: PMC6222706 DOI: 10.3390/molecules23081837] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/16/2018] [Accepted: 07/17/2018] [Indexed: 01/08/2023] Open
Abstract
A series of fourteen new asymmetrical 1,3-diketone derivatives have been synthesized and evaluated in the ABTS, FRAP and DPPH assays as a new chemotype with antioxidant and drug-like properties. All the compounds displayed low cytotoxicity in comparison to curcumin against the human neuroblastoma SH-SY5Y cell line. Among them, (3Z,5E)-6-(2,5-difluoro-4-hydroxy-phenyl)-1,1,1-trifluoro-4-hydroxyhexa-3,5-dien-2-one (6b) and (3Z,5E)-6-(2,3-difluoro-4-hydroxy-phenyl)-1,1,1-trifluoro-4-hydroxyhexa-3,5-dien-2-one (7b) with excellent solubility and chemical stability in biorelevant media, have also shown a similar Fe+2 chelation behavior to that of curcumin. Additionally, both derivatives 6b and 7b have afforded good neuroprotection activity against H₂O₂ induced oxidative stress in the same neuronal cell line, with a significant reduction of intracellular ROS levels, in parallel with a good recovery of the Mitochondrial Membrane Potential (ΔΨm). Compounds 6b and 7b with a promising antioxidant and drug-like profile, with low cytotoxic and good neuroprotectant activity, constitute a new interesting chemical class with high potential as new therapeutic agents against neurodegenerative diseases.
Collapse
Affiliation(s)
- Carla I Nieto
- Departamento de Química Orgánica y Bio-Orgánica, Facultad de Ciencias, Universidad Nacional de Educación a distancia (UNED), Paseo Senda del Rey 9, E-28040 Madrid, Spain.
| | - María Pilar Cornago
- Departamento de Química Orgánica y Bio-Orgánica, Facultad de Ciencias, Universidad Nacional de Educación a distancia (UNED), Paseo Senda del Rey 9, E-28040 Madrid, Spain.
| | - María Pilar Cabildo
- Departamento de Química Orgánica y Bio-Orgánica, Facultad de Ciencias, Universidad Nacional de Educación a distancia (UNED), Paseo Senda del Rey 9, E-28040 Madrid, Spain.
| | - Dionisia Sanz
- Departamento de Química Orgánica y Bio-Orgánica, Facultad de Ciencias, Universidad Nacional de Educación a distancia (UNED), Paseo Senda del Rey 9, E-28040 Madrid, Spain.
| | - Rosa M Claramunt
- Departamento de Química Orgánica y Bio-Orgánica, Facultad de Ciencias, Universidad Nacional de Educación a distancia (UNED), Paseo Senda del Rey 9, E-28040 Madrid, Spain.
| | - María Carmen Torralba
- Departamento de Química Inorgánica I and CAI de Difracción de Rayos-X, Facultad de Ciencias Químicas, Universidad Complutense de Madrid (UCM), E-28040 Madrid, Spain.
| | - María Rosario Torres
- Departamento de Química Inorgánica I and CAI de Difracción de Rayos-X, Facultad de Ciencias Químicas, Universidad Complutense de Madrid (UCM), E-28040 Madrid, Spain.
| | - Diana Martínez Casanova
- Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad CEU San Pablo, Campus de Montepríncipe, Boadilla, E-28668 Madrid, Spain.
| | - Yaiza Rebeca Sánchez-Alegre
- Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad CEU San Pablo, Campus de Montepríncipe, Boadilla, E-28668 Madrid, Spain.
| | - Esther Escudero
- Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad CEU San Pablo, Campus de Montepríncipe, Boadilla, E-28668 Madrid, Spain.
| | - José Luis Lavandera
- Instituto de Medicina Molecular Aplicada (IMMA), Facultad de Medicina, Universidad CEU San Pablo, Campus de Montepríncipe, Boadilla, E-28668 Madrid, Spain.
| |
Collapse
|