1
|
Nair AC, Benny S, Aneesh TP, Sudheesh MS, Lakshmi PK. Comprehensive profiling of traditional herbomineral formulation Manasamitra vatakam in rat brain following oral administration and in-silico screening of the identified compound for anti-Alzheimer's activity. JOURNAL OF ETHNOPHARMACOLOGY 2024; 338:119024. [PMID: 39489356 DOI: 10.1016/j.jep.2024.119024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Multi-targeted drug therapy has received substantial attention for the treatment of diseases of multifactorial origin, such as neurodegenerative diseases. Manasamitra vatakam (MMV) is a traditional Ayurvedic formulation used to improve cognitive impairment and mental illness. Here we have used a unique method for leveraging the barrier properties of the intestinal and blood-brain barrier (BBB) to screen and identify the bioactive molecules against Alzheimer's disease (AD). The current method exemplifies a facile method to expedite drug discovery from traditional formulations. AIM OF THE STUDY The present study aimed to identify the phytoconstituents of MMV that reach the brain tissue and to predict major bioactive constituents by computational docking studies. MATERIALS AND METHODS After oral administration of the formulation, brain samples from male Sprague Dawley rats were collected at different time intervals and analyzed by liquid chromatography-mass spectrometry (LC-MS) to identify the phytoconstituents. In silico molecular docking studies were carried out to analyze the binding affinity of the compounds to the target proteins of AD using Schrodinger Maestro. The molecular dynamic studies were carried out for all the docked complexes having higher docking scores. RESULTS 34 phytoconstituents were identified by LC-MS analysis of brain homogenates. In the in silico docking study, the phytoconstituents chrysin, convolvin, rutin, galangin, palmatoside G, isoliquiritigenin, quercetin, and naringenin showed higher docking score against the target proteins of AD. These compounds may serve as the primary bioactive compounds responsible for the neuroprotective activity of the herbal formulation. Furthermore, molecular dynamic studies indicated that the galangin-acetylcholinesterase enzyme complex has the highest stability among these eight compounds. CONCLUSION The study, together with previous in vivo and in vitro efficacy results, suggests that BBB-permeable compounds with high binding affinities for the target proteins of AD might be responsible for the effectiveness of MMV against AD.
Collapse
Affiliation(s)
- Anju C Nair
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India.
| | - Sonu Benny
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India.
| | - T P Aneesh
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India.
| | - M S Sudheesh
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India.
| | - P K Lakshmi
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India.
| |
Collapse
|
2
|
Niazi SK. Bioavailability as Proof to Authorize the Clinical Testing of Neurodegenerative Drugs-Protocols and Advice for the FDA to Meet the ALS Act Vision. Int J Mol Sci 2024; 25:10211. [PMID: 39337696 PMCID: PMC11432374 DOI: 10.3390/ijms251810211] [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: 08/10/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Although decades of intensive drug discovery efforts to treat neurodegenerative disorders (NDs) have failed, around half a million patients in more than 2000 studies continue being tested, costing over USD 100 billion, despite the conclusion that even those drugs which have been approved have no better effect than a placebo. The US Food and Drug Administration (FDA) has established multiple programs to innovate the treatment of rare diseases, particularly NDs, providing millions of USD in funding primarily by encouraging novel clinical trials to account for issues related to study sizes and adopting multi-arm studies to account for patient dropouts. Instead, the FDA should focus on the primary reason for failure: the poor bioavailability of drugs reaching the brain (generally 0.1% at most) due to the blood-brain barrier (BBB). There are several solutions to enhance entry into the brain, and the FDA must require proof of significant entry into the brain as the prerequisite to approving Investigational New Drug (IND) applications. The FDA should also rely on factors other than biomarkers to confirm efficacy, as these are rarely relevant to clinical use. This study summarizes how the drugs used to treat NDs can be made effective and how the FDA should change its guidelines for IND approval of these drugs.
Collapse
Affiliation(s)
- Sarfaraz K Niazi
- College of Pharmacy, University of Illinois, Chicago, IL 60612, USA
| |
Collapse
|
3
|
Lasure VU, Singh Gautam A, Singh RK. Quercetin ameliorates neuroinflammatory and neurodegenerative biomarkers in the brain and improves neurobehavioral parameters in a repeated intranasal amyloid-beta exposed model of Alzheimer's disease. Food Funct 2024; 15:8712-8728. [PMID: 39087409 DOI: 10.1039/d4fo02602k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
Objectives: The aim of the present study was to study the potential therapeutic effects of quercetin in protection against repeated intranasal exposure of an amyloid-beta-induced mouse model. Methods: Mice received intranasal Aβ1-42 (5 μg/10 μL) exposure once daily for seven consecutive days. Quercetin was orally administered to them at 30 mg kg-1 and 100 mg kg-1 doses for one week starting from day five following Aβ1-42 peptide administration. Following this, the animals were evaluated for neurobehavioral parameters using a Morris water maze test and a novel object recognition test. Further to this, the biomarkers for neuroinflammation and neurodegeneration were evaluated in the hippocampus and cortex regions of the brain in these animals. Results: Multiple exposures to intranasal Aβ led to a significant decline in the learning and cognitive memory of the animals, whereas oral treatment with quercetin at dosages of 30 and 100 mg kg-1 alleviated Aβ-induced effects. Quercetin treatment significantly reduced Aβ accumulation, oxidative stress and proinflammatory cytokine biomarkers in the brain. In addition, it also alleviated the activation of astrocytic biomarkers, amyloid precursor protein and phosphorylated-tau proteins in the brain. Conclusion: Quercetin was found to be a potent antioxidant, anti-inflammatory compound with protection against neurodegenerative damage and improved learning and cognitive memory in a repeated Aβ-exposure model of AD.
Collapse
Affiliation(s)
- Vaibhav Uttamrao Lasure
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Transit Campus, Bijnour-Sisendi Road, Sarojini Nagar, Lucknow-226002, Uttar Pradesh, India.
| | - Avtar Singh Gautam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Transit Campus, Bijnour-Sisendi Road, Sarojini Nagar, Lucknow-226002, Uttar Pradesh, India.
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Raebareli, Transit Campus, Bijnour-Sisendi Road, Sarojini Nagar, Lucknow-226002, Uttar Pradesh, India.
| |
Collapse
|
4
|
Coimbra JRM, Resende R, Custódio JBA, Salvador JAR, Santos AE. BACE1 Inhibitors for Alzheimer's Disease: Current Challenges and Future Perspectives. J Alzheimers Dis 2024; 101:S53-S78. [PMID: 38943390 DOI: 10.3233/jad-240146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2024]
Abstract
Disease-modifying therapies (DMT) for Alzheimer's disease (AD) are highly longed-for. In this quest, anti-amyloid therapies take center stage supported by genetic facts that highlight an imbalance between production and clearance of amyloid-β peptide (Aβ) in AD patients. Indeed, evidence from basic research, human genetic and biomarker studies, suggests the accumulation of Aβ as a driver of AD pathogenesis and progression. The aspartic protease β-site AβPP cleaving enzyme (BACE1) is the initiator for Aβ production. Underpinning a critical role for BACE1 in AD pathophysiology are the elevated BACE1 concentration and activity observed in the brain and body fluids of AD patients. Therefore, BACE1 is a prime drug target for reducing Aβ levels in early AD. Small-molecule BACE1 inhibitors have been extensively developed for the last 20 years. However, clinical trials with these molecules have been discontinued for futility or safety reasons. Most of the observed adverse side effects were due to other aspartic proteases cross-inhibition, including the homologue BACE2, and to mechanism-based toxicity since BACE1 has substrates with important roles for synaptic plasticity and synaptic homeostasis besides amyloid-β protein precursor (AβPP). Despite these setbacks, BACE1 persists as a well-validated therapeutic target for which a specific inhibitor with high substrate selectivity may yet to be found. In this review we provide an overview of the evolution in BACE1 inhibitors design pinpointing the molecules that reached advanced phases of clinical trials and the liabilities that precluded adequate trial effects. Finally, we ponder on the challenges that anti-amyloid therapies must overcome to achieve clinical success.
Collapse
Affiliation(s)
- Judite R M Coimbra
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Rosa Resende
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - José B A Custódio
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Laboratory of Biochemistry and Biology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Jorge A R Salvador
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Armanda E Santos
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Laboratory of Biochemistry and Biology, Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
5
|
Rivai B, Umar AK. Neuroprotective compounds from marine invertebrates. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2023; 12:71. [DOI: 10.1186/s43088-023-00407-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/22/2023] [Indexed: 09/01/2023] Open
Abstract
Abstract
Background
Neuroinflammation is a key pathological feature of a wide variety of neurological disorders, including Parkinson’s, multiple sclerosis, Alzheimer’s, and Huntington’s disease. While current treatments for these disorders are primarily symptomatic, there is a growing interest in developing new therapeutics that target the underlying neuroinflammatory processes.
Main body
Marine invertebrates, such as coral, sea urchins, starfish, sponges, and sea cucumbers, have been found to contain a wide variety of biologically active compounds that have demonstrated potential therapeutic properties. These compounds are known to target various key proteins and pathways in neuroinflammation, including 6-hydroxydopamine (OHDH), caspase-3 and caspase-9, p-Akt, p-ERK, p-P38, acetylcholinesterase (AChE), amyloid-β (Aβ), HSF-1, α-synuclein, cellular prion protein, advanced glycation end products (AGEs), paraquat (PQ), and mitochondria DJ-1.
Short conclusion
This review focuses on the current state of research on the neuroprotective effects of compounds found in marine invertebrates and the potential therapeutic implications of these findings for treating neuroinflammatory disorders. We also discussed the challenges and limitations of using marine-based compounds as therapeutics, such as sourcing and sustainability concerns, and the need for more preclinical and clinical studies to establish their efficacy and safety.
Graphical abstract
Collapse
|
6
|
Mycroft-West CJ, Devlin AJ, Cooper LC, Guimond SE, Procter P, Miller GJ, Guerrini M, Fernig DG, Yates EA, Lima MA, Skidmore MA. A sulphated glycosaminoglycan extract from Placopecten magellanicus inhibits the Alzheimer's disease β-site amyloid precursor protein cleaving enzyme 1 (BACE-1). Carbohydr Res 2023; 525:108747. [PMID: 36773398 DOI: 10.1016/j.carres.2023.108747] [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: 04/15/2022] [Revised: 10/05/2022] [Accepted: 01/19/2023] [Indexed: 01/27/2023]
Abstract
The clinically important anticoagulant heparin, a member of the glycosaminoglycan family of carbohydrates that is extracted predominantly from porcine and bovine tissue sources, has previously been shown to inhibit the β-site amyloid precursor protein cleaving enzyme 1 (BACE-1), a key drug target in Alzheimer's Disease. In addition, heparin has been shown to exert favourable bioactivities through a number of pathophysiological pathways involved in the disease processes of Alzheimer's Disease including inflammation, oxidative stress, tau phosphorylation and amyloid peptide generation. Despite the multi-target potential of heparin as a therapeutic option for Alzheimer's disease, the repurposing of this medically important biomolecule has to-date been precluded by its high anticoagulant potential. An alternative source to mammalian-derived glycosaminoglycans are those extracted from marine environments and these have been shown to display an expanded repertoire of sequence-space and heterogeneity compared to their mammalian counterparts. Furthermore, many marine-derived glycosaminoglycans appear to retain favourable bioactivities, whilst lacking the high anticoagulant potential of their mammalian counterparts. Here we describe a sulphated, marine-derived glycosaminoglycan extract from the Atlantic Sea Scallop, Placopecten magellanicus that displays high inhibitory potential against BACE-1 (IC50 = 4.8 μg.mL-1) combined with low anticoagulant activity; 25-fold less than that of heparin. This extract possesses a more favourable therapeutic profile compared to pharmaceutical heparin of mammalian provenance and is composed of a mixture of heparan sulphate (HS), with a high content of 6-sulphated N-acetyl glucosamine (64%), and chondroitin sulphate.
Collapse
Affiliation(s)
- Courtney J Mycroft-West
- Centre for Glycoscience Research and Training, Keele University, Keele, Staffordshire, ST5 5BG, UK.
| | - Anthony J Devlin
- Centre for Glycoscience Research and Training, Keele University, Keele, Staffordshire, ST5 5BG, UK; Istituto di Ricerche Chimiche e Biochimiche G. Ronzoni, Via G. Colombo 81, 20133, Milan, Italy.
| | - Lynsay C Cooper
- University of Gloucestershire, Francis Close Hall Campus, Swindon Rd, Cheltenham, GL50 4AZ, UK.
| | - Scott E Guimond
- Centre for Glycoscience Research and Training, Keele University, Keele, Staffordshire, ST5 5BG, UK.
| | - Patricia Procter
- Centre for Glycoscience Research and Training, Keele University, Keele, Staffordshire, ST5 5BG, UK.
| | - Gavin J Miller
- Centre for Glycoscience Research and Training, Keele University, Keele, Staffordshire, ST5 5BG, UK.
| | - Marco Guerrini
- Istituto di Ricerche Chimiche e Biochimiche G. Ronzoni, Via G. Colombo 81, 20133, Milan, Italy.
| | - David G Fernig
- Department of Biochemistry and Systems Biology, ISMIB, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK.
| | - Edwin A Yates
- Department of Biochemistry and Systems Biology, ISMIB, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK.
| | - Marcelo A Lima
- Centre for Glycoscience Research and Training, Keele University, Keele, Staffordshire, ST5 5BG, UK.
| | - Mark A Skidmore
- Centre for Glycoscience Research and Training, Keele University, Keele, Staffordshire, ST5 5BG, UK; Department of Biochemistry and Systems Biology, ISMIB, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK.
| |
Collapse
|
7
|
Rajah Kumaran K, Yunusa S, Perimal E, Wahab H, Müller CP, Hassan Z. Insights into the Pathophysiology of Alzheimer's Disease and Potential Therapeutic Targets: A Current Perspective. J Alzheimers Dis 2023; 91:507-530. [PMID: 36502321 DOI: 10.3233/jad-220666] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The aging population increases steadily because of a healthy lifestyle and medical advancements in healthcare. However, Alzheimer's disease (AD) is becoming more common and problematic among older adults. AD-related cases show an increasing trend annually, and the younger age population may also be at risk of developing this disorder. AD constitutes a primary form of dementia, an irreversible and progressive brain disorder that steadily damages cognitive functions and the ability to perform daily tasks. Later in life, AD leads to death as a result of the degeneration of specific brain areas. Currently, the cause of AD is poorly understood, and there is no safe and effective therapeutic agent to cure or slow down its progression. The condition is entirely preventable, and no study has yet demonstrated encouraging findings in terms of treatment. Identifying this disease's pathophysiology can help researchers develop safe and efficient therapeutic strategies to treat this ailment. This review outlines and discusses the pathophysiology that resulted in the development of AD including amyloid-β plaques, tau neurofibrillary tangles, neuroinflammation, oxidative stress, cholinergic dysfunction, glutamate excitotoxicity, and changes in neurotrophins level may sound better based on the literature search from Scopus, PubMed, ScienceDirect, and Google Scholar. Potential therapeutic strategies are discussed to provide more insights into AD mechanisms by developing some possible pharmacological agents for its treatment.
Collapse
Affiliation(s)
- Kesevan Rajah Kumaran
- Malaysian Institute of Pharmaceuticals and Nutraceuticals, National Institutes of Biotechnology Malaysia, Halaman Bukit Gambir, Gelugor, Pulau Pinang, Malaysia
| | - Suleiman Yunusa
- Centre for Drug Research, Universiti Sains Malaysia, Penang, Malaysia.,Department of Pharmacology, Bauchi State University Gadau, Bauchi State, Nigeria
| | - Enoch Perimal
- Curtin Medical School, Curtin University, Bentley, Western Australia, Australia.,Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Habibah Wahab
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang, Malaysia
| | - Christian P Müller
- Centre for Drug Research, Universiti Sains Malaysia, Penang, Malaysia.,Section of Addiction Medicine, Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Zurina Hassan
- Centre for Drug Research, Universiti Sains Malaysia, Penang, Malaysia.,Section of Addiction Medicine, Department of Psychiatry and Psychotherapy, University Clinic, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
8
|
Cuddy LK, Alia AO, Salvo MA, Chandra S, Grammatopoulos TN, Justman CJ, Lansbury PT, Mazzulli JR, Vassar R. Farnesyltransferase inhibitor LNK-754 attenuates axonal dystrophy and reduces amyloid pathology in mice. Mol Neurodegener 2022; 17:54. [PMID: 35987691 PMCID: PMC9392365 DOI: 10.1186/s13024-022-00561-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 08/11/2022] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Amyloid plaque deposition and axonal degeneration are early events in AD pathogenesis. Aβ disrupts microtubules in presynaptic dystrophic neurites, resulting in the accumulation of impaired endolysosomal and autophagic organelles transporting β-site amyloid precursor protein cleaving enzyme (BACE1). Consequently, dystrophic neurites generate Aβ42 and significantly contribute to plaque deposition. Farnesyltransferase inhibitors (FTIs) have recently been investigated for repositioning toward the treatment of neurodegenerative disorders and block the action of farnesyltransferase (FTase) to catalyze farnesylation, a post-translational modification that regulates proteins involved in lysosome function and microtubule stability. In postmortem AD brains, FTase and its downstream signaling are upregulated. However, the impact of FTIs on amyloid pathology and dystrophic neurites is unknown. METHODS We tested the effects of the FTIs LNK-754 and lonafarnib in the 5XFAD mouse model of amyloid pathology. RESULTS In 2-month-old 5XFAD mice treated chronically for 3 months, LNK-754 reduced amyloid plaque burden, tau hyperphosphorylation, and attenuated the accumulation of BACE1 and LAMP1 in dystrophic neurites. In 5-month-old 5XFAD mice treated acutely for 3 weeks, LNK-754 reduced dystrophic neurite size and LysoTracker-Green accumulation in the absence of effects on Aβ deposits. Acute treatment with LNK-754 improved memory and learning deficits in hAPP/PS1 amyloid mice. In contrast to LNK-754, lonafarnib treatment was less effective at reducing plaques, tau hyperphosphorylation and dystrophic neurites, which could have resulted from reduced potency against FTase compared to LNK-754. We investigated the effects of FTIs on axonal trafficking of endolysosomal organelles and found that lonafarnib and LNK-754 enhanced retrograde axonal transport in primary neurons, indicating FTIs could support the maturation of axonal late endosomes into lysosomes. Furthermore, FTI treatment increased levels of LAMP1 in mouse primary neurons and in the brains of 5XFAD mice, demonstrating that FTIs stimulated the biogenesis of endolysosomal organelles. CONCLUSIONS We show new data to suggest that LNK-754 promoted the axonal trafficking and function of endolysosomal compartments, which we hypothesize decreased axonal dystrophy, reduced BACE1 accumulation and inhibited amyloid deposition in 5XFAD mice. Our results agree with previous work identifying FTase as a therapeutic target for treating proteinopathies and could have important therapeutic implications in treating AD.
Collapse
Affiliation(s)
- Leah K. Cuddy
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Alia O. Alia
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Miranda A. Salvo
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Sidhanth Chandra
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | | | | | - Peter T. Lansbury
- Bial Biotech, Cambridge, MA 02139 USA
- Department of Neurology, Harvard Medical School, Cambridge, MA 02139 USA
| | - Joseph R. Mazzulli
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Robert Vassar
- The Ken and Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
- Mesulam Center for Cognitive Neurology and Alzheimer’s Disease, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| |
Collapse
|
9
|
Singh S, Yang F, Sivils A, Cegielski V, Chu XP. Amylin and Secretases in the Pathology and Treatment of Alzheimer's Disease. Biomolecules 2022; 12:996. [PMID: 35883551 PMCID: PMC9312829 DOI: 10.3390/biom12070996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer's disease remains a prevailing neurodegenerative condition which has an array physical, emotional, and financial consequences to patients and society. In the past decade, there has been a greater degree of investigation on therapeutic small peptides. This group of biomolecules have a profile of fundamentally sound characteristics which make them an intriguing area for drug development. Among these biomolecules, there are four modulatory mechanisms of interest in this review: alpha-, beta-, gamma-secretases, and amylin. These protease-based biomolecules all have a contributory role in the amyloid cascade hypothesis. Moreover, the involvement of various biochemical pathways intertwines these peptides to have shared regulators (i.e., retinoids). Further clinical and translational investigation must occur to gain a greater understanding of its potential application in patient care. The aim of this narrative review is to evaluate the contemporary literature on these protease biomolecule modulators and determine its utility in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | - Xiang-Ping Chu
- Department of Biomedical Sciences, School of Medicine, University of Missouri, Kansas City, MO 64108, USA; (S.S.); (F.Y.); (A.S.); (V.C.)
| |
Collapse
|
10
|
Marko DM, MacPherson REK. APP Processing: A Biochemical Competition Influenced by Exercise-Induced Signaling Mediators? Am J Physiol Regul Integr Comp Physiol 2022; 323:R169-R180. [PMID: 35608263 DOI: 10.1152/ajpregu.00297.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Neurodegenerative diseases, such as Alzheimer's disease (AD), are becoming more common in aging our society. One specific neuropathological hallmark of this disease is excessive accumulation of amyloid-β (Aβ) peptides, which can aggregate to form the plaques commonly associated with this disease. These plaques are often observed well before clinical diagnosis of AD. At the cellular level, both production and aggregation of Aβ peptides in the brain is detrimental to neuronal cell production, survival, and function, as well as often resulting in neuronal dysfunction and death. Exercise and physical activity have been shown to improve overall health, including brain health, and in the last several years there has been evidence to support that exercise may be able to regulate Aβ peptide production in the brain. Exercise promotes the release of a wide array of signaling mediators from various metabolically active tissues and organs in the body. These exercise-induced signaling mediators could be the driving force behind some of the beneficial effects observed in brain with exercise. This review will aim to discuss potential exercise-induced signaling mediators with the capacity to influence various proteins involved in the formation of Aβ peptide production in the brain.
Collapse
Affiliation(s)
- Daniel M Marko
- Department of Health Sciences, Brock University, St. Catharines, Ontario, Canada
| | - Rebecca E K MacPherson
- Department of Health Sciences, Brock University, St. Catharines, Ontario, Canada.,Centre for Neuroscience, Brock University, St. Catharines, Ontario, Canada
| |
Collapse
|
11
|
Resveratrol and neuroprotection: an insight into prospective therapeutic approaches against Alzheimer's disease from bench to bedside. Mol Neurobiol 2022; 59:4384-4404. [PMID: 35545730 DOI: 10.1007/s12035-022-02859-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/28/2022] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia and cognitive impairment; yet, there is currently no treatment. A buildup of Aβ, tau protein phosphorylation, oxidative stress, and inflammation in AD is pathogenic. The accumulation of amyloid-beta (Aβ) peptides in these neurocognitive areas is a significant characteristic of the disease. Therefore, inhibiting Aβ peptide aggregation has been proposed as the critical therapeutic approach for AD treatment. Resveratrol has been demonstrated in multiple studies to have a neuroprotective, anti-inflammatory, and antioxidant characteristic and the ability to minimize Aβ peptides aggregation and toxicity in the hippocampus of Alzheimer's patients, stimulating neurogenesis and inhibiting hippocampal degeneration. Furthermore, resveratrol's antioxidant effect promotes neuronal development by activating the silent information regulator-1 (SIRT1), which can protect against the detrimental effects of oxidative stress. Resveratrol-induced SIRT1 activation is becoming more crucial in developing novel therapeutic options for AD and other diseases that have neurodegenerative characteristics. This review highlighted a better knowledge of resveratrol's mechanism of action and its promising therapeutic efficacy in treating AD. We also highlighted the therapeutic potential of resveratrol as an AD therapeutic agent, which is effective against neurodegenerative disorders.
Collapse
|
12
|
Resende R, Ferreira-Marques M, Moreira P, Coimbra JRM, Baptista SJ, Isidoro C, Salvador JAR, Dinis TCP, Pereira CF, Santos AE. New BACE1 Chimeric Peptide Inhibitors Selectively Prevent AβPP-β Cleavage Decreasing Amyloid-β Production and Accumulation in Alzheimer's Disease Models. J Alzheimers Dis 2021; 76:1317-1337. [PMID: 32597812 DOI: 10.3233/jad-200381] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND A disease-modifying therapy for Alzheimer's disease (AD) is still an unmet clinical need. The formation of amyloid-β (Aβ) requires the initial cleavage of the amyloid-β protein precursor (AβPP) by BACE1 (beta-site AβPP cleaving enzyme 1), which is a prime therapeutic target for AD. OBJECTIVE We aimed to design and develop a selective BACE1 inhibitor suitable to AD treatment. METHODS The new BACE1 inhibitors consist on a chimeric peptide including a sequence related to the human Swedish mutant form of AβPP (AβPPswe) conjugated with the TAT carrier that facilitates cell membrane permeation and the crossing of the blood-brain barrier. Additionally to the chimeric peptide in the L-form, we developed a D-retroinverso chimeric peptide. The latter strategy, never used with BACE1 inhibitors, is considered to favor a significantly higher half-life and lower immunogenicity. RESULTS We found that both chimeric peptides inhibit recombinant BACE1 activity and decrease Aβ40/42 production in Neuro-2a (N2A) cells expressing AβPPswe without inducing cytotoxicity. The intraperitoneal administration of these peptides to 3xTg-AD mice decreased plasma and brain Aβ40/42 levels, as well as brain soluble AβPPβ production. Also, a reduction of insoluble Aβ was observed in the brain after chronic treatment. Noteworthy, the chimeric peptides selectively inhibited the AβPP-β cleavage relatively to the proteolysis of other BACE1 substrates such as close homologue of L1 (CHL1) and seizure-related gene 6 (SEZ6). CONCLUSIONS Overall these new BACE1 chimeric peptideshold promising potential as a selective disease-modifying therapy for AD.
Collapse
Affiliation(s)
- Rosa Resende
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), Coimbra, Portugal.,University of Coimbra, Institute for Interdisciplinary Research (IIIUC), Coimbra, Portugal
| | - Marisa Ferreira-Marques
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), Coimbra, Portugal.,University of Coimbra, Faculty of Pharmacy, Laboratory of Pharmacology, Coimbra, Portugal
| | - Patrícia Moreira
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), Coimbra, Portugal.,University of Coimbra, Faculty of Pharmacy, Laboratory of Pharmacology, Coimbra, Portugal
| | - Judite R M Coimbra
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), Coimbra, Portugal.,University of Coimbra, Faculty of Pharmacy, Laboratory of Pharmaceutical Chemistry, Coimbra, Portugal
| | - Salete J Baptista
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), Coimbra, Portugal.,Chem4Pharma, Coimbra, Portugal
| | - Ciro Isidoro
- Laboratory of Molecular Pathology, Department of Health Sciences, Università del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Jorge A R Salvador
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), Coimbra, Portugal.,University of Coimbra, Faculty of Pharmacy, Laboratory of Pharmaceutical Chemistry, Coimbra, Portugal
| | - Teresa C P Dinis
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), Coimbra, Portugal.,University of Coimbra, Faculty of Pharmacy, Laboratory of Biochemistry and Biology, Coimbra, Portugal
| | - Cláudia F Pereira
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), Coimbra, Portugal.,University of Coimbra, Faculty of Medicine, Institute of Biochemistry, Coimbra, Portugal
| | - Armanda E Santos
- University of Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), Coimbra, Portugal.,University of Coimbra, Faculty of Pharmacy, Laboratory of Biochemistry and Biology, Coimbra, Portugal
| |
Collapse
|
13
|
Mycroft-West CJ, Devlin AJ, Cooper LC, Guimond SE, Procter P, Guerrini M, Miller GJ, Fernig DG, Yates EA, Lima MA, Skidmore MA. Glycosaminoglycans from Litopenaeus vannamei Inhibit the Alzheimer's Disease β Secretase, BACE1. Mar Drugs 2021; 19:203. [PMID: 33916819 PMCID: PMC8067017 DOI: 10.3390/md19040203] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/23/2021] [Accepted: 03/26/2021] [Indexed: 11/16/2022] Open
Abstract
Only palliative therapeutic options exist for the treatment of Alzheimer's Disease; no new successful drug candidates have been developed in over 15 years. The widely used clinical anticoagulant heparin has been reported to exert beneficial effects through multiple pathophysiological pathways involved in the aetiology of Alzheimer's Disease, for example, amyloid peptide production and clearance, tau phosphorylation, inflammation and oxidative stress. Despite the therapeutic potential of heparin as a multi-target drug for Alzheimer's disease, the repurposing of pharmaceutical heparin is proscribed owing to the potent anticoagulant activity of this drug. Here, a heterogenous non-anticoagulant glycosaminoglycan extract, obtained from the shrimp Litopenaeus vannamei, was found to inhibit the key neuronal β-secretase, BACE1, displaying a more favorable therapeutic ratio compared to pharmaceutical heparin when anticoagulant activity is considered.
Collapse
Affiliation(s)
- Courtney J. Mycroft-West
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK; (C.J.M.-W.); (A.J.D.); (L.C.C.); (P.P.); (M.A.L.)
| | - Anthony J. Devlin
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK; (C.J.M.-W.); (A.J.D.); (L.C.C.); (P.P.); (M.A.L.)
| | - Lynsay C. Cooper
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK; (C.J.M.-W.); (A.J.D.); (L.C.C.); (P.P.); (M.A.L.)
| | - Scott E. Guimond
- School of Medicine, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK;
| | - Patricia Procter
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK; (C.J.M.-W.); (A.J.D.); (L.C.C.); (P.P.); (M.A.L.)
| | - Marco Guerrini
- Istituto di Ricerche Chimiche e Biochimiche G. Ronzoni, via G. Colombo 81, 20133 Milan, Italy;
| | - Gavin J. Miller
- School of Chemistry, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK;
| | - David G. Fernig
- Department of Biochemistry and Systems Biology, ISMIB, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK; (D.G.F.); (E.A.Y.)
| | - Edwin A. Yates
- Department of Biochemistry and Systems Biology, ISMIB, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK; (D.G.F.); (E.A.Y.)
| | - Marcelo A. Lima
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK; (C.J.M.-W.); (A.J.D.); (L.C.C.); (P.P.); (M.A.L.)
| | - Mark A. Skidmore
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK; (C.J.M.-W.); (A.J.D.); (L.C.C.); (P.P.); (M.A.L.)
- Department of Biochemistry and Systems Biology, ISMIB, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK; (D.G.F.); (E.A.Y.)
| |
Collapse
|
14
|
Das S, Sengupta S, Chakraborty S. Scope of β-Secretase (BACE1)-Targeted Therapy in Alzheimer's Disease: Emphasizing the Flavonoid Based Natural Scaffold for BACE1 Inhibition. ACS Chem Neurosci 2020; 11:3510-3522. [PMID: 33073981 DOI: 10.1021/acschemneuro.0c00579] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and the most common form of dementia in the world. Studies report the presence of extracellular amyloid plaques consisting of β-amyloid peptide and intracellular tangles consisting of hyperphosphorylated tau proteins as the histopathological indicators of AD. The process of β-amyloid peptide generation by sequential cleavage of amyloid precursor protein by β-secretase (BACE1) and γ-secretase, followed by its aggregation to form amyloid plaques, is the mechanistic basis of the amyloid hypothesis. Other popular hypotheses related to the pathogenesis of AD include the tau hypothesis and the oxidative stress hypothesis. Various targets of the amyloid cascade are now in prime focus to develop drugs for AD. Many BACE1 inhibitors, β-amyloid aggregation inhibitors, and Aβ clearance strategies using monoclonal antibodies are in various stages of clinical trials. This review provides an in-depth evaluation of the role of BACE1 in disease pathogenesis and also highlights the therapeutic approaches developed to find more potent but less toxic inhibitors for BACE1, particularly emphasizing the natural scaffold as a nontoxic lead for BACE1 inhibition. Cellular targets and signaling cascades involving BACE1 have been highlighted to understand the physiological role of BACE1. This knowledge is extremely crucial to understand the toxicity evaluations for BACE1-targeted therapy. We have particularly highlighted the scope of flavonoids as a new generation of nontoxic BACE1 inhibitory scaffolds. The structure-activity relationship of BACE1 inhibition for this group of compounds has been highlighted to provide a guideline to design more selective highly potent inhibitors. The review aims to provide a holistic overview of BACE1-targeted therapy for AD that paves the way for future drug development.
Collapse
Affiliation(s)
- Sucharita Das
- Department of Microbiology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, India
| | - Swaha Sengupta
- Amity Institute of Biotechnology, Amity University, Kolkata 700135, India
| | | |
Collapse
|
15
|
Mycroft-West CJ, Devlin AJ, Cooper LC, Procter P, Miller GJ, Fernig DG, Guerrini M, Guimond SE, Lima MA, Yates EA, Skidmore MA. Inhibition of BACE1, the β-secretase implicated in Alzheimer's disease, by a chondroitin sulfate extract from Sardina pilchardus. Neural Regen Res 2020; 15:1546-1553. [PMID: 31997821 PMCID: PMC7059579 DOI: 10.4103/1673-5374.274341] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 08/23/2019] [Accepted: 10/26/2019] [Indexed: 12/24/2022] Open
Abstract
The pharmaceutical and anticoagulant agent heparin, a member of the glycosaminoglycan family of carbohydrates, has previously been identified as a potent inhibitor of a key Alzheimer's disease drug target, the primary neuronal β-secretase, β-site amyloid precursor protein cleaving enzyme 1 (BACE1). The anticoagulant activity of heparin has, however, precluded the repurposing of this widely used pharmaceutical as an Alzheimer's disease therapeutic. Here, a glycosaminoglycan extract, composed predominantly of 4-sulfated chondroitin sulfate, has been isolated from Sardina pilchardus, which possess the ability to inhibit BACE1 (IC50 [half maximal inhibitory concentration] = 4.8 μg/mL), while displaying highly attenuated anticoagulant activities (activated partial thromboplastin time EC50 [median effective concentration] = 403.8 μg/mL, prothrombin time EC50 = 1.3 mg/mL). The marine-derived, chondroitin sulfate extract destabilizes BACE1, determined via differential scanning fluorimetry (ΔTm -5°C), to a similar extent as heparin, suggesting that BACE1 inhibition by glycosaminoglycans may occur through a common mode of action, which may assist in the screening of glycan-based BACE1 inhibitors for Alzheimer's disease.
Collapse
Affiliation(s)
- Courtney J. Mycroft-West
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire, ST5 5BG, UK
| | - Anthony J. Devlin
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire, ST5 5BG, UK
| | - Lynsay C. Cooper
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire, ST5 5BG, UK
| | - Patricia Procter
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire, ST5 5BG, UK
| | - Gavin J. Miller
- Lennard-Jones Laboratory, School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire, ST5 5BG, UK
| | - David G. Fernig
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | - Marco Guerrini
- Istituto di Ricerche Chimiche e Biochimiche G. Ronzoni, Via G. Colombo 81, 20133 Milan, Italy
| | - Scott E. Guimond
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire, ST5 5BG, UK
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
- School of Medicine, Keele, Staffordshire, ST5 5BG, UK
| | - Marcelo A. Lima
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire, ST5 5BG, UK
| | - Edwin A. Yates
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire, ST5 5BG, UK
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
| | - Mark Andrew Skidmore
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire, ST5 5BG, UK
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK
- School of Medicine, Keele, Staffordshire, ST5 5BG, UK
| |
Collapse
|
16
|
Gupta SP, Patil VM. Recent Studies on Design and Development of Drugs Against Alzheimer's Disease (AD) Based on Inhibition of BACE-1 and Other AD-causative Agents. Curr Top Med Chem 2020; 20:1195-1213. [PMID: 32297584 DOI: 10.2174/1568026620666200416091623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is one of the neurodegenerative diseases and has been hypothesized to be a protein misfolding disease. In the generation of AD, β-secretase, γ-secretase, and tau protein play an important role. A literature search reflects ever increasing interest in the design and development of anti-AD drugs targeting β-secretase, γ-secretase, and tau protein. OBJECTIVE The objective is to explore the structural aspects and role of β-secretase, γ-secretase, and tau protein in AD and the efforts made to exploit them for the design of effective anti-AD drugs. METHODS The manuscript covers the recent studies on design and development of anti-AD drugs exploiting amyloid and cholinergic hypotheses. RESULTS Based on amyloid and cholinergic hypotheses, effective anti-AD drugs have been searched out in which non-peptidic BACE1 inhibitors have been most prominent. CONCLUSION Further exploitation of the structural aspects and the inhibition mechanism for β-secretase, γ-secretase, and tau protein and the use of cholinergic hypothesis may lead still more potent anti-AD drugs.
Collapse
Affiliation(s)
- Satya P Gupta
- Department of Pharmaceutical Technology, Meerut Institute of Engineering and Technology, Meerut-250005, India
| | - Vaishali M Patil
- Computer Aided Drug Design Lab, Department of Pharmaceutical Chemistry, KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad-201206, Uttar Pradesh, India
| |
Collapse
|
17
|
Coimbra JRM, Baptista SJ, Dinis TCP, Silva MMC, Moreira PI, Santos AE, Salvador JAR. Combining Virtual Screening Protocol and In Vitro Evaluation towards the Discovery of BACE1 Inhibitors. Biomolecules 2020; 10:biom10040535. [PMID: 32244832 PMCID: PMC7226079 DOI: 10.3390/biom10040535] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/23/2020] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
The treatment options for a patient diagnosed with Alzheimer’s disease (AD) are currently limited. The cerebral accumulation of amyloid-β (Aβ) is a critical molecular event in the pathogenesis of AD. When the amyloidogenic β-secretase (BACE1) is inhibited, the production of Aβ peptide is reduced. Henceforth, the main goal of this study is the discovery of new small bioactive molecules that potentially reach the brain and inhibit BACE1. The work was conducted by a customized molecular modelling protocol, including pharmacophore-based and molecular docking-based virtual screening (VS). Structure-based (SB) and ligand-based (LB) pharmacophore models were designed to accurately screen several drug-like compound databases. The retrieved hits were subjected to molecular docking and in silico filtered to predict their ability to cross the blood–brain barrier (BBB). Additionally, 34 high-scoring compounds structurally distinct from known BACE1 inhibitors were selected for in vitro screening assay, which resulted in 13 novel hit-compounds for this relevant therapeutic target. This study disclosed new BACE1 inhibitors, proving the utility of combining computational and in vitro approaches for effectively predicting anti-BACE1 agents in the early drug discovery process.
Collapse
Affiliation(s)
- Judite R. M. Coimbra
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (J.R.M.C.); (M.M.C.S.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal; (S.J.B.); (T.C.P.D.); (P.I.M.); (A.E.S.)
| | - Salete J. Baptista
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal; (S.J.B.); (T.C.P.D.); (P.I.M.); (A.E.S.)
- Chem4Pharma, Edifício IPN Incubadora, 3030-199 Coimbra, Portugal
| | - Teresa C. P. Dinis
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal; (S.J.B.); (T.C.P.D.); (P.I.M.); (A.E.S.)
- Laboratory of Biochemistry and Biology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria M. C. Silva
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (J.R.M.C.); (M.M.C.S.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal; (S.J.B.); (T.C.P.D.); (P.I.M.); (A.E.S.)
| | - Paula I. Moreira
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal; (S.J.B.); (T.C.P.D.); (P.I.M.); (A.E.S.)
- Laboratory of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Armanda E. Santos
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal; (S.J.B.); (T.C.P.D.); (P.I.M.); (A.E.S.)
- Laboratory of Biochemistry and Biology, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Jorge A. R. Salvador
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal; (J.R.M.C.); (M.M.C.S.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal; (S.J.B.); (T.C.P.D.); (P.I.M.); (A.E.S.)
- Correspondence: ; Tel.: +351-239-488-479
| |
Collapse
|
18
|
Mycroft-West CJ, Cooper LC, Devlin AJ, Procter P, Guimond SE, Guerrini M, Fernig DG, Lima MA, Yates EA, Skidmore MA. A Glycosaminoglycan Extract from Portunus pelagicus Inhibits BACE1, the β Secretase Implicated in Alzheimer's Disease. Mar Drugs 2019; 17:E293. [PMID: 31100859 PMCID: PMC6562973 DOI: 10.3390/md17050293] [Citation(s) in RCA: 4] [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: 04/18/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/23/2022] Open
Abstract
Therapeutic options for Alzheimer's disease, the most common form of dementia, are currently restricted to palliative treatments. The glycosaminoglycan heparin, widely used as a clinical anticoagulant, has previously been shown to inhibit the Alzheimer's disease-relevant β-secretase 1 (BACE1). Despite this, the deployment of pharmaceutical heparin for the treatment of Alzheimer's disease is largely precluded by its potent anticoagulant activity. Furthermore, ongoing concerns regarding the use of mammalian-sourced heparins, primarily due to prion diseases and religious beliefs hinder the deployment of alternative heparin-based therapeutics. A marine-derived, heparan sulphate-containing glycosaminoglycan extract, isolated from the crab Portunus pelagicus, was identified to inhibit human BACE1 with comparable bioactivity to that of mammalian heparin (IC50 = 1.85 μg mL-1 (R2 = 0.94) and 2.43 μg mL-1 (R2 = 0.93), respectively), while possessing highly attenuated anticoagulant activities. The results from several structural techniques suggest that the interactions between BACE1 and the extract from P. pelagicus are complex and distinct from those of heparin.
Collapse
Affiliation(s)
- Courtney J Mycroft-West
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK.
| | - Lynsay C Cooper
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK.
| | - Anthony J Devlin
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK.
- Istituto di Ricerche Chimiche e Biochimiche G. Ronzoni, Via G. Colombo 81, 20133 Milan, Italy.
| | - Patricia Procter
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK.
| | - Scott E Guimond
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK.
- Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire ST5 5BG, UK.
| | - Marco Guerrini
- Istituto di Ricerche Chimiche e Biochimiche G. Ronzoni, Via G. Colombo 81, 20133 Milan, Italy.
| | - David G Fernig
- School of Biological Sciences, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK.
| | - Marcelo A Lima
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK.
| | - Edwin A Yates
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK.
- School of Biological Sciences, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK.
| | - Mark A Skidmore
- Molecular & Structural Biosciences, School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK.
- Institute for Science and Technology in Medicine, Keele University, Keele, Staffordshire ST5 5BG, UK.
- School of Biological Sciences, University of Liverpool, Crown Street, Liverpool L69 7ZB, UK.
| |
Collapse
|
19
|
Molecular mechanisms underlying protective role of quercetin in attenuating Alzheimer's disease. Life Sci 2019; 224:109-119. [PMID: 30914316 DOI: 10.1016/j.lfs.2019.03.055] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/22/2019] [Accepted: 03/22/2019] [Indexed: 12/17/2022]
Abstract
Quercetin belongs to the flavonoids family, which is present in most of the plants including fruits, vegetables, green tea and even in red wine having antioxidant activities. It is available as a food supplement in the market and has physiological health effects. Quercetin has anti-inflammatory, anticancer and anti-prostate activities along with its beneficial effects on high cholesterol, kidney transplantation, asthma, diabetes, viral infections, pulmonary, schizophrenia and cardiovascular diseases. Quercetin possesses scavenging potential of hydroxyl radical (OH-), hydrogen peroxide (H2O2), and superoxide anion (O2-). These reactive oxygen species (ROS) hampers lipid, protein, amino acids and deoxyribonucleic acid (DNA) processing leading to epigenetic alterations. Quercetin has the ability to combat these harmful effects. ROS plays a vital role in the progression of Alzheimer's disease (AD), and we propose that quercetin would be the best choice to overcome cellular and molecular signals in regulating normal physiological functions. However, data are not well documented regarding exact cellular mechanisms of quercetin. The neuroprotective effects of quercetin are mainly due to potential up- and/or down-regulation of cytokines via nuclear factor (erythroid-derived 2)-like 2 (Nrf2), Paraoxonase-2, c-Jun N-terminal kinase (JNK), Protein kinase C, Mitogen-activated protein kinase (MAPK) signalling cascades, and PI3K/Akt pathways. Therefore, the aim of the present review was to elaborate on the cellular and molecular mechanisms of the quercetin involved in the protection against AD.
Collapse
|
20
|
Coimbra JRM, Marques DFF, Baptista SJ, Pereira CMF, Moreira PI, Dinis TCP, Santos AE, Salvador JAR. Highlights in BACE1 Inhibitors for Alzheimer's Disease Treatment. Front Chem 2018; 6:178. [PMID: 29881722 PMCID: PMC5977085 DOI: 10.3389/fchem.2018.00178] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 05/04/2018] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disorder and the most common type of dementia in the elderly. The clinical symptoms of AD include a progressive loss of memory and impairment of cognitive functions interfering with daily life activities. The main neuropathological features consist in extracellular amyloid-β (Aβ) plaque deposition and intracellular Neurofibrillary tangles (NFTs) of hyperphosphorylated Tau. Understanding the pathophysiological mechanisms that underlie neurodegeneration in AD is essential for rational design of neuroprotective agents able to prevent disease progression. According to the "Amyloid Cascade Hypothesis" the critical molecular event in the pathogenesis of AD is the accumulation of Aβ neurotoxic oligomers. Since the proteolytic processing of Amyloid Precursor Protein (APP) by β-secretase (beta-site APP cleaving enzyme 1, BACE1) is the rate-limiting step in the production of Aβ, this enzyme is considered a major therapeutic target and BACE1 inhibitors have the potential to be disease-modifying drugs for AD treatment. Therefore, intensive efforts to discover and develop inhibitors that can reach the brain and effectively inhibit BACE1 have been pursued by several groups worldwide. The aim of this review is to highlight the progress in the discovery of potent and selective small molecule BACE1 inhibitors over the past decade.
Collapse
Affiliation(s)
- Judite R. M. Coimbra
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of CoimbraCoimbra, Portugal
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
| | - Daniela F. F. Marques
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of CoimbraCoimbra, Portugal
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
| | - Salete J. Baptista
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of CoimbraCoimbra, Portugal
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
- Chem4Pharma, Edifício IPN IncubadoraCoimbra, Portugal
| | - Cláudia M. F. Pereira
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
- Faculty of Medicine, University of CoimbraCoimbra, Portugal
| | - Paula I. Moreira
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
- Laboratory of Physiology, Faculty of Medicine, University of CoimbraCoimbra, Portugal
| | - Teresa C. P. Dinis
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
- Laboratory of Biochemistry, Faculty of Pharmacy, University of CoimbraCoimbra, Portugal
| | - Armanda E. Santos
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
- Laboratory of Biochemistry, Faculty of Pharmacy, University of CoimbraCoimbra, Portugal
| | - Jorge A. R. Salvador
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, University of CoimbraCoimbra, Portugal
- Center for Neuroscience and Cell Biology, University of CoimbraCoimbra, Portugal
| |
Collapse
|
21
|
|
22
|
Li R. Physical activity and prevention of Alzheimer's disease. JOURNAL OF SPORT AND HEALTH SCIENCE 2016; 5:381-382. [PMID: 29067218 PMCID: PMC5651177 DOI: 10.1016/j.jshs.2016.10.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/31/2016] [Indexed: 06/07/2023]
Affiliation(s)
- Rena Li
- Center for Hormone Advanced Science and Education, Roskamp Institute, Sarasota, FL 34243, USA,
| |
Collapse
|