1
|
Jang YJ, Kang SJ, Park HS, Lee DH, Kim JH, Kim JE, Kim DI, Chung CH, Yoon JK, Bhang SH. Drug delivery strategies with lipid-based nanoparticles for Alzheimer's disease treatment. J Nanobiotechnology 2025; 23:99. [PMID: 39930497 PMCID: PMC11809104 DOI: 10.1186/s12951-025-03109-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/11/2025] [Indexed: 02/14/2025] Open
Abstract
Alzheimer's disease (AD) is a distinctive form of dementia characterized by age-related cognitive decline and memory impairment. A key hallmark of AD is the irreversible overaccumulation of beta-amyloid (Aβ) in the brain, associated with neuroinflammation and neuronal death. Although Aβ clearance and immunoregulation have been the major therapeutic strategies for AD, highly selective transport across the blood-brain barrier (BBB) negatively affects the delivery efficacy of the drugs without the ability to cross the BBB. In this review, we discuss the potential of lipid-based nanoparticles (LBNs) as promising vehicles for drug delivery in AD treatment. LBNs, composed of phospholipid mono- or bilayer, have attracted attention due to their exceptional cellular penetration capabilities and drug loading capabilities, which also facilitate cargo transcytosis across the BBB. Recent advances in the development and engineering of LBNs overcome the existing limitations of the current clinical approaches for AD treatment by addressing off-target effects and low therapeutic efficacy. Here, we review the transport pathways across the BBB, as well as various types of LBNs for AD therapy, including exosomes, liposomes, solid lipid nanoparticles (SLNs), and nanostructured lipid carriers (NLCs), to elucidate their distinctive properties, preparation methodologies, and therapeutic efficacy, thereby offering innovative avenues for novel drug development for clinical translation in AD therapy.
Collapse
Affiliation(s)
- Young-Ju Jang
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
| | - Seong-Jun Kang
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, 17546, Gyeonggi-do, Republic of Korea
| | - Hyun Su Park
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
| | - Dong-Hyun Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
| | - Jae Hoon Kim
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, 17546, Gyeonggi-do, Republic of Korea
| | - Ju-El Kim
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, 17546, Gyeonggi-do, Republic of Korea
| | - Dong-Ik Kim
- Division of Vascular Surgery, Samsung Medical Center, School of Medicine, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Chan-Hwa Chung
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, 17546, Gyeonggi-do, Republic of Korea.
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon-si, 16419, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
2
|
Chen C, Wu C, Yang T, Zhao W, Lei J, Lin D. Changing the amyloid nucleation process using small molecules and substrates: a way to build two-dimensional materials. NANOSCALE 2025. [PMID: 39902864 DOI: 10.1039/d4nr04624b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
The assembly of two-dimensional (2D) materials on substrates presents a wide range of potential applications in nanomaterials. However, there is limited information available in the literature regarding the tunable nucleation process in molecular assembly. In this paper, a neurodegenerative disease-related short peptide and a small molecule named Fast Green (FG) were selected for their binding affinity with mica/highly oriented pyrolytic graphite (HOPG) substrates. Based on atomic force microscopy (AFM) and molecular dynamics (MD) simulation, we investigated the control of 2D assemblies. By tuning FG small molecules and substrates, the assemblies grew epitaxially from nanosheets to nanofilms on mica and highly ordered nanofilaments on HOPG substrates. Notably, the nuclei formed an orderly array without a critical size or lag phase in the presence of FG molecules on the HOPG substrate, facilitating a quicker co-assembly of ordered filaments compared to bulk conditions. Our MD simulations further demonstrated that the interaction between Aβ16-22 molecules and the HOPG substrate was primarily due to π-π interactions between aromatic rings, which led to the formation of single-layer filaments by lying on the surface of HOPG. Additionally, parallel π-π stacking acted as the primary force to inhibit the aggregation of peptides into fibrils. Overall, our results provide a strategy for modulating the interaction of amyloid peptides with small molecules and substrates in the assembly of 2D nanomaterials.
Collapse
Affiliation(s)
- Chao Chen
- School of Physical Science and Technology, Ningbo University, 818 Fenghua Road, Ningbo 315211, P. R. China.
| | - Chenyang Wu
- School of Physical Science and Technology, Ningbo University, 818 Fenghua Road, Ningbo 315211, P. R. China.
| | - Tiantian Yang
- School of Physical Science and Technology, Ningbo University, 818 Fenghua Road, Ningbo 315211, P. R. China.
| | - Wenhui Zhao
- School of Physical Science and Technology, Ningbo University, 818 Fenghua Road, Ningbo 315211, P. R. China.
| | - Jiangtao Lei
- School of Physics and Materials Science, Nanchang University, Xuefu Avenue 999, Nanchang City 330031, P. R. China
| | - Dongdong Lin
- School of Physical Science and Technology, Ningbo University, 818 Fenghua Road, Ningbo 315211, P. R. China.
| |
Collapse
|
3
|
Ahanger IA, Dar TA. Small molecule modulators of alpha-synuclein aggregation and toxicity: Pioneering an emerging arsenal against Parkinson's disease. Ageing Res Rev 2024; 101:102538. [PMID: 39389237 DOI: 10.1016/j.arr.2024.102538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
Parkinson's disease (PD) is primarily characterized by loss of dopaminergic neurons in the substantia nigra pars compacta region of the brain and accumulation of aggregated forms of alpha-synuclein (α-Syn), an intrinsically disordered protein, in the form of Lewy Bodies and Lewy Neurites. Substantial evidences point to the aggregated/fibrillar forms of α-Syn as a central event in PD pathogenesis, underscoring the modulation of α-Syn aggregation as a promising strategy for PD treatment. Consequently, numerous anti-aggregation agents, spanning from small molecules to polymers, have been scrutinized for their potential to mitigate α-Syn aggregation and its associated toxicity. Among these, small molecule modulators like osmoprotectants, polyphenols, cellular metabolites, metals, and peptides have emerged as promising candidates with significant potential in PD management. This article offers a comprehensive overview of the effects of these small molecule modulators on the aggregation propensity and associated toxicity of α-Syn and its PD-associated mutants. It serves as a valuable resource for identifying and developing potent, non-invasive, non-toxic, and highly specific small molecule-based therapeutic arsenal for combating PD. Additionally, it raises pertinent questions aimed at guiding future research endeavours in the field of α-Syn aggregation remodelling.
Collapse
Affiliation(s)
- Ishfaq Ahmad Ahanger
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India.
| | - Tanveer Ali Dar
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India.
| |
Collapse
|
4
|
Fan J, Liu X, Wang Z, Cui N, Zhang Y, Zhang Y, Song J, Li T, Wang Y. Roseibium algae sp. nov., isolated from a marine alga ( Grateloupia sp.). Int J Syst Evol Microbiol 2024; 74. [PMID: 39073406 DOI: 10.1099/ijsem.0.006475] [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: 07/30/2024] Open
Abstract
A novel Gram-stain-negative, rod-shaped, non-spore-forming, aerobic, motile bacterium with a single polar or subpolar flagellum, designated strain H3510T, was isolated from marine alga collected on sea shore of Yantai, PR China. The organism grew optimally at 28 °C and pH 7.0 and in presence of 3.0 % (w/v) NaCl. The strain exhibited positive catalase activity but negative oxidase and nitrate reduction activities. The predominant cellular fatty acids were C18 : 1 ω7c and/or C18 : 1 ω6c, 11-methyl C18 : 1 ω7c, and C16 : 0. Additionally, the major polar lipids were phosphatidylglycerol, phosphatidylmonomethylethanolamine, diphosphatidylglycerol, and phosphatidylethanolamine; the respiratory quinone was ubiquinone 10 (Q-10). The genomic DNA G+C content of strain H3510T was 54.2%. The novel strain showed the closest relationship with Roseibium polysiphoniae KMM 9699T with 98.2 % 16S rRNA gene sequence similarity. The calculated values for average nucleotide identity and DNA-DNA hybridization between strain H3510T and the phylogenetically related Roseibium species were in the range of 71.3-74.9 % and 13.7-19.9 %, respectively. Based on polyphasic analyses, strain H3510T was identified as representing a novel species of the genus Roseibium, for which the name Roseibium algae sp. nov. is proposed. The type strain is H3510T (=KCTC 8206T=MCCC 1K04325T). The heterologously expressed inositol 2-dehydrogenase gene from strain H3510T displayed high oxidation activity on myo-inositol and showed potential in the production of rare stereoisomers of inositol, such as scyllo-inositol.
Collapse
Affiliation(s)
- Jiwu Fan
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, 601 Jinsui Avenue, Xinxiang 453003, PR China
| | - Xinqi Liu
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, 601 Jinsui Avenue, Xinxiang 453003, PR China
| | - Ziwei Wang
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, 601 Jinsui Avenue, Xinxiang 453003, PR China
| | - Ning Cui
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, 601 Jinsui Avenue, Xinxiang 453003, PR China
| | - Yao Zhang
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, 601 Jinsui Avenue, Xinxiang 453003, PR China
| | - Yanfeng Zhang
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, 601 Jinsui Avenue, Xinxiang 453003, PR China
| | - Jiale Song
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, 601 Jinsui Avenue, Xinxiang 453003, PR China
| | - Tao Li
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, 601 Jinsui Avenue, Xinxiang 453003, PR China
| | - Yan Wang
- Synthetic Biology Engineering Lab of Henan Province, School of Life Sciences and Technology, Xinxiang Medical University, 601 Jinsui Avenue, Xinxiang 453003, PR China
| |
Collapse
|
5
|
Odorskaya MV, Mavletova DA, Nesterov AA, Tikhonova OV, Soloveva NA, Reznikova DA, Galanova OO, Vatlin AA, Slynko NM, Vasilieva AR, Peltek SE, Danilenko VN. The use of omics technologies in creating LBP and postbiotics based on the Limosilactobacillus fermentum U-21. Front Microbiol 2024; 15:1416688. [PMID: 38919499 PMCID: PMC11197932 DOI: 10.3389/fmicb.2024.1416688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
In recent years, there has been an increasing tendency to create drugs based on certain commensal bacteria of the human microbiota and their ingredients, primarily focusing on live biotherapeutics (LBPs) and postbiotics. The creation of such drugs, termed pharmacobiotics, necessitates an understanding of their mechanisms of action and the identification of pharmacologically active ingredients that determine their target properties. Typically, these are complexes of biologically active substances synthesized by specific strains, promoted as LBPs or postbiotics (including vesicles): proteins, enzymes, low molecular weight metabolites, small RNAs, etc. This study employs omics technologies, including genomics, proteomics, and metabolomics, to explore the potential of Limosilactobacillus fermentum U-21 for innovative LBP and postbiotic formulations targeting neuroinflammatory processes. Proteomic techniques identified and quantified proteins expressed by L. fermentum U-21, highlighting their functional attributes and potential applications. Key identified proteins include ATP-dependent Clp protease (ClpL), chaperone protein DnaK, protein GrpE, thioredoxin reductase, LysM peptidoglycan-binding domain-containing protein, and NlpC/P60 domain-containing protein, which have roles in disaggregase, antioxidant, and immunomodulatory activities. Metabolomic analysis provided insights into small-molecule metabolites produced during fermentation, revealing compounds with anti-neuroinflammatory activity. Significant metabolites produced by L. fermentum U-21 include GABA (γ-aminobutyric acid), niacin, aucubin, and scyllo-inositol. GABA was found to stabilize neuronal activity, potentially counteracting neurodegenerative processes. Niacin, essential for optimal nervous system function, was detected in vesicles and culture fluid, and it modulates cytokine production, maintaining immune homeostasis. Aucubin, an iridoid glycoside usually secreted by plants, was identified as having antioxidant properties, addressing issues of bioavailability for therapeutic use. Scyllo-inositol, identified in vesicles, acts as a chemical chaperone, reducing abnormal protein clumps linked to neurodegenerative diseases. These findings demonstrate the capability of L. fermentum U-21 to produce bioactive substances that could be harnessed in the development of pharmacobiotics for neurodegenerative diseases, contributing to their immunomodulatory, anti-neuroinflammatory, and neuromodulatory activities. Data of the HPLC-MS/MS analysis are available via ProteomeXchange with identifier PXD050857.
Collapse
Affiliation(s)
- Maya V. Odorskaya
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics Russian Academy of Sciences, Moscow, Russia
| | - Dilara A. Mavletova
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics Russian Academy of Sciences, Moscow, Russia
| | - Andrey A. Nesterov
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics Russian Academy of Sciences, Moscow, Russia
- Institute of Environmental Engineering, RUDN University, Moscow, Russia
| | | | | | - Diana A. Reznikova
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics Russian Academy of Sciences, Moscow, Russia
- Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Olesya O. Galanova
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics Russian Academy of Sciences, Moscow, Russia
- Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Aleksey A. Vatlin
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics Russian Academy of Sciences, Moscow, Russia
| | - Nikolai M. Slynko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Asya R. Vasilieva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Sergey E. Peltek
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Valery N. Danilenko
- Laboratory of Bacterial Genetics, Vavilov Institute of General Genetics Russian Academy of Sciences, Moscow, Russia
- Brain Science Institute, Research Center of Neurology, Moscow, Russia
| |
Collapse
|
6
|
Yoshida KI, Bott M. Microbial synthesis of health-promoting inositols. Curr Opin Biotechnol 2024; 87:103114. [PMID: 38520822 DOI: 10.1016/j.copbio.2024.103114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/25/2024]
Abstract
D-chiro-inositol and scyllo-inositol are known for their health-promoting properties and promising as ingredients for functional foods. Strains of Bacillus subtilis and Corynebacterium glutamicum were created by metabolic engineering capable of inexpensive production of these two rare inositols from myo-inositol, which is the most common inositol in nature. In addition, further modifications have enabled the synthesis of the two rare inositols from the much-cheaper carbon sources, glucose or sucrose.
Collapse
Affiliation(s)
- Ken-Ichi Yoshida
- Graduate School of Science, Technology and Innovation, University of Kobe, Kobe, Japan.
| | - Michael Bott
- Institute of Bio- and Geosciences, IBG-1: Biotechnology, Forschungszentrum Jülich, Jülich, Germany.
| |
Collapse
|
7
|
Tandon S, Aggarwal P, Sarkar S. Polyglutamine disorders: Pathogenesis and potential drug interventions. Life Sci 2024; 344:122562. [PMID: 38492921 DOI: 10.1016/j.lfs.2024.122562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/27/2024] [Accepted: 03/13/2024] [Indexed: 03/18/2024]
Abstract
Polyglutamine/poly(Q) diseases are a group nine hereditary neurodegenerative disorders caused due to abnormally expanded stretches of CAG trinucleotide in functionally distinct genes. All human poly(Q) diseases are characterized by the formation of microscopically discernable poly(Q) positive aggregates, the inclusion bodies. These toxic inclusion bodies are responsible for the impairment of several cellular pathways such as autophagy, transcription, cell death, etc., that culminate in disease manifestation. Although, these diseases remain largely without treatment, extensive research has generated mounting evidences that various events of poly(Q) pathogenesis can be developed as potential drug targets. The present review article briefly discusses the key events of disease pathogenesis, model system-based investigations that support the development of effective therapeutic interventions against pathogenesis of human poly(Q) disorders, and a comprehensive list of pharmacological and bioactive compounds that have been experimentally shown to alleviate poly(Q)-mediated neurotoxicity. Interestingly, due to the common cause of pathogenesis, all poly(Q) diseases share etiology, thus, findings from one disease can be potentially extrapolated to other poly(Q) diseases as well.
Collapse
Affiliation(s)
- Shweta Tandon
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| | - Prerna Aggarwal
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India
| | - Surajit Sarkar
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110021, India.
| |
Collapse
|
8
|
Monteiro KLC, de Aquino TM, da Silva-Júnior EF. Natural Compounds as Inhibitors of Aβ Peptide and Tau Aggregation. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:1234-1250. [PMID: 38018200 DOI: 10.2174/0118715273273539231114095300] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/08/2023] [Accepted: 10/16/2023] [Indexed: 11/30/2023]
Abstract
Neurodegenerative conditions like Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS) encompass disorders characterized by the degeneration of neurons in specific circumstances. The quest for novel agents to influence these diseases, particularly AD, has unearthed various natural compounds displaying multifaceted activities and diverse pharmacological mechanisms. Given the ongoing extensive study of pathways associated with the accumulation of neurofibrillary aggregates and amyloid plaques, this paper aims to comprehensively review around 130 studies exploring natural products. These studies focus on inhibiting the formation of amyloid plaques and tau protein tangles, with the objective of potentially alleviating or delaying AD.
Collapse
Affiliation(s)
- Kadja Luana Chagas Monteiro
- Research Group on Therapeutic Strategies - GPET, Laboratory of Synthesis and Research in Medicinal Chemistry - LSPMED, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Alagoas, Brazil
| | - Thiago Mendonça de Aquino
- Research Group on Therapeutic Strategies - GPET, Laboratory of Synthesis and Research in Medicinal Chemistry - LSPMED, Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Alagoas, Brazil
| | | |
Collapse
|
9
|
Fanton S, Menezes J, Krock E, Sandström A, Tour J, Sandor K, Jurczak A, Hunt M, Baharpoor A, Kadetoff D, Jensen KB, Fransson P, Ellerbrock I, Sitnikov R, Svensson CI, Kosek E. Anti-satellite glia cell IgG antibodies in fibromyalgia patients are related to symptom severity and to metabolite concentrations in thalamus and rostral anterior cingulate cortex. Brain Behav Immun 2023; 114:371-382. [PMID: 37683961 DOI: 10.1016/j.bbi.2023.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 08/29/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023] Open
Abstract
Recent translational work has shown that fibromyalgia might be an autoimmune condition with pathogenic mechanisms mediated by a peripheral, pain-inducing action of immunoglobulin G (IgG) antibodies binding to satellite glia cells (SGC) in the dorsal root ganglia. A first clinical assessment of the postulated autoimmunity showed that fibromyalgia subjects (FMS) had elevated levels of antibodies against SGC (termed anti-SGC IgG) compared to healthy controls and that anti-SGC IgG were associated with a more severe disease status. The overarching aim of the current study was to determine whether the role of anti-SGC IgG in driving pain is exclusively through peripheral mechanisms, as indirectly shown so far, or could be attributed also to central mechanisms. To this end, we wanted to first confirm, in a larger cohort of FMS, the relation between anti-SGC IgG and pain-related clinical measures. Secondly, we explored the associations of these autoantibodies with brain metabolite concentrations (assessed via magnetic resonance spectroscopy, MRS) and pressure-evoked cerebral pain processing (assessed via functional magnetic resonance imaging, fMRI) in FMS. Proton MRS was performed in the thalamus and rostral anterior cingulate cortex (rACC) of FMS and concentrations of a wide spectrum of metabolites were assessed. During fMRI, FMS received individually calibrated painful pressure stimuli corresponding to low and high pain intensities. Our results confirmed a positive correlation between anti-SGC IgG and clinical measures assessing condition severity. Additionally, FMS with high anti-SGC IgG levels had higher pain intensity and a worse disease status than FMS with low anti-SGC IgG levels. Further, anti-SGC IgG levels negatively correlated with metabolites such as scyllo-inositol in thalamus and rACC as well as with total choline and macromolecule 12 in thalamus, thus linking anti-SGC IgG levels to the concentration of metabolites in the brain of FMS. However, anti-SGC IgG levels in FMS were not associated with the sensitivity to pressure pain or the cerebral processing of evoked pressure pain. Taken together, our results suggest that anti-SGC IgG might be clinically relevant for spontaneous, non-evoked pain. Our current and previous translational and clinical findings could provide a rationale to try new antibody-related treatments in FMS.
Collapse
Affiliation(s)
- Silvia Fanton
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden.
| | - Joana Menezes
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Emerson Krock
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden; Faculty of Dental Medicine and Oral Health Sciences, Alan Edwards Centre for Research on Pain, McGill University, Montreal, Canada
| | - Angelica Sandström
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden; Department of Radiology, Massachusetts General Hospital, A.A. Martinos Center for Biomedical Imaging, Harvard Medical School, Boston, MA, USA
| | - Jeanette Tour
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Katalin Sandor
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Alexandra Jurczak
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Matthew Hunt
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Azar Baharpoor
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Diana Kadetoff
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden; Stockholm Spine Center, Löwenströmska Hospital, Upplands Väsby, Sweden
| | - Karin B Jensen
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Fransson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Isabel Ellerbrock
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden
| | - Rouslan Sitnikov
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden; MRI Research Center, Karolinska University Hospital, Stockholm, Sweden
| | - Camilla I Svensson
- Department of Physiology and Pharmacology, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Eva Kosek
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Neuroradiology, Karolinska University Hospital, Stockholm, Sweden; Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
10
|
Cutuli D, Petrosini L, Gelfo F. Advance in Neurotoxicity Research from Development to Aging. Int J Mol Sci 2023; 24:15112. [PMID: 37894793 PMCID: PMC10606676 DOI: 10.3390/ijms242015112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
A substance capable of inducing a consistent pattern of neural dysfunction in the chemistry or structure of the nervous system may be defined as neurotoxic [...].
Collapse
Affiliation(s)
- Debora Cutuli
- IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
- Department of Psychology, Sapienza University of Rome, Via dei Marsi 78, 00185 Rome, Italy
| | - Laura Petrosini
- IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
| | - Francesca Gelfo
- IRCCS Fondazione Santa Lucia, Via Ardeatina 306, 00179 Rome, Italy
- Department of Human Sciences, Guglielmo Marconi University, Via Plinio 44, 00193 Rome, Italy
| |
Collapse
|
11
|
Basli A, Bounaas J. Pathophysiological mechanism and natural preventive and therapeutic strategies of Alzheimer's disease. Nutr Health 2023; 29:403-413. [PMID: 36377316 DOI: 10.1177/02601060221137104] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Alzheimer's disease (AD) is characterized by the presence of two types of protein deposits in the brain, amyloid plaques and neurofibrillary tangles. The first one are dense deposits of beta amyloid protein, the second one are dense deposits of the protein tau. These proteins are present in all of our brains, but in AD they act unusually, leading to neuronal degeneration. This review will provide an overview of the AD, including the role of amyloid beta and tau, and mechanisms that lead to the formation of plaques and tangles. The review will also cover the existing researches that have focused on the inhibition of amyloid beta formation, cholinesterase, tau hyperphosphorylation, the pathogenic mechanisms of apoE4, and GSK-3 as a solution that could be used to slow or prevent the disease.
Collapse
Affiliation(s)
- Abdelkader Basli
- Laboratory of Interaction Research, Biodiversity, Ecosystems and Biotechnology, Faculty of Sciences, University of Skikda, Skikda, Algeria
| | - Jihane Bounaas
- Laboratory of Interaction Research, Biodiversity, Ecosystems and Biotechnology, Faculty of Sciences, University of Skikda, Skikda, Algeria
| |
Collapse
|
12
|
Sanchez-Rodriguez D, Gonzalez-Figueroa I, Alvarez-Berríos MP. Chaperone Activity and Protective Effect against Aβ-Induced Cytotoxicity of Artocarpus camansi Blanco and Amaranthus dubius Mart. ex Thell Seed Protein Extracts. Pharmaceuticals (Basel) 2023; 16:820. [PMID: 37375767 DOI: 10.3390/ph16060820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/29/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia and is listed as the sixth-leading cause of death in the United States. Recent findings have linked AD to the aggregation of amyloid beta peptides (Aβ), a proteolytic fragment of 39-43 amino acid residues derived from the amyloid precursor protein. AD has no cure; thus, new therapies to stop the progression of this deadly disease are constantly being searched for. In recent years, chaperone-based medications from medicinal plants have gained significant interest as an anti-AD therapy. Chaperones are responsible for maintaining the three-dimensional shape of proteins and play an important role against neurotoxicity induced by the aggregation of misfolded proteins. Therefore, we hypothesized that proteins extracted from the seeds of Artocarpus camansi Blanco (A. camansi) and Amaranthus dubius Mart. ex Thell (A. dubius) could possess chaperone activity and consequently may exhibit a protective effect against Aβ1-40-induced cytotoxicity. To test this hypothesis, the chaperone activity of these protein extracts was measured using the enzymatic reaction of citrate synthase (CS) under stress conditions. Then, their ability to inhibit the aggregation of Aβ1-40 using a thioflavin T (ThT) fluorescence assay and DLS measurements was determined. Finally, the neuroprotective effect against Aβ1-40 in SH-SY5Y neuroblastoma cells was evaluated. Our results demonstrated that A. camansi and A. dubius protein extracts exhibited chaperone activity and inhibited Aβ1-40 fibril formation, with A. dubius showing the highest chaperone activity and inhibition at the concentration assessed. Additionally, both protein extracts showed neuroprotective effects against Aβ1-40-induced toxicity. Overall, our data demonstrated that the plant-based proteins studied in this research work can effectively overcome one of the most important characteristics of AD.
Collapse
Affiliation(s)
- David Sanchez-Rodriguez
- Department of Science and Technology, Inter American University of Puerto Rico at Ponce, Ponce, PR 00715-1602, USA
| | - Idsa Gonzalez-Figueroa
- Department of Science and Technology, Inter American University of Puerto Rico at Ponce, Ponce, PR 00715-1602, USA
| | - Merlis P Alvarez-Berríos
- Department of Science and Technology, Inter American University of Puerto Rico at Ponce, Ponce, PR 00715-1602, USA
| |
Collapse
|
13
|
Andrade S, Nunes D, Dabur M, Ramalho MJ, Pereira MC, Loureiro JA. Therapeutic Potential of Natural Compounds in Neurodegenerative Diseases: Insights from Clinical Trials. Pharmaceutics 2023; 15:pharmaceutics15010212. [PMID: 36678841 PMCID: PMC9860553 DOI: 10.3390/pharmaceutics15010212] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/01/2023] [Accepted: 01/04/2023] [Indexed: 01/10/2023] Open
Abstract
Neurodegenerative diseases are caused by the gradual loss of neurons' function. These neurological illnesses remain incurable, and current medicines only alleviate the symptoms. Given the social and economic burden caused by the rising frequency of neurodegenerative diseases, there is an urgent need for the development of appropriate therapeutics. Natural compounds are gaining popularity as alternatives to synthetic drugs due to their neuroprotective properties and higher biocompatibility. While natural compounds' therapeutic effects for neurodegenerative disease treatment have been investigated in numerous in vitro and in vivo studies, only few have moved to clinical trials. This article provides the first systematic review of the clinical trials evaluating natural compounds' safety and efficacy for the treatment of the five most prevalent neurodegenerative disorders: Alzheimer's disease, Parkinson's disease, multiple sclerosis, amyotrophic lateral sclerosis, and Huntington's disease.
Collapse
Affiliation(s)
- Stéphanie Andrade
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Débora Nunes
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Meghna Dabur
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Maria J. Ramalho
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Maria C. Pereira
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- Correspondence: (M.C.P.); (J.A.L.)
| | - Joana A. Loureiro
- LEPABE—Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- Correspondence: (M.C.P.); (J.A.L.)
| |
Collapse
|
14
|
Wu Y, Kawabata H, Kita K, Ishikawa S, Tanaka K, Yoshida KI. Constitutive glucose dehydrogenase elevates intracellular NADPH levels and luciferase luminescence in Bacillus subtilis. Microb Cell Fact 2022; 21:266. [PMID: 36539761 PMCID: PMC9768902 DOI: 10.1186/s12934-022-01993-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Genetic modifications in Bacillus subtilis have allowed the conversion of myo-inositol into scyllo-inositol, which is proposed as a therapeutic agent for Alzheimer's disease. This conversion comprises two reactions catalyzed by two distinct inositol dehydrogenases, IolG and IolW. The IolW-mediated reaction requires the intracellular regeneration of NADPH, and there appears to be a limit to the endogenous supply of NADPH, which may be one of the rate-determining factors for the conversion of inositol. The primary mechanism of NADPH regeneration in this bacterium remains unclear. RESULTS The gdh gene of B. subtilis encodes a sporulation-specific glucose dehydrogenase that can use NADP+ as a cofactor. When gdh was modified to be constitutively expressed, the intracellular NADPH level was elevated, increasing the conversion of inositol. In addition, the bacterial luciferase derived from Photorhabdus luminescens became more luminescent in cells in liquid culture and colonies on culture plates. CONCLUSION The results indicated that the luminescence of luciferase was representative of intracellular NADPH levels. Luciferase can therefore be employed to screen for mutations in genes involved in NADPH regeneration in B. subtilis, and artificial manipulation to enhance NADPH regeneration can promote the production of substances such as scyllo-inositol.
Collapse
Affiliation(s)
- Yuzheng Wu
- grid.31432.370000 0001 1092 3077Department of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe, 657 8501 Japan
| | - Honami Kawabata
- grid.31432.370000 0001 1092 3077Department of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe, 657 8501 Japan
| | - Kyosuke Kita
- grid.31432.370000 0001 1092 3077Department of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe, 657 8501 Japan
| | - Shu Ishikawa
- grid.31432.370000 0001 1092 3077Department of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe, 657 8501 Japan
| | - Kan Tanaka
- grid.32197.3e0000 0001 2179 2105Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, Tokyo, Japan ,grid.419082.60000 0004 1754 9200Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Tokyo, Japan
| | - Ken-ichi Yoshida
- grid.31432.370000 0001 1092 3077Department of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe, 657 8501 Japan
| |
Collapse
|
15
|
Almeida ZL, Brito RMM. Amyloid Disassembly: What Can We Learn from Chaperones? Biomedicines 2022; 10:3276. [PMID: 36552032 PMCID: PMC9776232 DOI: 10.3390/biomedicines10123276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 12/23/2022] Open
Abstract
Protein aggregation and subsequent accumulation of insoluble amyloid fibrils with cross-β structure is an intrinsic characteristic of amyloid diseases, i.e., amyloidoses. Amyloid formation involves a series of on-pathway and off-pathway protein aggregation events, leading to mature insoluble fibrils that eventually accumulate in multiple tissues. In this cascade of events, soluble oligomeric species are formed, which are among the most cytotoxic molecular entities along the amyloid cascade. The direct or indirect action of these amyloid soluble oligomers and amyloid protofibrils and fibrils in several tissues and organs lead to cell death in some cases and organ disfunction in general. There are dozens of different proteins and peptides causing multiple amyloid pathologies, chief among them Alzheimer's, Parkinson's, Huntington's, and several other neurodegenerative diseases. Amyloid fibril disassembly is among the disease-modifying therapeutic strategies being pursued to overcome amyloid pathologies. The clearance of preformed amyloids and consequently the arresting of the progression of organ deterioration may increase patient survival and quality of life. In this review, we compiled from the literature many examples of chemical and biochemical agents able to disaggregate preformed amyloids, which have been classified as molecular chaperones, chemical chaperones, and pharmacological chaperones. We focused on their mode of action, chemical structure, interactions with the fibrillar structures, morphology and toxicity of the disaggregation products, and the potential use of disaggregation agents as a treatment option in amyloidosis.
Collapse
Affiliation(s)
| | - Rui M. M. Brito
- Chemistry Department and Coimbra Chemistry Centre—Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
16
|
Near-Infrared Photothermally Enhanced Photo-Oxygenation for Inhibition of Amyloid-β Aggregation Based on RVG-Conjugated Porphyrinic Metal-Organic Framework and Indocyanine Green Nanoplatform. Int J Mol Sci 2022; 23:ijms231810885. [PMID: 36142796 PMCID: PMC9505608 DOI: 10.3390/ijms231810885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/08/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Amyloid aggregation is associated with many neurodegenerative diseases such as Alzheimer's disease (AD). The current technologies using phototherapy for amyloid inhibition are usually photodynamic approaches based on evidence that reactive oxygen species can inhibit Aβ aggregation. Herein, we report a novel combinational photothermally assisted photo-oxygenation treatment based on a nano-platform of the brain-targeting peptide RVG conjugated with the 2D porphyrinic PCN-222 metal-organic framework and indocyanine green (PCN-222@ICG@RVG) with enhanced photo-inhibition in Alzheimer's Aβ aggregation. A photothermally assisted photo-oxygenation treatment based on PCN@ICG could largely enhance the photo-inhibition effect on Aβ42 aggregation and lead to much lower neurotoxicity upon near-infrared (NIR) irradiation at 808 nm compared with a single modality of photo-treatment in both cell-free and in vitro experiments. Generally, local photothermal heat increases the instability of Aβ aggregates and keeps Aβ in the status of monomers, which facilitates the photo-oxygenation process of generating oxidized Aβ monomers with low aggregation capability. In addition, combined with the brain-targeting peptide RVG, the PCN-222@ICG@RVG nanoprobe shows high permeability of the human blood-brain barrier (BBB) on a human brain-on-a-chip platform. The ex vivo study also demonstrates that NIR-activated PCN-222@ICG@RVG could efficiently dissemble Aβ plaques. Our work suggests that the combination of photothermal treatment with photo-oxygenation can synergistically enhance the inhibition of Aβ aggregation, which may boost NIR-based combinational phototherapy of AD in the future.
Collapse
|
17
|
Tyler SEB, Tyler LDK. Therapeutic roles of plants for 15 hypothesised causal bases of Alzheimer's disease. NATURAL PRODUCTS AND BIOPROSPECTING 2022; 12:34. [PMID: 35996065 PMCID: PMC9395556 DOI: 10.1007/s13659-022-00354-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/15/2022] [Indexed: 05/26/2023]
Abstract
Alzheimer's disease (AD) is progressive and ultimately fatal, with current drugs failing to reverse and cure it. This study aimed to find plant species which may provide therapeutic bioactivities targeted to causal agents proposed to be driving AD. A novel toolkit methodology was employed, whereby clinical symptoms were translated into categories recognized in ethnomedicine. These categories were applied to find plant species with therapeutic effects, mined from ethnomedical surveys. Survey locations were mapped to assess how this data is at risk. Bioactivities were found of therapeutic relevance to 15 hypothesised causal bases for AD. 107 species with an ethnological report of memory improvement demonstrated therapeutic activity for all these 15 causal bases. The majority of the surveys were found to reside within biodiversity hotspots (centres of high biodiversity under threat), with loss of traditional knowledge the most common threat. Our findings suggest that the documented plants provide a large resource of AD therapeutic potential. In demonstrating bioactivities targeted to these causal bases, such plants may have the capacity to reduce or reverse AD, with promise as drug leads to target multiple AD hallmarks. However, there is a need to preserve ethnomedical knowledge, and the habitats on which this knowledge depends.
Collapse
Affiliation(s)
| | - Luke D K Tyler
- School of Natural Sciences, Bangor University, Gwynedd, UK
| |
Collapse
|
18
|
Carder HM, Wang Y, Wendlandt AE. Selective Axial-to-Equatorial Epimerization of Carbohydrates. J Am Chem Soc 2022; 144:11870-11877. [PMID: 35731921 DOI: 10.1021/jacs.2c04743] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Radical-mediated transformations have emerged as powerful methods for the synthesis of rare and unnatural branched, deoxygenated, and isomeric sugars. Here, we describe a radical-mediated axial-to-equatorial alcohol epimerization method to transform abundant glycans into rare isomers. The method delivers highly predictable and selective reaction outcomes that are complementary to other sugar isomerization methods. The synthetic utility of isomer interconversion is showcased through expedient glycan synthesis, including one-step glycodiversification. Mechanistic studies reveal that both site- and diastereoselectivities are achieved by highly selective H atom abstraction of equatorially disposed α-hydroxy C-H bonds.
Collapse
Affiliation(s)
- Hayden M Carder
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Yong Wang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Alison E Wendlandt
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
19
|
Xiao M, Xiang W, Chen Y, Peng N, Du X, Lu S, Zuo Y, Li B, Hu Y, Li X. DHA Ameliorates Cognitive Ability, Reduces Amyloid Deposition, and Nerve Fiber Production in Alzheimer's Disease. Front Nutr 2022; 9:852433. [PMID: 35782939 PMCID: PMC9240638 DOI: 10.3389/fnut.2022.852433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Background The etiology of Alzheimer's disease (AD) is very complex. Docosahexaenoic acid (DHA) is important in cognitive ability and nervous system development. A limited number of studies have evaluated the efficacy of DHA in the treatment of AD. Introduction We detected neurofibrillary tangles (NFT) in the hippocampus and cortex of transgenic mice brain through silver glycine staining. We determined the activity of neurons by staining Nissl bodies, used liquid NMR to detect metabolites in the brain, and functional magnetic resonance imaging results to observe the connection signal value between brain regions. Materials and Methods We fed 3-month-old APP/PS1 double transgenic mice with DHA mixed feeds for 4 months to assess the effects of DHA on cognitive ability in AD mice through the Morris water maze and open field tests. To evaluate its effects with AD pathology, continuous feeding was done until the mice reached 9 months of age. Results Compared to AD mice, escape latency significantly decreased on the fifth day while swimming speed, target quadrant stay time, and the crossing number of platforms increased by varying degrees after DHA treatment. Brain tissue section staining revealed that DHA significantly reduced Aβ and nerve fibers in the brain of AD mice. Conclusion DHA significantly reduced the deposition of Aβ in the brain and inhibited the production of nerve fibers, thereby increasing cognitive abilities in AD mice. In addition, DHA suppressed blood lipid levels, and restored uric acid and urea levels, implying that DHA is a potential therapeutic option for early AD.
Collapse
Affiliation(s)
- Min Xiao
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
- CABIO Biotech (Wuhan) Co., Ltd., Wuhan, China
| | - Wei Xiang
- CABIO Biotech (Wuhan) Co., Ltd., Wuhan, China
| | - Yashu Chen
- Key Laboratory of Oil Crop Biology and Genetic Breeding, Oil Crops Research Institute, Ministry of Agriculture, Chinese Academy of Agricultural Sciences, Wuhan, China
| | - Nan Peng
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiubo Du
- Shenzhen Key Laboratory of Microbial Genetic Engineering, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Shuhuan Lu
- CABIO Biotech (Wuhan) Co., Ltd., Wuhan, China
| | - Yao Zuo
- CABIO Biotech (Wuhan) Co., Ltd., Wuhan, China
| | - Boling Li
- CABIO Biotech (Wuhan) Co., Ltd., Wuhan, China
| | - Yonggang Hu
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Xiangyu Li
- CABIO Biotech (Wuhan) Co., Ltd., Wuhan, China
| |
Collapse
|
20
|
Xiao M, Yao C, Liu F, Xiang W, Zuo Y, Feng K, Lu S, Xiang L, Li M, Li X, Du X. Sialic Acid Ameliorates Cognitive Deficits by Reducing Amyloid Deposition, Nerve Fiber Production, and Neuronal Apoptosis in a Mice Model of Alzheimer's Disease. NEUROSCI 2022; 3:28-40. [PMID: 39484667 PMCID: PMC11523747 DOI: 10.3390/neurosci3010002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 12/22/2021] [Indexed: 11/03/2024] Open
Abstract
(1) Background: As a natural carbohydrate, sialic acid (SA) is helpful for brain development, cognitive ability, and the nervous system, but there are few reports about the effect of SA on Alzheimer's disease (AD). (2) Method: The present study evaluated the effect of SA on cognitive ability, neuronal activity, Aβ formation, and tau hyperphosphorylation in a double transgenic AD (2×Tg-AD) mice model. The 2×Tg-AD mice were randomly divided into four groups: the AD control group, 17 mg/kg SA-treated AD group, 84 mg/kg SA-treated AD group, and 420 mg/kg SA-treated AD group. Mice from all four groups were fed to 7 months of age for the behavioral test and to 9 months of age for the pathological factors investigation. (3) Results: In the Morris water maze, the escape latency significantly decreased on the fifth day in the SA-treated groups. The number of rearing and crossing times in the open field test also increased significantly, compared with the control group. SA treatment significantly reduced amyloid β-peptide (Aβ) and nerve fibers and increased the number of Nissl bodies in the brain of AD mice. (4) Conclusions: SA reduced the neuron damage by reducing Aβ and inhibited tau protein hyperphosphorylation, which improved the cognitive ability and mobility of AD mice.
Collapse
Affiliation(s)
- Min Xiao
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Chuangyu Yao
- Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China;
| | - Fang Liu
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Wei Xiang
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Yao Zuo
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Kejue Feng
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Shuhuan Lu
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Li Xiang
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Muzi Li
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Xiangyu Li
- R&D Center, Cabio Biotech (Wuhan) Co., Ltd., No. 999 Gaoxin Rd., Wuhan 430073, China; (M.X.); (F.L.); (W.X.); (Y.Z.); (K.F.); (S.L.); (L.X.); (M.L.)
| | - Xiubo Du
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
21
|
La Manna S, Leone M, Iacobucci I, Annuziata A, Di Natale C, Lagreca E, Malfitano AM, Ruffo F, Merlino A, Monti M, Marasco D. Glucosyl Platinum(II) Complexes Inhibit Aggregation of the C-Terminal Region of the Aβ Peptide. Inorg Chem 2022; 61:3540-3552. [PMID: 35171608 PMCID: PMC9951207 DOI: 10.1021/acs.inorgchem.1c03540] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Neurodegenerative diseases are often caused by uncontrolled amyloid aggregation. Hence, many drug discovery processes are oriented to evaluate new compounds that are able to modulate self-recognition mechanisms. Herein, two related glycoconjugate pentacoordinate Pt(II) complexes were analyzed in their capacity to affect the self-aggregation processes of two amyloidogenic fragments, Aβ21-40 and Aβ25-35, of the C-terminal region of the β-amyloid (Aβ) peptide, the major component of Alzheimer's disease (AD) neuronal plaques. The most water-soluble complex, 1Ptdep, is able to bind both fragments and to deeply influence the morphology of peptide aggregates. Thioflavin T (ThT) binding assays, electrospray ionization mass spectrometry (ESI-MS), and ultraviolet-visible (UV-vis) absorption spectroscopy indicated that 1Ptdep shows different kinetics and mechanisms of inhibition toward the two sequences and demonstrated that the peptide aggregation inhibition is associated with a direct coordinative bond of the compound metal center to the peptides. These data support the in vitro ability of pentacoordinate Pt(II) complexes to inhibit the formation of amyloid aggregates and pave the way for the application of this class of compounds as potential neurotherapeutics.
Collapse
Affiliation(s)
- Sara La Manna
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
| | - Marilisa Leone
- Institute
of Biostructures and Bioimaging - CNR, 80134 Naples, Italy
| | - Ilaria Iacobucci
- Department
of Chemical Sciences, University of Naples
“Federico II”, 80126 Naples, Italy
- CEINGE
Biotecnologie Avanzate S.c.a r.l., “University
of Naples Federico II”, 80131 Naples, Italy
| | - Alfonso Annuziata
- Department
of Chemical Sciences, University of Naples
“Federico II”, 80126 Naples, Italy
| | - Concetta Di Natale
- Interdisciplinary
Research Centre on Biomaterials (CRIB), Department of Ingegneria Chimica
del Materiali e della Produzione Industriale (DICMAPI), University “Federico II”, 80125 Naples, Italy
| | - Elena Lagreca
- Interdisciplinary
Research Centre on Biomaterials (CRIB), Department of Ingegneria Chimica
del Materiali e della Produzione Industriale (DICMAPI), University “Federico II”, 80125 Naples, Italy
| | - Anna Maria Malfitano
- Department
of Translational Medical Science, University
of Naples “Federico II”, 80131 Naples, Italy
| | - Francesco Ruffo
- Department
of Chemical Sciences, University of Naples
“Federico II”, 80126 Naples, Italy
| | - Antonello Merlino
- Department
of Chemical Sciences, University of Naples
“Federico II”, 80126 Naples, Italy
| | - Maria Monti
- Department
of Chemical Sciences, University of Naples
“Federico II”, 80126 Naples, Italy
- CEINGE
Biotecnologie Avanzate S.c.a r.l., “University
of Naples Federico II”, 80131 Naples, Italy
| | - Daniela Marasco
- Department
of Pharmacy, University of Naples “Federico
II”, 80131 Naples, Italy
- Institute
of Biostructures and Bioimaging - CNR, 80134 Naples, Italy
| |
Collapse
|
22
|
Siracusa L, Napoli E, Ruberto G. Novel Chemical and Biological Insights of Inositol Derivatives in Mediterranean Plants. Molecules 2022; 27:1525. [PMID: 35268625 PMCID: PMC8912080 DOI: 10.3390/molecules27051525] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
Inositols (Ins) are natural compounds largely widespread in plants and animals. Bio-sinthetically they derive from sugars, possessing a molecular structure very similar to the simple sugars, and this aspect concurs to define them as primary metabolites, even though it is much more correct to place them at the boundary between primary and secondary metabolites. This dichotomy is well represented by the fact that as primary metabolites they are essential cellular components in the form of phospholipid derivatives, while as secondary metabolites they are involved in a plethora of signaling pathways playing an important role in the surviving of living organisms. myo-Inositol is the most important and widespread compound of this family, it derives directly from d-glucose, and all known inositols, including stereoisomers and derivatives, are the results of metabolic processes on this unique molecule. In this review, we report the new insights of these compounds and their derivatives concerning their occurrence in Nature with a particular emphasis on the plant of the Mediterranean area, as well as the new developments about their biological effectiveness.
Collapse
Affiliation(s)
| | | | - Giuseppe Ruberto
- Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Via Paolo Gaifami 18, 95126 Catania, Italy; (L.S.); (E.N.)
| |
Collapse
|
23
|
Wu B, Bagshaw AP, Hickey C, Kühn S, Wilson M. Evidence for distinct neuro-metabolic phenotypes in humans. Neuroimage 2022; 249:118902. [PMID: 35033676 DOI: 10.1016/j.neuroimage.2022.118902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 01/05/2022] [Accepted: 01/09/2022] [Indexed: 11/17/2022] Open
Abstract
Advances in magnetic resonance imaging have shown how individual differences in the structure and function of the human brain relate to health and cognition. The relationship between individual differences and the levels of neuro-metabolites, however, remains largely unexplored - despite the potential for the discovery of novel behavioural and disease phenotypes. In this study, we measured 14 metabolite levels, normalised as ratios to total-creatine, with 1H magnetic resonance spectroscopy (MRS) acquired from the bilateral anterior cingulate cortices of six healthy participants, repeatedly over a period of four months. ANOVA tests revealed statistically significant differences of 3 metabolites and 3 commonly used combinations (total-choline, glutamate + glutamine and total-N-acetylaspartate) between the participants, with scyllo-inositol (F=85, p=6e-26) and total-choline (F=39, p=1e-17) having the greatest discriminatory power. This was not attributable to structural differences. When predicting individuals from the repeated MRS measurements, a leave-one-out classification accuracy of 88% was achieved using a support vector machine based on scyllo-inositol and total-choline levels. Accuracy increased to 98% with the addition of total-N-acetylaspartate and myo-inositol - demonstrating the efficacy of combining MRS with machine learning and metabolomic methodology. These results provide evidence for the existence of neuro-metabolic phenotypes, which may be non-invasively measured using widely available 3 Tesla MRS. Establishing these phenotypes in a larger cohort and investigating their connection to brain health and function presents an important area for future study.
Collapse
Affiliation(s)
- Bofan Wu
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham, UK
| | - Andrew P Bagshaw
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham, UK
| | - Clayton Hickey
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham, UK
| | - Simone Kühn
- Department of Psychiatry and Psychotherapy, Neuronal Plasticity Working Group, University Medical Center Hamburg-Eppendorf, Germany; Max Planck Institute for Human Development, Lise Meitner Group for Environmental Neuroscience, Germany
| | - Martin Wilson
- Centre for Human Brain Health and School of Psychology, University of Birmingham, Birmingham, UK.
| |
Collapse
|
24
|
An SSA, Shim KH, Kang S, Kim YK, Subedi L, Cho H, Hong SM, Tan MA, Jeon R, Chang KA, Kim SY. The potential anti-amyloidogenic candidate, SPA1413, for Alzheimer's disease. Br J Pharmacol 2021; 179:1033-1048. [PMID: 34610141 DOI: 10.1111/bph.15691] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 08/23/2021] [Accepted: 09/02/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND PURPOSE Recently, isoflavone derivatives have been shown to have neuroprotective effects against neurological disorders. For instance, genistein attenuated the neuroinflammation and amyloid-β accumulation in Alzheimer's disease animal models, suggesting the potential for use to prevent and treat Alzheimer's disease. EXPERIMENTAL APPROACH Here, 50 compounds, including isoflavone derivatives, were constructed and screened for the inhibitory effects on amyloid-β42 fibrilization and oligomerization using the high-throughput screening formats of thioflavin T assay and multimer detection system, respectively. The potential neuroprotective effect of t3-(4-hydroxyphenyl)-2H-chromen-7-ol (SPA1413), also known as dehydroequol, idronoxil or phenoxodiol, was evaluated in cells and in 5xFAD (B6SJL) transgenic mouse, a model of Alzheimer's disease. KEY RESULTS SPA1413 had a potent inhibitory action on both amyloid-β fibrilization and oligomerization. In the cellular assay, SPA1413 prevented amyloid-β-induced cytotoxicity and reduced neuroinflammation. Remarkably, the oral administration of SPA1413 ameliorated cognitive impairment, decreased amyloid-β plaques and activated microglia in the brain of 5xFAD (B6SJL) transgenic mouse. CONCLUSION AND IMPLICATIONS Our results strongly support the repurposing of SPA1413, which has already received fast-track status from the US Food and Drug Administration (FDA) for cancer treatment, for the treatment of Alzheimer's disease due to its potent anti-amyloidogenic and anti-neuroinflammatory actions.
Collapse
Affiliation(s)
- Seong Soo A An
- Department of Bionano Technology, Gachon University, Gyeonggi-do, Republic of Korea
| | - Kyu Hwan Shim
- Department of Bionano Technology, Gachon University, Gyeonggi-do, Republic of Korea.,Department of Neurology, Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul, Republic of Korea
| | - Shinwoo Kang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea.,Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea
| | - Young Kyo Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Lalita Subedi
- Department of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Hyewon Cho
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Seong-Min Hong
- Department of Pharmacy, Gachon University, Incheon, Republic of Korea
| | - Mario A Tan
- Department of Bionano Technology, Gachon University, Gyeonggi-do, Republic of Korea.,College of Science, University of Santo Tomas, Manila, Philippines
| | - Raok Jeon
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Sookmyung Women's University, Seoul, Republic of Korea
| | - Keun-A Chang
- Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea.,Neuroscience Research Institute, Gachon University, Incheon, Republic of Korea.,Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Sun Yeou Kim
- Department of Pharmacy, Gachon University, Incheon, Republic of Korea
| |
Collapse
|
25
|
Liu Y, Cong L, Han C, Li B, Dai R. Recent Progress in the Drug Development for the Treatment of Alzheimer's Disease Especially on Inhibition of Amyloid-peptide Aggregation. Mini Rev Med Chem 2021; 21:969-990. [PMID: 33245270 DOI: 10.2174/1389557520666201127104539] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/25/2020] [Accepted: 09/14/2020] [Indexed: 11/22/2022]
Abstract
As the world 's population is aging, Alzheimer's disease (AD) has become a big concern since AD has started affecting younger people and the population of AD patients is increasing worldwide. It has been revealed that the neuropathological hallmarks of AD are typically characterized by the presence of neurotoxic extracellular amyloid plaques in the brain, which are surrounded by tangles of neuronal fibers. However, the causes of AD have not been completely understood yet. Currently, there is no drug to effectively prevent AD or to completely reserve the symptoms in the patients. This article reviews the pathological features associated with AD, the recent progress in research on the drug development to treat AD, especially on the discovery of natural product derivatives to inhibit Aβ peptide aggregation as well as the design and synthesis of Aβ peptide aggregation inhibitors to treat AD.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Lin Cong
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, 10081, China
| | - Chu Han
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Bo Li
- Advanced Research Institute of Multidisciplinary Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Rongji Dai
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing, 10081, China
| |
Collapse
|
26
|
Ramp P, Lehnert A, Matamouros S, Wirtz A, Baumgart M, Bott M. Metabolic engineering of Corynebacterium glutamicum for production of scyllo-inositol, a drug candidate against Alzheimer's disease. Metab Eng 2021; 67:173-185. [PMID: 34224896 DOI: 10.1016/j.ymben.2021.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/17/2021] [Accepted: 06/30/2021] [Indexed: 10/20/2022]
Abstract
Scyllo-inositol has been identified as a potential drug for the treatment of Alzheimer's disease. Therefore, cost-efficient processes for the production of this compound are desirable. In this study, we analyzed and engineered Corynebacterium glutamicum with the aim to develop competitive scyllo-inositol producer strains. Initial studies revealed that C. glutamicum naturally produces scyllo-inositol when cultured with myo-inositol as carbon source. The conversion involves NAD+-dependent oxidation of myo-inositol to 2-keto-myo-inositol followed by NADPH-dependent reduction to scyllo-inositol. Use of myo-inositol for biomass formation was prevented by deletion of a cluster of 16 genes involved in myo-inositol catabolism (strain MB001(DE3)Δiol1). Deletion of a second cluster of four genes (oxiC-cg3390-oxiD-oxiE) related to inositol metabolism prevented conversion of 2-keto-myo-inositol to undesired products causing brown coloration (strain MB001(DE3)Δiol1Δiol2). The two chassis strains were used for plasmid-based overproduction of myo-inositol dehydrogenase (IolG) and scyllo-inositol dehydrogenase (IolW). In BHI medium containing glucose and myo-inositol, a complete conversion of the consumed myo-inositol into scyllo-inositol was achieved with the Δiol1Δiol2 strain. To enable scyllo-inositol production from cheap carbon sources, myo-inositol 1-phosphate synthase (Ino1) and myo-inositol 1-phosphatase (ImpA), which convert glucose 6-phosphate into myo-inositol, were overproduced in addition to IolG and IolW using plasmid pSI. Strain MB001(DE3)Δiol1Δiol2 (pSI) produced 1.8 g/L scyllo-inositol from 20 g/L glucose and even 4.4 g/L scyllo-inositol from 20 g/L sucrose within 72 h. Our results demonstrate that C. glutamicum is an attractive host for the biotechnological production of scyllo-inositol and potentially further myo-inositol-derived products.
Collapse
Affiliation(s)
- Paul Ramp
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Alexander Lehnert
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Susana Matamouros
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Astrid Wirtz
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Meike Baumgart
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany
| | - Michael Bott
- IBG-1: Biotechnology, Institute of Bio- and Geosciences, Forschungszentrum Jülich, Jülich, Germany.
| |
Collapse
|
27
|
Jani V, Sonavane U, Joshi R. Destabilization potential of beta sheet breaker peptides on Abeta fibril structure: an insight from molecular dynamics simulation study. RSC Adv 2021; 11:23557-23573. [PMID: 35479797 PMCID: PMC9036544 DOI: 10.1039/d1ra03609b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 06/29/2021] [Indexed: 02/02/2023] Open
Abstract
Alzheimer's disease is characterized by amyloid-β aggregation. Currently, all the approved medications are to treat the symptoms but there is no clinically approved treatment for the cure or to prevent the progression of Alzheimer's disease (AD). Earlier reports suggest the use of small molecules and peptides to target and destabilize the amyloid fibril. The use of Beta Sheet Breaker (BSB) peptides seems to be a promising and attractive therapeutic approach as it can strongly bind and destabilize the preformed amyloid fibril. There are experimental studies describing the destabilization role of various BSB peptides, but the exact mechanism remains elusive. In the current work, an attempt is made to study the destabilization mechanism of different BSB peptides on preformed amyloid protofibril using molecular docking and simulations. Molecular docking of eight different BSB peptides of varying length (5-mer to 10-mer) on the Abeta protofibril was done. Docking was followed by multiple sets of molecular simulations for the Abeta protofibril–BSB peptide complex for each of the top ranked poses of the eight BSB peptides. As a control, multiple sets of simulations for the Abeta protofibril (APO) were also carried out. An increase in the RMSD, decrease in the number of interchain hydrogen bonds, destabilization of important salt bridge interactions (D23–K28), and destabilization of interchain hydrophobic interactions suggested the destabilization of Abeta protofibril by BSB peptides. The MM-GBSA free energy of binding for each of the BSB peptides was calculated to measure the binding affinity of BSB peptides to Abeta protofibril. Further residue wise contribution of free energy of binding was also calculated. The study showed that 7-mer peptides tend to bind strongly to Abeta protofibril as compared to other BSB peptides. The KKLVFFA peptide showed better destabilization potential as compared to the other BSB peptides. The details about the destabilization mechanism of BSB peptides will help in the design of other peptides for the therapeutic intervention for AD. Destabilzation of Abeta protofibril by Beta Sheet Breaker (BSB) peptides.![]()
Collapse
Affiliation(s)
- Vinod Jani
- Centre for Development of Advanced Computing (C-DAC) Panchavati, Pashan Pune India
| | - Uddhavesh Sonavane
- Centre for Development of Advanced Computing (C-DAC) Panchavati, Pashan Pune India
| | - Rajendra Joshi
- Centre for Development of Advanced Computing (C-DAC) Panchavati, Pashan Pune India
| |
Collapse
|
28
|
Wang K, Na L, Duan M. The Pathogenesis Mechanism, Structure Properties, Potential Drugs and Therapeutic Nanoparticles against the Small Oligomers of Amyloid-β. Curr Top Med Chem 2021; 21:151-167. [PMID: 32938351 DOI: 10.2174/1568026620666200916123000] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/02/2020] [Accepted: 08/13/2020] [Indexed: 12/27/2022]
Abstract
Alzheimer's Disease (AD) is a devastating neurodegenerative disease that affects millions of people in the world. The abnormal aggregation of amyloid β protein (Aβ) is regarded as the key event in AD onset. Meanwhile, the Aβ oligomers are believed to be the most toxic species of Aβ. Recent studies show that the Aβ dimers, which are the smallest form of Aβ oligomers, also have the neurotoxicity in the absence of other oligomers in physiological conditions. In this review, we focus on the pathogenesis, structure and potential therapeutic molecules against small Aβ oligomers, as well as the nanoparticles (NPs) in the treatment of AD. In this review, we firstly focus on the pathogenic mechanism of Aβ oligomers, especially the Aβ dimers. The toxicity of Aβ dimer or oligomers, which attributes to the interactions with various receptors and the disruption of membrane or intracellular environments, were introduced. Then the structure properties of Aβ dimers and oligomers are summarized. Although some structural information such as the secondary structure content is characterized by experimental technologies, detailed structures are still absent. Following that, the small molecules targeting Aβ dimers or oligomers are collected; nevertheless, all of these ligands have failed to come into the market due to the rising controversy of the Aβ-related "amyloid cascade hypothesis". At last, the recent progress about the nanoparticles as the potential drugs or the drug delivery for the Aβ oligomers are present.
Collapse
Affiliation(s)
- Ke Wang
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| | - Liu Na
- School of Biological and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Mojie Duan
- Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Chinese Academy of Sciences, Wuhan 430071, China
| |
Collapse
|
29
|
Araújo AR, Castro VI, Reis RL, Pires RA. Glucosamine and Its Analogues as Modulators of Amyloid-β Toxicity. ACS Med Chem Lett 2021; 12:548-554. [PMID: 33859794 PMCID: PMC8040036 DOI: 10.1021/acsmedchemlett.0c00350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 03/17/2021] [Indexed: 01/04/2023] Open
Abstract
In Alzheimer's disease (AD), amyloid-β (Aβ) oligomers are considered key mediators of synaptic dysfunction and cognitive impairment. These unstable intermediate Aβ species can interfere with different cellular organelles, leading to neuronal cell death, through the formation of Ca2+-permeable membrane pores, impairment in the levels of acetylcholine neurotransmitters, increased insulin resistance, promotion of pro-inflammatory cascades, among others. Based on a series of evidences that indicate the key role of glycosaminoglycans (GAGs) in amyloid plaque formation, we evaluated the capacity of four monosaccharides, i.e., glucosamine (GlcN), N-acetyl glucosamine (GlcNAc), glucosamine-6-sulfate (GlcN6S), and glucosamine-6-phosphate (GlcN6P), to reduce the Aβ-mediated pathological hallmarks. The tested monosaccharides, in particular, GlcN6S and GlcN6P, were able to interact with Aβ aggregates, reducing neuronal cell death, Aβ-mediated damage to the cellular membrane, acetylcholinesterase activity, insulin resistance, and pro-inflammation levels.
Collapse
Affiliation(s)
- Ana R. Araújo
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials,
Biodegradables and Biomimetics, University
of Minho, Headquarters of the European
Institute of Excellence on Tissue Engineering and Regenerative Medicine,
AvePark, Parque de Ciência e Tecnologia, Zona Industrial da
Gandra, 4805-017 Barco, Portugal
- ICVS/3B’s
− PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Vânia I.
B. Castro
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials,
Biodegradables and Biomimetics, University
of Minho, Headquarters of the European
Institute of Excellence on Tissue Engineering and Regenerative Medicine,
AvePark, Parque de Ciência e Tecnologia, Zona Industrial da
Gandra, 4805-017 Barco, Portugal
- ICVS/3B’s
− PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Rui L. Reis
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials,
Biodegradables and Biomimetics, University
of Minho, Headquarters of the European
Institute of Excellence on Tissue Engineering and Regenerative Medicine,
AvePark, Parque de Ciência e Tecnologia, Zona Industrial da
Gandra, 4805-017 Barco, Portugal
- ICVS/3B’s
− PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Ricardo A. Pires
- 3B’s
Research Group, I3Bs − Research Institute on Biomaterials,
Biodegradables and Biomimetics, University
of Minho, Headquarters of the European
Institute of Excellence on Tissue Engineering and Regenerative Medicine,
AvePark, Parque de Ciência e Tecnologia, Zona Industrial da
Gandra, 4805-017 Barco, Portugal
- ICVS/3B’s
− PT Government Associate Laboratory, 4806-909 Braga/Guimarães, Portugal
| |
Collapse
|
30
|
Mueed Z, Mehta D, Rai PK, Kamal MA, Poddar NK. Cross-Interplay between Osmolytes and mTOR in Alzheimer's Disease Pathogenesis. Curr Pharm Des 2021; 26:4699-4711. [PMID: 32418522 DOI: 10.2174/1381612826666200518112355] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease, categorized by the piling of amyloid-β (Aβ), hyperphosphorylated tau, PHFs, NFTs and mTOR hyperactivity, is a neurodegenerative disorder, affecting people across the globe. Osmolytes are known for osmoprotectants and play a pivotal role in protein folding, function and protein stability, thus, preventing proteins aggregation, and counteracting effects of denaturing solutes on proteins. Osmolytes (viz., sorbitol, inositol, and betaine) perform a pivotal function of maintaining homeostasis during hyperosmotic stress. The selective advantage of utilising osmolytes over inorganic ions by cells is in maintaining cell volume without compromising cell function, which is important for organs such as the brain. Osmolytes have been documented not only as neuroprotectors but they also seem to act as neurodegenerators. Betaine, sucrose and trehalose supplementation has been seen to induce autophagy thereby inhibiting the accumulation of Aβ. In contrast, sucrose has also been associated with mTOR hyperactivity, a hallmark of AD pathology. The neuroprotective action of taurine is revealed when taurine supplementation is seen to inhibit neural damage, apoptosis and oxidative damage. Inositol stereoisomers (viz., scyllo-inositol and myo-inositol) have also been seen to inhibit Aβ production and plaque formation in the brain, inhibiting AD pathogenesis. However, TMAO affects the aging process adversely by deregulating the mTOR signalling pathway and then kindling cognitive dysfunction via degradation of chemical synapses and synaptic plasticity. Thus, it can be concluded that osmolytes may act as a probable therapeutic approach for neurodevelopmental disorders. Here, we have reviewed and focussed upon the impact of osmolytes on mTOR signalling pathway and thereby its role in AD pathogenesis.
Collapse
Affiliation(s)
- Zeba Mueed
- Department of Biotechnology, Invertis University, Bareilly, Uttar Pradesh, India
| | - Devanshu Mehta
- Kusuma School of Biological Sciences, Indian Institute of Technology, Delhi, India
| | - Pankaj K Rai
- Department of Biotechnology, Invertis University, Bareilly, Uttar Pradesh, India
| | - Mohammad A Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia,Enzymoics; Novel Global Community Educational Foundation, 7 Peterlee Place, Hebersham, NSW 2770, Australia
| | - Nitesh K Poddar
- Department of Biosciences, Manipal University Jaipur, Rajasthan, India
| |
Collapse
|
31
|
Yanckello LM, Hoffman JD, Chang YH, Lin P, Nehra G, Chlipala G, McCulloch SD, Hammond TC, Yackzan AT, Lane AN, Green SJ, Hartz AMS, Lin AL. Apolipoprotein E genotype-dependent nutrigenetic effects to prebiotic inulin for modulating systemic metabolism and neuroprotection in mice via gut-brain axis. Nutr Neurosci 2021; 25:1669-1679. [PMID: 33666538 PMCID: PMC8799401 DOI: 10.1080/1028415x.2021.1889452] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The goal of the study was to identify the potential nutrigenetic effects to inulin, a prebiotic fiber, in mice with different human apolipoprotein E (APOE) genetic variants. Specifically, we compared responses to inulin for the potential modulation of the systemic metabolism and neuroprotection via gut-brain axis in mice with human APOE ϵ3 and ϵ4 alleles. METHOD We performed experiments with young mice expressing the human APOE3 (E3FAD mice and APOE4 gene (E4FAD mice). We fed mice with either inulin or control diet for 16 weeks starting from 3 months of age. We determined gut microbiome diversity and composition using16s rRNA sequencing, systemic metabolism using in vivo MRI and metabolomics, and blood-brain barrier (BBB) tight junction expression using Western blot. RESULTS In both E3FAD and E4FAD mice, inulin altered the alpha and beta diversity of the gut microbiome, increased beneficial taxa of bacteria and elevated cecal short chain fatty acid and hippocampal scyllo-inositol. E3FAD mice had altered metabolism related to tryptophan and tyrosine, while E4FAD mice had changes in the tricarboxylic acid cycle, pentose phosphate pathway, and bile acids. Differences were found in levels of brain metabolites related to oxidative stress, and levels of Claudin-1 and Claudin-5 BBB tight junction expression. DISCUSSION We found that inulin had many similar beneficial effects in the gut and brain for both E3FAD and E4FAD mice, which may be protective for brain functions and reduce risk for neurodegeneration. . E3FAD and E4FAD mice also had distinct responses in several metabolic pathways, suggesting an APOE-dependent nutrigenetic effects in modulating systemic metabolism and neuroprotection.
Collapse
Affiliation(s)
- Lucille M Yanckello
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA.,Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
| | - Jared D Hoffman
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA.,Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
| | - Ya-Hsuan Chang
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA.,Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
| | - Penghui Lin
- Center for Environmental Systems Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Geetika Nehra
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - George Chlipala
- Research Resources Center, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | - Tyler C Hammond
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA.,Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA
| | - Andrew T Yackzan
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Andrew N Lane
- Center for Environmental Systems Biochemistry, University of Kentucky, Lexington, Kentucky, USA.,Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky, USA.,Markey Cancer Center, University of Kentucky, Lexington, Kentucky, USA
| | - Stefan J Green
- Research Resources Center, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA.,Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA
| | - Ai-Ling Lin
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA.,Department of Pharmacology and Nutritional Science, University of Kentucky, Lexington, Kentucky, USA.,Department of Neuroscience, University of Kentucky, Lexington, Kentucky, USA.,F. Joseph Halcomb III, M.D. Department of Biomedical Engineering, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
32
|
Minakawa EN, Nagai Y. Protein Aggregation Inhibitors as Disease-Modifying Therapies for Polyglutamine Diseases. Front Neurosci 2021; 15:621996. [PMID: 33642983 PMCID: PMC7907447 DOI: 10.3389/fnins.2021.621996] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/18/2021] [Indexed: 12/11/2022] Open
Abstract
The polyglutamine (polyQ) diseases are a group of inherited neurodegenerative diseases caused by the abnormal expansion of a CAG trinucleotide repeat that are translated into an expanded polyQ stretch in the disease-causative proteins. The expanded polyQ stretch itself plays a critical disease-causative role in the pathomechanisms underlying polyQ diseases. Notably, the expanded polyQ stretch undergoes a conformational transition from the native monomer into the β-sheet-rich monomer, followed by the formation of soluble oligomers and then insoluble aggregates with amyloid fibrillar structures. The intermediate soluble species including the β-sheet-rich monomer and oligomers exhibit substantial neurotoxicity. Therefore, protein conformation stabilization and aggregation inhibition that target the upstream of the insoluble aggregate formation would be a promising approach toward the development of disease-modifying therapies for polyQ diseases. PolyQ aggregation inhibitors of different chemical categories, such as intrabodies, peptides, and small chemical compounds, have been identified through intensive screening methods. Among them, recent advances in the brain delivery methods of several peptides and the screening of small chemical compounds have brought them closer to clinical utility. Notably, the recent discovery of arginine as a potent conformation stabilizer and aggregation inhibitor of polyQ proteins both in vitro and in vivo have paved way to the clinical trial for the patients with polyQ diseases. Meanwhile, expression reduction of expanded polyQ proteins per se would be another promising approach toward disease modification of polyQ diseases. Gene silencing, especially by antisense oligonucleotides (ASOs), have succeeded in reducing the expression of polyQ proteins in the animal models of various polyQ diseases by targeting the aberrant mRNA with expanded CAG repeats. Of note, some of these ASOs have recently been translated into clinical trials. Here we overview and discuss these recent advances toward the development of disease modifying therapies for polyQ diseases. We envision that combination therapies using aggregation inhibitors and gene silencing would meet the needs of the patients with polyQ diseases and their caregivers in the near future to delay or prevent the onset and progression of these currently intractable diseases.
Collapse
Affiliation(s)
- Eiko N Minakawa
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Yoshitaka Nagai
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan.,Department of Neurotherapeutics, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
33
|
Mahmoudinobar F, Nilsson BL, Dias CL. Effects of Ions and Small Compounds on the Structure of Aβ 42 Monomers. J Phys Chem B 2021; 125:1085-1097. [PMID: 33481611 DOI: 10.1021/acs.jpcb.0c09617] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Aggregation of amyloid-β (Aβ) proteins in the brain is a hallmark of Alzheimer's disease. This phenomenon can be promoted or inhibited by adding small molecules to the solution where Aβ is embedded. These molecules affect the ensemble of conformations sampled by Aβ monomers even before aggregation starts. Here, we perform extensive all-atom replica exchange molecular dynamics (REMD) simulations to provide a comparative study of the ensemble of conformations sampled by Aβ42 monomers in solutions that promote (i.e., aqueous solution containing NaCl) and inhibit (i.e., aqueous solutions containing scyllo-inositol or 4-aminophenol) aggregation. Simulations performed in pure water are used as our reference. We find that secondary-structure content is only affected in an antagonistic manner by promoters and inhibitors at the C-terminus and the central hydrophilic core. Moreover, the end of the C-terminus binds more favorably to the central hydrophobic core region of Aβ42 in NaCl adopting a type of strand-loop-strand structure that is disfavored by inhibitors. Nonpolar residues that form the dry core of larger aggregates of Aβ42 (e.g., PDB ID 2BEG) are found at close proximity in these strand-loop-strand structures, suggesting that their formation could play an important role in initiating nucleation. In the presence of inhibitors, the C-terminus binds the central hydrophilic core with a higher probability than in our reference simulation. This sensitivity of the C-terminus, which is affected in an antagonistic manner by inhibitors and promoters, provides evidence for its critical role in accounting for aggregation.
Collapse
Affiliation(s)
- Farbod Mahmoudinobar
- Department of Physics, New Jersey Institute of Technology, Newark, New Jersey 07102-1982, United States
| | - Bradley L Nilsson
- Department of Chemistry, University of Rochester, Rochester, New York 14627, United States
| | - Cristiano L Dias
- Department of Physics, New Jersey Institute of Technology, Newark, New Jersey 07102-1982, United States
| |
Collapse
|
34
|
Minakawa EN, Popiel HA, Tada M, Takahashi T, Yamane H, Saitoh Y, Takahashi Y, Ozawa D, Takeda A, Takeuchi T, Okamoto Y, Yamamoto K, Suzuki M, Fujita H, Ito C, Yagihara H, Saito Y, Watase K, Adachi H, Katsuno M, Mochizuki H, Shiraki K, Sobue G, Toda T, Wada K, Onodera O, Nagai Y. Arginine is a disease modifier for polyQ disease models that stabilizes polyQ protein conformation. Brain 2021; 143:1811-1825. [PMID: 32436573 DOI: 10.1093/brain/awaa115] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 01/12/2020] [Accepted: 02/23/2020] [Indexed: 12/15/2022] Open
Abstract
The polyglutamine (polyQ) diseases are a group of inherited neurodegenerative diseases that include Huntington's disease, various spinocerebellar ataxias, spinal and bulbar muscular atrophy, and dentatorubral pallidoluysian atrophy. They are caused by the abnormal expansion of a CAG repeat coding for the polyQ stretch in the causative gene of each disease. The expanded polyQ stretches trigger abnormal β-sheet conformational transition and oligomerization followed by aggregation of the polyQ proteins in the affected neurons, leading to neuronal toxicity and neurodegeneration. Disease-modifying therapies that attenuate both symptoms and molecular pathogenesis of polyQ diseases remain an unmet clinical need. Here we identified arginine, a chemical chaperone that facilitates proper protein folding, as a novel compound that targets the upstream processes of polyQ protein aggregation by stabilizing the polyQ protein conformation. We first screened representative chemical chaperones using an in vitro polyQ aggregation assay, and identified arginine as a potent polyQ aggregation inhibitor. Our in vitro and cellular assays revealed that arginine exerts its anti-aggregation property by inhibiting the toxic β-sheet conformational transition and oligomerization of polyQ proteins before the formation of insoluble aggregates. Arginine exhibited therapeutic effects on neurological symptoms and protein aggregation pathology in Caenorhabditis elegans, Drosophila, and two different mouse models of polyQ diseases. Arginine was also effective in a polyQ mouse model when administered after symptom onset. As arginine has been safely used for urea cycle defects and for mitochondrial myopathy, encephalopathy, lactic acid and stroke syndrome patients, and efficiently crosses the blood-brain barrier, a drug-repositioning approach for arginine would enable prompt clinical application as a promising disease-modifier drug for the polyQ diseases.
Collapse
Affiliation(s)
- Eiko N Minakawa
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Helena Akiko Popiel
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Division of Clinical Genetics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masayoshi Tada
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Niigata, Japan
| | - Toshiaki Takahashi
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Niigata, Japan
| | - Hiroshi Yamane
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Yuji Saitoh
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | | | - Daisaku Ozawa
- Department of Neurotherapeutics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Akiko Takeda
- Department of Neurotherapeutics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Toshihide Takeuchi
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Department of Neurotherapeutics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuma Okamoto
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Division of Clinical Genetics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kazuhiro Yamamoto
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Mari Suzuki
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Department of Neurotherapeutics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiromi Fujita
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Chiyomi Ito
- Division of Clinical Genetics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Hiroko Yagihara
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Yuko Saito
- Department of Pathology and Laboratory Medicine, National Center Hospital, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Kei Watase
- Center for Brain Integration Research, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Hiroaki Adachi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Hideki Mochizuki
- Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kentaro Shiraki
- Faculty of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Tatsushi Toda
- Division of Clinical Genetics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Keiji Wada
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, Niigata, Niigata, Japan
| | - Yoshitaka Nagai
- Department of Degenerative Neurological Diseases, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Division of Clinical Genetics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Department of Neurotherapeutics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Department of Neurology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
35
|
Engineering Bacillus subtilis Cells as Factories: Enzyme Secretion and Value-added Chemical Production. BIOTECHNOL BIOPROC E 2020. [DOI: 10.1007/s12257-020-0104-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
36
|
Yoshida KI, Shirae Y, Nishimura R, Fukui K, Ishikawa S. Identification of a repressor for the two iol operons required for inositol catabolism in Geobacillus kaustophilus. MICROBIOLOGY-SGM 2020; 167. [PMID: 33320079 DOI: 10.1099/mic.0.001008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Geobacillus kaustophilus HTA426, a thermophilic Gram-positive bacterium, feeds on inositol as its sole carbon source, and an iol gene cluster required for inositol catabolism has been postulated with reference to the iol genes in Bacillus subtilis. The iol gene cluster of G. kaustophilus comprises two tandem operons induced in the presence of inositol; however, the mechanism underlying this induction remains unclear. B. subtilis iolQ is known to be involved in the regulation of iolX encoding scyllo-inositol dehydrogenase, and its homologue in HTA426 was found two genes upstream of the first gene (gk1899) of the iol gene cluster and was termed iolQ in G. kaustophilus. When iolQ was inactivated in G. kaustophilus, not only cellular myo-inositol dehydrogenase activity due to gk1899 expression but also the transcription of the two iol operons became constitutive. IolQ was produced and purified as a C-terminal histidine (His)-tagged fusion protein in Escherichia coli and subjected to an in vitro gel electrophoresis mobility shift assay to examine its DNA-binding property. It was observed that IolQ bound to the DNA fragments containing each of the two iol promoter regions and that DNA binding was antagonized by myo-inositol. Moreover, DNase I footprinting analyses identified two tandem binding sites of IolQ within each of the iol promoter regions. By comparing the sequences of the binding sites, a consensus sequence for IolQ binding was deduced to form a palindrome of 5'-RGWAAGCGCTTSCY-3' (where R=A or G, W=A or T, S=G or C, and Y=C or T). IolQ functions as a transcriptional repressor regulating the induction of the two iol operons responding to myo-inositol.
Collapse
Affiliation(s)
- Ken-Ichi Yoshida
- Department of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe 657 8501, Japan
| | - Yusuke Shirae
- Department of Agrobioscience, Kobe University, 1-1 Rokkodai, Nada, Kobe 657 8501, Japan
| | - Ryo Nishimura
- Department of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe 657 8501, Japan
| | - Kaho Fukui
- Department of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe 657 8501, Japan
| | - Shu Ishikawa
- Department of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai, Nada, Kobe 657 8501, Japan
| |
Collapse
|
37
|
Rashid S, Bhat BA, Mehta G. Conceptualization and Synthesis of the First Inosito-Inositol (Decahydroxydecalin, DHD): In silico Binding to β-Amyloid Protein. Chemistry 2020; 26:17005-17010. [PMID: 32783222 DOI: 10.1002/chem.202003367] [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: 07/17/2020] [Revised: 08/04/2020] [Indexed: 11/10/2022]
Abstract
Previously unknown entities in the form of 1,2,3,4,5,6,7,8,9,10-decahydroxydecalins (DHDs) have been conceptualized and the first member of this class, an inosito-inositol, has been synthesized from aromatic hydrocarbon naphthalene following a flexible strategy that is amenable to diversity creation. The DHD accessed here has been subjected to preliminary in silico evaluation with Aβ and may hold some promise in Alzheimer's disease therapeutics.
Collapse
Affiliation(s)
- Showkat Rashid
- CSIR -, Indian Institute of Integrative Medicine, Sanatnagar, Srinagar, 190005 Jammu & Kashmir, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, 180001, India
| | - Bilal A Bhat
- CSIR -, Indian Institute of Integrative Medicine, Sanatnagar, Srinagar, 190005 Jammu & Kashmir, India.,Academy of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu Tawi, 180001, India
| | - Goverdhan Mehta
- School of Chemistry, University of Hyderabad, Hyderabad, 500046, India
| |
Collapse
|
38
|
Alzheimer's disease: Recent treatment strategies. Eur J Pharmacol 2020; 887:173554. [DOI: 10.1016/j.ejphar.2020.173554] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/03/2020] [Accepted: 09/11/2020] [Indexed: 12/12/2022]
|
39
|
Role of Inositols and Inositol Phosphates in Energy Metabolism. Molecules 2020; 25:molecules25215079. [PMID: 33139672 PMCID: PMC7663797 DOI: 10.3390/molecules25215079] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/12/2022] Open
Abstract
Recently, inositols, especially myo-inositol and inositol hexakisphosphate, also known as phytic acid or IP6, with their biological activities received much attention for their role in multiple health beneficial effects. Although their roles in cancer treatment and prevention have been extensively reported, interestingly, they may also have distinctive properties in energy metabolism and metabolic disorders. We review inositols and inositol phosphate metabolism in mammalian cells to establish their biological activities and highlight their potential roles in energy metabolism. These molecules are known to decrease insulin resistance, increase insulin sensitivity, and have diverse properties with importance from cell signaling to metabolism. Evidence showed that inositol phosphates might enhance the browning of white adipocytes and directly improve insulin sensitivity through adipocytes. In addition, inositol pyrophosphates containing high-energy phosphate bonds are considered in increasing cellular energetics. Despite all recent advances, many aspects of the bioactivity of inositol phosphates are still not clear, especially their effects on insulin resistance and alteration of metabolism, so more research is needed.
Collapse
|
40
|
García-Viñuales S, Ahmed R, Sciacca MFM, Lanza V, Giuffrida ML, Zimbone S, Romanucci V, Zarrelli A, Bongiorno C, Spinella N, Galati C, Di Fabio G, Melacini G, Milardi D. Trehalose Conjugates of Silybin as Prodrugs for Targeting Toxic Aβ Aggregates. ACS Chem Neurosci 2020; 11:2566-2576. [PMID: 32687307 DOI: 10.1021/acschemneuro.0c00232] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Alzheimer's disease (AD) is linked to the abnormal accumulation of amyloid β peptide (Aβ) aggregates in the brain. Silybin B, a natural compound extracted from milk thistle (Silybum marianum), has been shown to significantly inhibit Aβ aggregation in vitro and to exert neuroprotective properties in vivo. However, further explorations of silybin B's clinical potential are currently limited by three main factors: (a) poor solubility, (b) instability in blood serum, and (c) only partial knowledge of silybin's mechanism of action. Here, we address these three limitations. We demonstrate that conjugation of a trehalose moiety to silybin significantly increases both water solubility and stability in blood serum without significantly compromising its antiaggregation properties. Furthermore, using a combination of biophysical techniques with different spatial resolution, that is, TEM, ThT fluorescence, CD, and NMR spectroscopy, we profile the interactions of the trehalose conjugate with both Aβ monomers and oligomers and evidence that silybin may shield the "toxic" surfaces formed by the N-terminal and central hydrophobic regions of Aβ. Finally, comparative analysis with silybin A, a less active diastereoisomer of silybin B, revealed how even subtle differences in chemical structure may entail different effects on amyloid inhibition. The resulting insight on the mechanism of action of silybins as aggregation inhibitors is anticipated to facilitate the future investigation of silybin's therapeutic potential.
Collapse
Affiliation(s)
- Sara García-Viñuales
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Sede Secondaria di Catania, Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Rashik Ahmed
- Departments of Chemistry and Chemical Biology & Biochemistry and Biomedical Sciences, McMaster University, Hamilton ON L8S 4M1, Canada
| | - Michele F. M. Sciacca
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Sede Secondaria di Catania, Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Valeria Lanza
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Sede Secondaria di Catania, Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Maria Laura Giuffrida
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Sede Secondaria di Catania, Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Stefania Zimbone
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Sede Secondaria di Catania, Via Paolo Gaifami 18, 95126 Catania, Italy
| | - Valeria Romanucci
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, I-80126 Napoli, Italy
| | - Armando Zarrelli
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, I-80126 Napoli, Italy
| | - Corrado Bongiorno
- Institute for Microelectronics and Microsystems, National Research Council, Stradale Primosole 50, 95121 Catania, Italy
| | | | - Clelia Galati
- STMicroelectronics, Stradale Primosole 50, 95121 Catania, Italy
| | - Giovanni Di Fabio
- Department of Chemical Sciences, University of Naples Federico II, Via Cintia 4, I-80126 Napoli, Italy
| | - Giuseppe Melacini
- Departments of Chemistry and Chemical Biology & Biochemistry and Biomedical Sciences, McMaster University, Hamilton ON L8S 4M1, Canada
| | - Danilo Milardi
- Consiglio Nazionale delle Ricerche, Istituto di Cristallografia, Sede Secondaria di Catania, Via Paolo Gaifami 18, 95126 Catania, Italy
| |
Collapse
|
41
|
Pinheiro L, Faustino C. Therapeutic Strategies Targeting Amyloid-β in Alzheimer's Disease. Curr Alzheimer Res 2020; 16:418-452. [PMID: 30907320 DOI: 10.2174/1567205016666190321163438] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/16/2019] [Accepted: 03/17/2019] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder linked to protein misfolding and aggregation. AD is pathologically characterized by senile plaques formed by extracellular Amyloid-β (Aβ) peptide and Intracellular Neurofibrillary Tangles (NFT) formed by hyperphosphorylated tau protein. Extensive synaptic loss and neuronal degeneration are responsible for memory impairment, cognitive decline and behavioral dysfunctions typical of AD. Amyloidosis has been implicated in the depression of acetylcholine synthesis and release, overactivation of N-methyl-D-aspartate (NMDA) receptors and increased intracellular calcium levels that result in excitotoxic neuronal degeneration. Current drugs used in AD treatment are either cholinesterase inhibitors or NMDA receptor antagonists; however, they provide only symptomatic relief and do not alter the progression of the disease. Aβ is the product of Amyloid Precursor Protein (APP) processing after successive cleavage by β- and γ-secretases while APP proteolysis by α-secretase results in non-amyloidogenic products. According to the amyloid cascade hypothesis, Aβ dyshomeostasis results in the accumulation and aggregation of Aβ into soluble oligomers and insoluble fibrils. The former are synaptotoxic and can induce tau hyperphosphorylation while the latter deposit in senile plaques and elicit proinflammatory responses, contributing to oxidative stress, neuronal degeneration and neuroinflammation. Aβ-protein-targeted therapeutic strategies are thus a promising disease-modifying approach for the treatment and prevention of AD. This review summarizes recent findings on Aβ-protein targeted AD drugs, including β-secretase inhibitors, γ-secretase inhibitors and modulators, α-secretase activators, direct inhibitors of Aβ aggregation and immunotherapy targeting Aβ, focusing mainly on those currently under clinical trials.
Collapse
Affiliation(s)
- Lídia Pinheiro
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto 1649-003 Lisboa, Portugal
| | - Célia Faustino
- iMed.ULisboa - Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto 1649-003 Lisboa, Portugal
| |
Collapse
|
42
|
López-Gambero AJ, Sanjuan C, Serrano-Castro PJ, Suárez J, Rodríguez de Fonseca F. The Biomedical Uses of Inositols: A Nutraceutical Approach to Metabolic Dysfunction in Aging and Neurodegenerative Diseases. Biomedicines 2020; 8:biomedicines8090295. [PMID: 32825356 PMCID: PMC7554709 DOI: 10.3390/biomedicines8090295] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 02/05/2023] Open
Abstract
Inositols are sugar-like compounds that are widely distributed in nature and are a part of membrane molecules, participating as second messengers in several cell-signaling processes. Isolation and characterization of inositol phosphoglycans containing myo- or d-chiro-inositol have been milestones for understanding the physiological regulation of insulin signaling. Other functions of inositols have been derived from the existence of multiple stereoisomers, which may confer antioxidant properties. In the brain, fluctuation of inositols in extracellular and intracellular compartments regulates neuronal and glial activity. Myo-inositol imbalance is observed in psychiatric diseases and its use shows efficacy for treatment of depression, anxiety, and compulsive disorders. Epi- and scyllo-inositol isomers are capable of stabilizing non-toxic forms of β-amyloid proteins, which are characteristic of Alzheimer’s disease and cognitive dementia in Down’s syndrome, both associated with brain insulin resistance. However, uncertainties of the intrinsic mechanisms of inositols regarding their biology are still unsolved. This work presents a critical review of inositol actions on insulin signaling, oxidative stress, and endothelial dysfunction, and its potential for either preventing or delaying cognitive impairment in aging and neurodegenerative diseases. The biomedical uses of inositols may represent a paradigm in the industrial approach perspective, which has generated growing interest for two decades, accompanied by clinical trials for Alzheimer’s disease.
Collapse
Affiliation(s)
- Antonio J. López-Gambero
- Departamento de Biología Celular, Genética y Fisiología, Campus de Teatinos s/n, Universidad de Málaga, Andalucia Tech, 29071 Málaga, Spain;
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, 29010 Málaga, Spain
| | | | - Pedro Jesús Serrano-Castro
- UGC Neurología, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, 29010 Málaga, Spain;
| | - Juan Suárez
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, 29010 Málaga, Spain
- Correspondence: (J.S.); (F.R.d.F.); Tel.: +34-952614012 (J.S.)
| | - Fernando Rodríguez de Fonseca
- UGC Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Regional de Málaga, 29010 Málaga, Spain
- Correspondence: (J.S.); (F.R.d.F.); Tel.: +34-952614012 (J.S.)
| |
Collapse
|
43
|
Morrone CD, Bazzigaluppi P, Beckett TL, Hill ME, Koletar MM, Stefanovic B, McLaurin J. Regional differences in Alzheimer's disease pathology confound behavioural rescue after amyloid-β attenuation. Brain 2020; 143:359-373. [PMID: 31782760 PMCID: PMC6935751 DOI: 10.1093/brain/awz371] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/16/2019] [Accepted: 10/01/2019] [Indexed: 12/31/2022] Open
Abstract
Failure of Alzheimer’s disease clinical trials to improve or stabilize cognition has led to the need for a better understanding of the driving forces behind cognitive decline in the presence of active disease processes. To dissect contributions of individual pathologies to cognitive function, we used the TgF344-AD rat model, which recapitulates the salient hallmarks of Alzheimer’s disease pathology observed in patient populations (amyloid, tau inclusions, frank neuronal loss, and cognitive deficits). scyllo-Inositol treatment attenuated amyloid-β peptide in disease-bearing TgF344-AD rats, which rescued pattern separation in the novel object recognition task and executive function in the reversal learning phase of the Barnes maze. Interestingly, neither activities of daily living in the burrowing task nor spatial memory in the Barnes maze were rescued by attenuating amyloid-β peptide. To understand the pathological correlates leading to behavioural rescue, we examined the neuropathology and in vivo electrophysiological signature of the hippocampus. Amyloid-β peptide attenuation reduced hippocampal tau pathology and rescued adult hippocampal neurogenesis and neuronal function, via improvements in cross-frequency coupling between theta and gamma bands. To investigate mechanisms underlying the persistence of spatial memory deficits, we next examined neuropathology in the entorhinal cortex, a region whose input to the hippocampus is required for spatial memory. Reduction of amyloid-β peptide in the entorhinal cortex had no effect on entorhinal tau pathology or entorhinal-hippocampal neuronal network dysfunction, as measured by an impairment in hippocampal response to entorhinal stimulation. Thus, rescue or not of cognitive function is dependent on regional differences of amyloid-β, tau and neuronal network dysfunction, demonstrating the importance of staging disease in patients prior to enrolment in clinical trials. These results further emphasize the need for combination therapeutic approaches across disease progression.
Collapse
Affiliation(s)
- Christopher D Morrone
- Sunnybrook Research Institute, Biological Sciences, 2075 Bayview Ave, Toronto, ON, Canada.,University of Toronto, Faculty of Medicine, Department of Laboratory Medicine and Pathobiology, 1 King's College Cir, Toronto, ON, Canada
| | - Paolo Bazzigaluppi
- Sunnybrook Research Institute, Physical Sciences, 2075 Bayview Ave, Toronto, ON, Canada
| | - Tina L Beckett
- Sunnybrook Research Institute, Biological Sciences, 2075 Bayview Ave, Toronto, ON, Canada
| | - Mary E Hill
- Sunnybrook Research Institute, Biological Sciences, 2075 Bayview Ave, Toronto, ON, Canada
| | - Margaret M Koletar
- Sunnybrook Research Institute, Physical Sciences, 2075 Bayview Ave, Toronto, ON, Canada
| | - Bojana Stefanovic
- Sunnybrook Research Institute, Physical Sciences, 2075 Bayview Ave, Toronto, ON, Canada.,University of Toronto, Faculty of Medicine, Department of Medical Biophysics, 101 College St Suite 15-701, Toronto, ON, Canada
| | - JoAnne McLaurin
- Sunnybrook Research Institute, Biological Sciences, 2075 Bayview Ave, Toronto, ON, Canada.,University of Toronto, Faculty of Medicine, Department of Laboratory Medicine and Pathobiology, 1 King's College Cir, Toronto, ON, Canada
| |
Collapse
|
44
|
Uddin MS, Kabir MT, Rahman MS, Behl T, Jeandet P, Ashraf GM, Najda A, Bin-Jumah MN, El-Seedi HR, Abdel-Daim MM. Revisiting the Amyloid Cascade Hypothesis: From Anti-Aβ Therapeutics to Auspicious New Ways for Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21165858. [PMID: 32824102 PMCID: PMC7461598 DOI: 10.3390/ijms21165858] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/03/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is the most prevalent neurodegenerative disorder related to age, characterized by the cerebral deposition of fibrils, which are made from the amyloid-β (Aβ), a peptide of 40–42 amino acids. The conversion of Aβ into neurotoxic oligomeric, fibrillar, and protofibrillar assemblies is supposed to be the main pathological event in AD. After Aβ accumulation, the clinical symptoms fall out predominantly due to the deficient brain clearance of the peptide. For several years, researchers have attempted to decline the Aβ monomer, oligomer, and aggregate levels, as well as plaques, employing agents that facilitate the reduction of Aβ and antagonize Aβ aggregation, or raise Aβ clearance from brain. Unluckily, broad clinical trials with mild to moderate AD participants have shown that these approaches were unsuccessful. Several clinical trials are running involving patients whose disease is at an early stage, but the preliminary outcomes are not clinically impressive. Many studies have been conducted against oligomers of Aβ which are the utmost neurotoxic molecular species. Trials with monoclonal antibodies directed against Aβ oligomers have exhibited exciting findings. Nevertheless, Aβ oligomers maintain equivalent states in both monomeric and aggregation forms; so, previously administered drugs that precisely decrease Aβ monomer or Aβ plaques ought to have displayed valuable clinical benefits. In this article, Aβ-based therapeutic strategies are discussed and several promising new ways to fight against AD are appraised.
Collapse
Affiliation(s)
- Md. Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka 1213, Bangladesh
- Pharmakon Neuroscience Research Network, Dhaka 1207, Bangladesh
- Correspondence: ; Tel.: +880-171-022-0110
| | - Md. Tanvir Kabir
- Department of Pharmacy, BRAC University, Dhaka 1212, Bangladesh;
| | - Md. Sohanur Rahman
- Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi 6205, Bangladesh;
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India;
| | - Philippe Jeandet
- Research Unit, Induced Resistance and Plant Bioprotection, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences, University of Reims Champagne-Ardenne, PO Box 1039, 51687 Reims CEDEX 2, France;
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Agnieszka Najda
- Laboratory of Quality of Vegetables and Medicinal Plants, Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland;
| | - May N. Bin-Jumah
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia;
| | - Hesham R. El-Seedi
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China;
- Pharmacognosy Group, Department of Pharmaceutical Biosciences, Uppsala University, SE-751 23 Uppsala, Sweden
- Department of Chemistry, Faculty of Science, Menoufia University, Shebin El-Koom 32512, Egypt
| | - Mohamed M. Abdel-Daim
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
45
|
Yoshida KI, Ishikawa S. Production of scyllo-Inositol: Conversion of Rice Bran into a Promising Disease-Modifying Therapeutic Agent for Alzheimer's Disease. J Nutr Sci Vitaminol (Tokyo) 2020; 65:S139-S142. [PMID: 31619615 DOI: 10.3177/jnsv.65.s139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
scyllo-Inositol (SI) is one of the inositol stereoisomers, rare in the nature, and expected as a promising disease-modifying therapeutic agent for Alzheimer's disease. On the other hand, myo-inositol (MI) is another inositol stereoisomer most abundant in nature and thus supplied from agricultural byproducts including rice bran. Bacillus subtilis was genetically modified in its inositol metabolism and phytase secretion, to develope the bioconversion processes to produce SI from rice bran. Phytase, an enzyme that degrades phytate in rice bran into MI, was secreted in a B. subtilis strain with the optimized signal peptide. Another B. subtilis strain was constructed with the constitutive and simultaneous overexpression of IolG and IolW, which are the two inositol dehydrogenases responsible for the conversion, to demonstrate an efficient conversion of MI into SI with a rate up to 10 g/L/48 h. In order to devise further elevation in the conversion efficiency, we attempted to improve the substrate uptake by overexpressing iolT for the major MI transporter. In addition, Escherichia coli pntAB encoding the membrane-bound transhydrogenase was introduced aiming at enhanced supply of NADPH required for the rate-limiting IolW reaction. These additional modifications successfully elevated the conversion efficiency with an improved rate up to almost 30 g/L/48 h. Together with the improved phytase secretion, technological infrastructure for social implementation of SI production from rice bran is on the way.
Collapse
Affiliation(s)
- Ken-Ichi Yoshida
- Graduate School of Science, Technology and Innovation, Kobe University
| | - Shu Ishikawa
- Graduate School of Science, Technology and Innovation, Kobe University
| |
Collapse
|
46
|
Yang T, Zhu Z, Yin E, Wang Y, Zhang C, Yuan H, Zhang H, Jin S, Guo Z, Wang X. Alleviation of symptoms of Alzheimer's disease by diminishing Aβ neurotoxicity and neuroinflammation. Chem Sci 2019; 10:10149-10158. [PMID: 32055369 PMCID: PMC6979322 DOI: 10.1039/c9sc03042e] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/16/2019] [Indexed: 01/22/2023] Open
Abstract
Alzheimer's disease (AD) is one of the most prevailing neurodegenerative illnesses in the elderly. Accumulation of amyloid-β peptide (Aβ) and inflammation play critical roles in the pathogenesis and development of AD. Multi-target drugs may interdict the progress of AD through a synergistic mechanism. A neuromodulator, 2-((1H-benzo[d]imidazole-2-yl)methoxy)benzoic acid (BIBA), consisting of an Aβ-targeting group and a derivative of anti-inflammatory aspirin was designed as a potential anti-AD agent. BIBA exhibits a remarkable inhibitory effect on the self- and metal-induced Aβ aggregations and shows outstanding anti-inflammatory activity simultaneously. The neurotoxicity of Aβ aggregates is attenuated, and the production of pro-inflammatory cytokines (PICs), such as IL-6, IL-1β and TNF-α, in microglia stimulated by lipopolysaccharide (LPS) or Aβ is reduced. Owing to the synergy between the inhibition of Aβ oligomerization and downregulation of PICs, BIBA markedly prolongs the lifespan and relieves the Aβ-induced paralysis of Aβ-transgenic Caenorhabditis elegans, thus showing the potential to ameliorate the symptoms of AD through inhibiting Aβ neurotoxicity and deactivating microglia. These findings demonstrate that both Aβ aggregation and neuroinflammation are therapeutic targets for anti-AD drugs, and dual-functional agents that integrate anti-Aβ and anti-inflammatory capabilities have great advantages over the traditional single-target agents for AD treatment.
Collapse
Affiliation(s)
- Tao Yang
- State Key Laboratory of Pharmaceutical Biotechnology , School of Life Sciences , Nanjing University , Nanjing 210023 , P. R. China .
| | - Zhenzhu Zhu
- State Key Laboratory of Pharmaceutical Biotechnology , School of Life Sciences , Nanjing University , Nanjing 210023 , P. R. China .
| | - Enmao Yin
- State Key Laboratory of Pharmaceutical Biotechnology , School of Life Sciences , Nanjing University , Nanjing 210023 , P. R. China .
| | - Yanqing Wang
- School of Chemistry and Chemical Engineering , Yancheng Teachers University , Yancheng 224002 , P. R. China
| | - Changli Zhang
- Department of Chemistry , Nanjing Xiaozhuang College , Nanjing , 210017 , P. R. China
| | - Hao Yuan
- State Key Laboratory of Coordination Chemistry , School of Chemistry and Chemical Engineering , Nanjing University , Nanjing 210023 , P. R. China
| | - Hongmei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology , School of Life Sciences , Nanjing University , Nanjing 210023 , P. R. China .
| | - Suxing Jin
- State Key Laboratory of Pharmaceutical Biotechnology , School of Life Sciences , Nanjing University , Nanjing 210023 , P. R. China .
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry , School of Chemistry and Chemical Engineering , Nanjing University , Nanjing 210023 , P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology , School of Life Sciences , Nanjing University , Nanjing 210023 , P. R. China .
| |
Collapse
|
47
|
Multi-enzyme systems and recombinant cells for synthesis of valuable saccharides: Advances and perspectives. Biotechnol Adv 2019; 37:107406. [DOI: 10.1016/j.biotechadv.2019.06.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/30/2019] [Accepted: 06/08/2019] [Indexed: 02/07/2023]
|
48
|
Li Y, Liu S, You C. Permeabilized
Escherichia coli
Whole Cells Containing Co‐Expressed Two Thermophilic Enzymes Facilitate the Synthesis of
scyllo
‐Inositol from
myo
‐Inositol. Biotechnol J 2019; 15:e1900191. [DOI: 10.1002/biot.201900191] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 08/28/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Yuan Li
- Tianjin Institute of Industrial BiotechnologyChinese Academy of Sciences Tianjin 300308 P. R. China
| | - Shan Liu
- Tianjin Institute of Industrial BiotechnologyChinese Academy of Sciences Tianjin 300308 P. R. China
| | - Chun You
- Tianjin Institute of Industrial BiotechnologyChinese Academy of Sciences Tianjin 300308 P. R. China
- University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
49
|
Bazzigaluppi P, Beckett TL, Koletar MM, Hill ME, Lai A, Trivedi A, Thomason L, Dorr A, Gallagher D, Librach CL, Joo IL, McLaurin J, Stefanovic B. Combinatorial Treatment Using Umbilical Cord Perivascular Cells and Aβ Clearance Rescues Vascular Function Following Transient Hypertension in a Rat Model of Alzheimer Disease. Hypertension 2019; 74:1041-1051. [PMID: 31476904 PMCID: PMC6739147 DOI: 10.1161/hypertensionaha.119.13187] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Supplemental Digital Content is available in the text. Transient hypertension is a risk factor for Alzheimer disease (AD), but the effects of this interaction on brain vasculature are understudied. Addressing vascular pathology is a promising avenue to potentiate the efficacy of treatments for AD. We used arterial spin labeling magnetic resonance imaging to longitudinally assess brain vascular function and immunohistopathology to examine cerebrovascular remodeling and amyloid load. Hypertension was induced for 1 month by administration of l-NG-nitroarginine-methyl-ester in TgF344-AD rats at the prodromal stage. Following hypertension, nontransgenic rats showed transient cerebrovascular changes, whereas TgF344-AD animals exhibited sustained alterations in cerebrovascular function. Human umbilical cord perivascular cells in combination with scyllo-inositol, an inhibitor of Aβ oligomerization, resulted in normalization of hippocampal vascular function and remodeling, in contrast to either treatment alone. Prodromal stage hypertension exacerbates latter AD pathology, and the combination of human umbilical cord perivascular cells with amyloid clearance promotes cerebrovascular functional recovery.
Collapse
Affiliation(s)
- Paolo Bazzigaluppi
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Tina L Beckett
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Margaret M Koletar
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Mary E Hill
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Aaron Lai
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Arunachala Trivedi
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Lynsie Thomason
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Adrienne Dorr
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | | | - Clifford L Librach
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.).,Division of Obstetrics and Gynaecology, Laboratory Medicine and Pathobiology (C.L.), University of Toronto, Canada.,CReATe Research Program, Toronto, Canada (D.G., C.L.L.)
| | - Illsung L Joo
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - JoAnne McLaurin
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.)
| | - Bojana Stefanovic
- From the Sunnybrook Research Institute, Toronto, Canada (P.B., T.L.B., M.M.K., M.E.H., A.L., A.T., L.T., A.D., C.L.L., I.L.J., J.M., B.S.).,Department of Medical Biophysics (B.S.), University of Toronto, Canada
| |
Collapse
|
50
|
Andrade S, Ramalho MJ, Loureiro JA, Pereira MDC. Natural Compounds for Alzheimer's Disease Therapy: A Systematic Review of Preclinical and Clinical Studies. Int J Mol Sci 2019; 20:E2313. [PMID: 31083327 PMCID: PMC6539304 DOI: 10.3390/ijms20092313] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/04/2019] [Accepted: 05/07/2019] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's Disease (AD) is a neurodegenerative disorder related with the increase of age and it is the main cause of dementia in the world. AD affects cognitive functions, such as memory, with an intensity that leads to several functional losses. The continuous increase of AD incidence demands for an urgent development of effective therapeutic strategies. Despite the extensive research on this disease, only a few drugs able to delay the progression of the disease are currently available. In the last years, several compounds with pharmacological activities isolated from plants, animals and microorganisms, revealed to have beneficial effects for the treatment of AD, targeting different pathological mechanisms. Thus, a wide range of natural compounds may play a relevant role in the prevention of AD and have proven to be efficient in different preclinical and clinical studies. This work aims to review the natural compounds that until this date were described as having significant benefits for this neurological disease, focusing on studies that present clinical trials.
Collapse
Affiliation(s)
- Stephanie Andrade
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal.
| | - Maria João Ramalho
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal.
| | - Joana Angélica Loureiro
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal.
| | - Maria do Carmo Pereira
- LEPABE, Department of Chemical Engineering, Faculty of Engineering of the University of Porto, 4200-465 Porto, Portugal.
| |
Collapse
|