1
|
Budamagunta MS, Mori H, Silk J, Slez RR, Bognár B, Mendiola UR, Kálai T, Maezawa I, Voss JC. Nitroxyl Hybrids with Curcumin and Stilbene Scaffolds Display Potent Antioxidant Activity, Remodel the Amyloid Beta Oligomer, and Reverse Amyloid Beta-Induced Cytotoxicity. Antioxidants (Basel) 2024; 13:1411. [PMID: 39594552 PMCID: PMC11591036 DOI: 10.3390/antiox13111411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
The disorder and heterogeneity of low-molecular-weight amyloid-beta oligomers (AβOs) underlie their participation in multiple modes of cellular dysfunction associated with the etiology of Alzheimer's disease (AD). The lack of specified conformational states in these species complicates efforts to select or design small molecules to targeting discrete pathogenic states. Furthermore, targeting AβOs alone may be therapeutically insufficient, as AD progresses as a multifactorial, self-amplifying cascade. To address these challenges, we have screened the activity of seven new candidates that serve as Paramagnetic Amyloid Ligand (PAL) candidates. PALs are bifunctional small molecules that both remodel the AβO structure and localize a potent antioxidant that mimics the activity of SOD within live cells. The candidates are built from either a stilbene or curcumin scaffold with nitroxyl moiety to serve as catalytic antioxidants. Measurements of PAL AβO binding and remolding along with assessments of bioactivity allow for the extraction of useful SAR information from screening data. One candidate (HO-4450; PMT-307), with a six-membered nitroxyl ring attached to a stilbene ring, displays the highest potency in protecting against cell-derived Aβ. A preliminary low-dose evaluation in AD model mice provides evidence of modest treatment effects by HO-4450. The results for the curcumin PALs demonstrate that the retention of the native curcumin phenolic groups is advantageous to the design of the hybrid PAL candidates. Finally, the PAL remodeling of AβO secondary structures shows a reasonable correlation between a candidate's bioactivity and its ability to reduce the fraction of antiparallel β-strand.
Collapse
Affiliation(s)
- Madhu S. Budamagunta
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA 95616, USA; (M.S.B.); (J.S.); (R.R.S.)
| | - Hidetoshi Mori
- Center for Genomic Pathology, University of California Davis, Sacramento, CA 95817, USA
| | - Joshua Silk
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA 95616, USA; (M.S.B.); (J.S.); (R.R.S.)
- Paramag Biosciences Inc., 720 Olive Drive, Davis, CA 95616, USA
| | - Ryan R. Slez
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA 95616, USA; (M.S.B.); (J.S.); (R.R.S.)
| | - Balázs Bognár
- Institute of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Pécs, Honvéd St. 1., H-7624 Pécs, Hungary; (B.B.); (T.K.)
- János Szentágothai Research Center, Ifjúság St. 20., H-7624 Pécs, Hungary
| | - Ulises Ruiz Mendiola
- M.I.N.D. Institute and Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA 95817, USA;
| | - Tamás Kálai
- Institute of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Pécs, Honvéd St. 1., H-7624 Pécs, Hungary; (B.B.); (T.K.)
- János Szentágothai Research Center, Ifjúság St. 20., H-7624 Pécs, Hungary
| | - Izumi Maezawa
- M.I.N.D. Institute and Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento, CA 95817, USA;
| | - John C. Voss
- Department of Biochemistry & Molecular Medicine, University of California, Davis, CA 95616, USA; (M.S.B.); (J.S.); (R.R.S.)
- Paramag Biosciences Inc., 720 Olive Drive, Davis, CA 95616, USA
| |
Collapse
|
2
|
Jin LW, di Lucente J, Mendiola UR, Tang X, Zivkovic AM, Lebrilla CB, Maezawa I. The role of FUT8-catalyzed core fucosylation in Alzheimer's amyloid-β oligomer-induced activation of human microglia. Glia 2023; 71:1346-1359. [PMID: 36692036 PMCID: PMC11021125 DOI: 10.1002/glia.24345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/03/2023] [Accepted: 01/10/2023] [Indexed: 01/25/2023]
Abstract
Fucosylation, especially core fucosylation of N-glycans catalyzed by α1-6 fucosyltransferase (fucosyltransferase 8 or FUT8), plays an important role in regulating the peripheral immune system and inflammation. However, its role in microglial activation is poorly understood. Here we used human induced pluripotent stem cells-derived microglia (hiMG) as a model to study the role of FUT8-catalyzed core fucosylation in amyloid-β oligomer (AβO)-induced microglial activation, in view of its significant relevance to the pathogenesis of Alzheimer's disease (AD). HiMG responded to AβO and lipopolysaccharides (LPS) with a pattern of pro-inflammatory activation as well as enhanced core fucosylation and FUT8 expression within 24 h. Furthermore, we found increased FUT8 expression in both human AD brains and microglia isolated from 5xFAD mice, a model of AD-like cerebral amyloidosis. Inhibition of fucosylation in AβO-stimulated hiMG reduced the induction of pro-inflammatory cytokines, suppressed the activation of p38MAPK, and rectified phagocytic deficits. Specific inhibition of FUT8 by siRNA-mediated knockdown also reduced AβO-induced pro-inflammatory cytokines. We further showed that p53 binds to the two consensus binding sites in the Fut8 promoter, and that p53 knockdown abolished FUT8 overexpression in AβO-activated hiMG. Taken together, our evidence supports that FUT8-catalyzed core fucosylation is a signaling pathway required for AβO-induced microglia activation and that FUT8 is a component of the p53 signaling cascade regulating microglial behavior. Because microglia are a key driver of AD pathogenesis, our results suggest that microglial FUT8 could be an anti-inflammatory therapeutic target for AD.
Collapse
Affiliation(s)
- Lee-Way Jin
- Department of Pathology and Laboratory Medicine and M.I.N.D. Institute, University of California Davis Medical Center, 2805 50 Street, Sacramento, CA 95817
| | - Jacopo di Lucente
- Department of Pathology and Laboratory Medicine and M.I.N.D. Institute, University of California Davis Medical Center, 2805 50 Street, Sacramento, CA 95817
| | - Ulises R. Mendiola
- Department of Pathology and Laboratory Medicine and M.I.N.D. Institute, University of California Davis Medical Center, 2805 50 Street, Sacramento, CA 95817
| | - Xinyu Tang
- Department of Nutrition, University of California, Davis, CA 95618
| | | | | | - Izumi Maezawa
- Department of Pathology and Laboratory Medicine and M.I.N.D. Institute, University of California Davis Medical Center, 2805 50 Street, Sacramento, CA 95817
| |
Collapse
|
3
|
Tanaka M, Takarada T, Nadanaka S, Kojima R, Hosoi K, Machiba Y, Kitagawa H, Yamada T. Influences of amino-terminal modifications on amyloid fibril formation of human serum amyloid A. Arch Biochem Biophys 2023; 742:109615. [PMID: 37105512 DOI: 10.1016/j.abb.2023.109615] [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: 02/26/2023] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 04/29/2023]
Abstract
Human serum amyloid A (SAA) is a precursor protein involved in AA amyloidosis. The N-terminal region of the SAA molecule is crucial for amyloid fibril formation, and therefore modifications in this region are considered to influence the pathogenesis of AA amyloidosis. In the present study, using the N-terminal peptide corresponding to the putative first helix region of the SAA molecule, we investigated the influences of N-terminal modifications on amyloid fibril formation. Spectroscopic analyses revealed that carbamoylation of the N-terminal amino group delayed the onset of amyloid fibril formation. From transmission electron microscopic observations, the N-terminal carbamoylated aggregate showed remarkably different morphologies from the unmodified control. In contrast, acetylation of the N-terminal amino group or truncation of N-terminal amino acid(s) considerably diminished amyloidogenic properties. Furthermore, we also tested the cell toxicity of each peptide aggregate on cultured cells by two cytotoxic assays. Irrespective of carbamoylation or acetylation, MTT assay revealed that SAA peptides reduced the reductive activity of MTT on cells, whereas no apparent increase in LDH release was observed during an LDH assay. In contrast, N-terminal truncation did not affect either MTT reduction or LDH release. These results suggest that N-terminal modification of SAA molecules can act as a switch to regulate susceptibility to AA amyloidosis.
Collapse
Affiliation(s)
- Masafumi Tanaka
- Laboratory of Functional Molecular Chemistry, Kobe Pharmaceutical University, Kobe, 658-8558, Japan.
| | - Toru Takarada
- Laboratory of Functional Molecular Chemistry, Kobe Pharmaceutical University, Kobe, 658-8558, Japan
| | - Satomi Nadanaka
- Laboratory of Biochemistry, Kobe Pharmaceutical University, Kobe, 658-8558, Japan
| | - Risa Kojima
- Laboratory of Functional Molecular Chemistry, Kobe Pharmaceutical University, Kobe, 658-8558, Japan
| | - Kimiko Hosoi
- Laboratory of Functional Molecular Chemistry, Kobe Pharmaceutical University, Kobe, 658-8558, Japan
| | - Yuki Machiba
- Laboratory of Functional Molecular Chemistry, Kobe Pharmaceutical University, Kobe, 658-8558, Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry, Kobe Pharmaceutical University, Kobe, 658-8558, Japan
| | - Toshiyuki Yamada
- Department of Clinical Laboratory Medicine, Jichi Medical University, Shimotsuke, 329-0498, Japan
| |
Collapse
|
4
|
Hilt S, Liu R, Maezawa I, Rojalin T, Aung HH, Budamagunta M, Slez R, Gong Q, Carney RP, Voss JC. Novel Stilbene-Nitroxyl Hybrid Compounds Display Discrete Modulation of Amyloid Beta Toxicity and Structure. Front Chem 2022; 10:896386. [PMID: 35720993 PMCID: PMC9204515 DOI: 10.3389/fchem.2022.896386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
Several neurodegenerative diseases are driven by misfolded proteins that assemble into soluble aggregates. These "toxic oligomers" have been associated with a plethora of cellular dysfunction and dysregulation, however the structural features underlying their toxicity are poorly understood. A major impediment to answering this question relates to the heterogeneous nature of the oligomers, both in terms of structural disorder and oligomer size. This not only complicates elucidating the molecular etiology of these disorders, but also the druggability of these targets as well. We have synthesized a class of bifunctional stilbenes to modulate both the conformational toxicity within amyloid beta oligomers (AβO) and the oxidative stress elicited by AβO. Using a neuronal culture model, we demonstrate this bifunctional approach has the potential to counter the molecular pathogenesis of Alzheimer's disease in a powerful, synergistic manner. Examination of AβO structure by various biophysical tools shows that each stilbene candidate uniquely alters AβO conformation and toxicity, providing insight towards the future development of structural correctors for AβO. Correlations of AβO structural modulation and bioactivity displayed by each provides insights for future testing in vivo. The multi-target activity of these hybrid molecules represents a highly advantageous feature for disease modification in Alzheimer's, which displays a complex, multifactorial etiology. Importantly, these novel small molecules intervene with intraneuronal AβO, a necessary feature to counter the cycle of dysregulation, oxidative stress and inflammation triggered during the earliest stages of disease progression.
Collapse
Affiliation(s)
- Silvia Hilt
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, United States
| | - Ruiwu Liu
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, United States
| | - Izumi Maezawa
- M.I.N.D. Institute and Department of Pathology and Laboratory Medicine, University of California, Davis, Davis, CA, United States
| | - Tatu Rojalin
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - Hnin H. Aung
- Division of Cardiovascular Medicine, Department of Internal Medicine, School of Medicine, University of California, Davis, Davis, CA, United States
- Research Division, California Air Resource Board, Sacramento, CA, United States
| | - Madhu Budamagunta
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, United States
| | - Ryan Slez
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, United States
| | - Qizhi Gong
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Randy P. Carney
- Department of Biomedical Engineering, University of California, Davis, Davis, CA, United States
| | - John C. Voss
- Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, United States
- Paramag Biosciences Inc., Davis, CA, United States
| |
Collapse
|
5
|
Rishton GM, Look GC, Ni ZJ, Zhang J, Wang Y, Huang Y, Wu X, Izzo NJ, LaBarbera KM, Limegrover CS, Rehak C, Yurko R, Catalano SM. Discovery of Investigational Drug CT1812, an Antagonist of the Sigma-2 Receptor Complex for Alzheimer's Disease. ACS Med Chem Lett 2021; 12:1389-1395. [PMID: 34531947 DOI: 10.1021/acsmedchemlett.1c00048] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 08/03/2021] [Indexed: 02/08/2023] Open
Abstract
An unbiased phenotypic neuronal assay was developed to measure the synaptotoxic effects of soluble Aβ oligomers. A collection of CNS druglike small molecules prepared by conditioned extraction was screened. Compounds that prevented and reversed synaptotoxic effects of Aβ oligomers in neurons were discovered to bind to the sigma-2 receptor complex. Select development compounds displaced receptor-bound Aβ oligomers, rescued synapses, and restored cognitive function in transgenic hAPP Swe/Ldn mice. Our first-in-class orally administered small molecule investigational drug 7 (CT1812) has been advanced to Phase II clinical studies for Alzheimer's disease.
Collapse
Affiliation(s)
- Gilbert M. Rishton
- Cognition Therapeutics, 2403 Sidney Street, Suite 261, Pittsburgh, Pennsylvania 15203, United States
| | - Gary C. Look
- Cognition Therapeutics, 2403 Sidney Street, Suite 261, Pittsburgh, Pennsylvania 15203, United States
| | - Zhi-Jie Ni
- Acme Bioscience, Inc., 3941 East Bayshore Road, Palo Alto, California 94303, United States
| | - Jason Zhang
- Acme Bioscience, Inc., 3941 East Bayshore Road, Palo Alto, California 94303, United States
| | - Yingcai Wang
- Acme Bioscience, Inc., 3941 East Bayshore Road, Palo Alto, California 94303, United States
| | - Yaodong Huang
- Acme Bioscience, Inc., 3941 East Bayshore Road, Palo Alto, California 94303, United States
| | - Xiaodong Wu
- Acme Bioscience, Inc., 3941 East Bayshore Road, Palo Alto, California 94303, United States
| | - Nicholas J. Izzo
- Cognition Therapeutics, 2403 Sidney Street, Suite 261, Pittsburgh, Pennsylvania 15203, United States
| | - Kelsie M LaBarbera
- Cognition Therapeutics, 2403 Sidney Street, Suite 261, Pittsburgh, Pennsylvania 15203, United States
| | - Colleen S. Limegrover
- Cognition Therapeutics, 2403 Sidney Street, Suite 261, Pittsburgh, Pennsylvania 15203, United States
| | - Courtney Rehak
- Cognition Therapeutics, 2403 Sidney Street, Suite 261, Pittsburgh, Pennsylvania 15203, United States
| | - Raymond Yurko
- Cognition Therapeutics, 2403 Sidney Street, Suite 261, Pittsburgh, Pennsylvania 15203, United States
| | - Susan M. Catalano
- Cognition Therapeutics, 2403 Sidney Street, Suite 261, Pittsburgh, Pennsylvania 15203, United States
| |
Collapse
|
6
|
Saathoff J, Green J, Jiang Y, Xu Y, Kellogg GE, Zhang S. Structural understanding of 5-(4-hydroxy-phenyl)-N-(2-(5-methoxy-1H-indol-3-yl)-ethyl)-3-oxopentanamide as a neuroprotectant for Alzheimer's disease. Bioorg Med Chem Lett 2021; 43:128081. [PMID: 33964442 PMCID: PMC8187328 DOI: 10.1016/j.bmcl.2021.128081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 10/21/2022]
Abstract
In our continuing efforts to develop novel neuroprotectants for Alzheimer's disease (AD), a series of analogs based on a lead compound that was recently shown to target the mitochondrial complex I were designed, synthesized and biologically characterized to understand the structure features that are important for neuroprotective activities. The results from a cellular AD model highlighted the important roles of the 4-OH on the phenyl ring and the 5-OCH3 on the indole ring of the lead compound. The results also demonstrated that the β-keto moiety can be modified to retain or improve the neuroprotective activity. Docking studies of selected analogs to the FMN site of mitochondrial complex I also supported the observed neuroprotective activities. Collectively, the results provide further information to guide optimization and development of analogs based on this chemical scaffold as neuroprotectants with a novel mechanism of action for AD.
Collapse
Affiliation(s)
- John Saathoff
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Jakob Green
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Yuqi Jiang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Yiming Xu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Glen E Kellogg
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
7
|
Limegrover CS, LeVine H, Izzo NJ, Yurko R, Mozzoni K, Rehak C, Sadlek K, Safferstein H, Catalano SM. Alzheimer's protection effect of A673T mutation may be driven by lower Aβ oligomer binding affinity. J Neurochem 2021; 157:1316-1330. [PMID: 33025581 PMCID: PMC8246829 DOI: 10.1111/jnc.15212] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/16/2022]
Abstract
Several mutations conferring protection against Alzheimer's disease (AD) have been described, none as profound as the A673T mutation, where carriers are four times less likely to get AD compared to noncarriers. This mutation results in reduced amyloid beta (Aβ) protein production in vitro and lower lifetime Aβ concentration in carriers. Better understanding of the protective mechanisms of the mutation may provide important insights into AD pathophysiology and identify productive therapeutic intervention strategies for disease modification. Aβ(1-42) protein forms oligomers that bind saturably to a single receptor site on neuronal synapses, initiating the downstream toxicities observed in AD. Decreased formation, toxicity, or stability of soluble Aβ oligomers, or reduction of synaptic binding of these oligomers, may combine with overall lower Aβ concentration to underlie A673T's disease protecting mechanism. To investigate these possibilities, we compared the formation rate of soluble oligomers made from Icelandic A673T mutant and wild type (wt) Aβ(1-42) synthetic protein, the amount and intensity of oligomer bound to mature primary rat hippocampal/cortical neuronal synapses, and the potency of bound oligomers to impact trafficking rate in neurons in vitro using a physiologically relevant oligomer preparation method. At equal protein concentrations, mutant protein forms approximately 50% or fewer oligomers of high molecular weight (>50 kDa) compared to wt protein. Mutant oligomers are twice as potent at altering the cellular vesicle trafficking rate as wt at equivalent concentrations, however, mutant oligomers have a >4-fold lower binding affinity to synaptic receptors (Kd = 1,950 vs. 442 nM). The net effect of these differences is a lower overall toxicity at a given concentration. This study demonstrates for the first time that mutant A673T Aβ oligomers prepared with this method have fundamentally different assembly characteristics and biological impact from wt protein and indicates that its disease protecting mechanism may result primarily from the mutant protein's much lower binding affinity to synaptic receptors. This suggests that therapeutics that effectively reduce oligomer binding to synapses in the brain may be beneficial in AD.
Collapse
Affiliation(s)
| | - Harry LeVine
- Sanders‐Brown Center on AgingUniversity of KentuckyLexingtonKYUSA
| | | | | | | | | | | | | | | |
Collapse
|
8
|
LaBarbera KM, Limegrover C, Rehak C, Yurko R, Izzo NJ, Knezovich N, Watto E, Waybright L, Catalano SM. Modeling the mature CNS: A predictive screening platform for neurodegenerative disease drug discovery. J Neurosci Methods 2021; 358:109180. [PMID: 33836174 PMCID: PMC8217273 DOI: 10.1016/j.jneumeth.2021.109180] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 12/20/2022]
Abstract
Background: Mature primary neuronal cultures are an important model of the nervous system, but limited scalability has been a major challenge in their use for drug discovery of neurodegenerative diseases. This work describes a method for improving scalability through the use of larger format microtiter plates while preserving culture quality. New Method: Here we describe a method and quality control procedures for growing embryonic day 18 rat hippocampal/cortical neuronal cultures in 384-well microtiter plates for three weeks in vitro. Results: We use these cultures in two assays measuring intracellular lipid vesicle trafficking and synapse density for routine screening of small molecule libraries. Together this culture system and screening platform have successfully identified therapeutics capable of improving cognitive function in transgenic models of Alzheimer’s disease that have advanced to clinical trials, validating their translational applicability. Comparison with Existing Methods: Our method enables the growth of healthy, mature neurons in larger format microtiter plates than in traditional primary neuronal culturing protocols, making it ideal for drug screening and mechanism of action studies. Conclusion: The predictive capacity of this culture system and screening platform provides a method for rapidly identifying novel disease-modifying neurodegenerative therapeutics.
Collapse
Affiliation(s)
| | | | - Courtney Rehak
- Cognition Therapeutics Inc., Pittsburgh, PA, 15203, United States
| | - Raymond Yurko
- Cognition Therapeutics Inc., Pittsburgh, PA, 15203, United States
| | | | - Nicole Knezovich
- Cognition Therapeutics Inc., Pittsburgh, PA, 15203, United States
| | - Emily Watto
- Cognition Therapeutics Inc., Pittsburgh, PA, 15203, United States
| | - Lora Waybright
- Cognition Therapeutics Inc., Pittsburgh, PA, 15203, United States
| | - Susan M Catalano
- Cognition Therapeutics Inc., Pittsburgh, PA, 15203, United States.
| |
Collapse
|
9
|
Limegrover CS, Yurko R, Izzo NJ, LaBarbera KM, Rehak C, Look G, Rishton G, Safferstein H, Catalano SM. Sigma-2 receptor antagonists rescue neuronal dysfunction induced by Parkinson's patient brain-derived α-synuclein. J Neurosci Res 2021; 99:1161-1176. [PMID: 33480104 PMCID: PMC7986605 DOI: 10.1002/jnr.24782] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/03/2020] [Accepted: 12/13/2020] [Indexed: 12/11/2022]
Abstract
α‐Synuclein oligomers are thought to have a pivotal role in sporadic and familial Parkinson's disease (PD) and related α‐synucleinopathies, causing dysregulation of protein trafficking, autophagy/lysosomal function, and protein clearance, as well as synaptic function impairment underlying motor and cognitive symptoms of PD. Moreover, trans‐synaptic spread of α‐synuclein oligomers is hypothesized to mediate disease progression. Therapeutic approaches that effectively block α‐synuclein oligomer‐induced pathogenesis are urgently needed. Here, we show for the first time that α‐synuclein species isolated from human PD patient brain and recombinant α‐synuclein oligomers caused similar deficits in lipid vesicle trafficking rates in cultured rat neurons and glia, while α‐synuclein species isolated from non‐PD human control brain samples did not. Recombinant α‐synuclein oligomers also increased neuronal expression of lysosomal‐associated membrane protein‐2A (LAMP‐2A), the lysosomal receptor that has a critical role in chaperone‐mediated autophagy. Unbiased screening of several small molecule libraries (including the NIH Clinical Collection) identified sigma‐2 receptor antagonists as the most effective at blocking α‐synuclein oligomer‐induced trafficking deficits and LAMP‐2A upregulation in a dose‐dependent manner. These results indicate that antagonists of the sigma‐2 receptor complex may alleviate α‐synuclein oligomer‐induced neurotoxicity and are a novel therapeutic approach for disease modification in PD and related α‐synucleinopathies.
Collapse
Affiliation(s)
| | | | | | | | | | - Gary Look
- Cognition Therapeutics Inc., Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
10
|
Green JC, Jiang Y, He L, Xu Y, Sun D, Keoprasert T, Nelson C, Oh U, Lesnefsky EJ, Kellogg GE, Chen Q, Zhang S. Characterization and Discovery of a Selective Small-Molecule Modulator of Mitochondrial Complex I Targeting a Unique Binding Site. J Med Chem 2020; 63:11819-11830. [PMID: 32945676 DOI: 10.1021/acs.jmedchem.0c01021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mitochondrial dysfunction has been recognized as an essential contributor to many human diseases including neurodegenerative disorders. However, the exact pathological role of mitochondrial dysfunction, especially in mitochondrial reactive oxygen species-associated oxidative stress, remains elusive, partially due to the lack of chemical probes with well-defined mechanisms of action. Herein, we describe the characterization and discovery of a rationally designed small molecule ZCM-I-1 as a selective modulator of the production of reactive oxygen species from mitochondrial complex I that does not alter mitochondrial membrane potential and bioenergetics. Chemical biology studies employing photoaffinity probes derived from ZCM-I-1 demonstrated its novel mechanism of action of modulating complex I via interactions with the flavin mononucleotide site, proximal in the reaction pathway within complex I.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Edward J Lesnefsky
- Medical Service, McGuire Department of Veteran Affairs Medical Center, Richmond, Virginia 23224, United States
| | | | | | | |
Collapse
|
11
|
Balavigneswaran CK, Kumar G, Vignesh Kumar C, Sellamuthu S, Kasiviswanathan U, Ray B, Muthuvijayan V, Mahto SK, Misra N. Gelatin grafted poly(D,L-lactide) as an inhibitor of protein aggregation: An in vitro case study. Biopolymers 2020; 111:e23383. [PMID: 32604473 DOI: 10.1002/bip.23383] [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: 01/23/2020] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 11/10/2022]
Abstract
Amyloids are a group of proteins that are capable of forming aggregated amyloid fibrils, which is responsible for many neurodegenerative diseases including Alzheimer's disease (AD). In our previous study, synthesis and characterization of star-shaped poly(D,L-lactide)-b-gelatin (ss-pLG) have been reported. In the present work, we have extended our work to study ss-pLG against protein aggregation. To the best of our knowledge, this is the first report on the inhibition of amyloid fibrillation by protein grafted poly(D,L-lactide). Bovine serum albumin (BSA) was chosen as the model protein, which readily forms fibril under high temperature. We found that ss-pLG efficiently suppressed the fibril formation of BSA compared with gelatin (Gel), which was supported by Thioflavin T assay, circular dichroism (CD) spectroscopy and atomic force microscopy (AFM). In addition, ss-pLG significantly curtailed amyloid-induced hemolysis. We also found that incubation of ss-pLG with neuroblastoma cells (MC65) protected the cells from fibril-induced toxicity. The rescuing efficiency of ss-pLG was better than Gel, which could be attributed to the reduced lamella thickness in branched ss-pLG. These results suggest the significance of gelatin grafting, which probably allows gelatin to interact with the key residues of the amyloidogenic core of BSA effectively.
Collapse
Affiliation(s)
- Chelladurai Karthikeyan Balavigneswaran
- Polymer Engineering Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India.,Tissue Engineering and Biomaterials Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Gaurav Kumar
- Electrophysiology Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India.,Department of Clinical Research, School of Biosciences and Biomedical Engineering, Galgotias University, Greater Noida, Uttar Pradesh, India
| | | | - Satheeshkumar Sellamuthu
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Uvanesh Kasiviswanathan
- Bioelectronics and Instrumentation Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Biswajit Ray
- Department of Chemistry, Institute of Science, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh, India
| | - Vignesh Muthuvijayan
- Tissue Engineering and Biomaterials Laboratory, Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu, India
| | - Sanjeev Kumar Mahto
- Tissue Engineering and Biomicrofluidics Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Nira Misra
- Polymer Engineering Laboratory, School of Biomedical Engineering, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| |
Collapse
|
12
|
Esculetin as a Bifunctional Antioxidant Prevents and Counteracts the Oxidative Stress and Neuronal Death Induced by Amyloid Protein in SH-SY5Y Cells. Antioxidants (Basel) 2020; 9:antiox9060551. [PMID: 32630394 PMCID: PMC7346165 DOI: 10.3390/antiox9060551] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/21/2020] [Accepted: 06/23/2020] [Indexed: 12/18/2022] Open
Abstract
Oxidative stress (OS) appears to be an important determinant during the different stages of progression of Alzheimer’s Disease (AD). In particular, impaired antioxidant defense mechanisms, such as the decrease of glutathione (GSH) and nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2), a master regulator of antioxidant genes, including those for GSH, are associated with OS in the human AD brain. Among the neuropathological hallmarks of AD, the soluble oligomers of amyloid beta (Aβ) peptides seem to promote neuronal death through mitochondrial dysfunction and OS. In this regard, bifunctional antioxidants can exert a dual neuroprotective role by scavenging reactive oxygen species (ROS) directly and concomitant induction of antioxidant genes. In this study, among natural coumarins (esculetin, scopoletin, fraxetin and daphnetin), we demonstrated the ability of esculetin (ESC) to prevent and counteract ROS formation in neuronal SH-SY5Y cells, suggesting its profile as a bifunctional antioxidant. In particular, ESC increased the resistance of the SH-SY5Y cells against OS through the activation of Nrf2 and increase of GSH. In similar experimental conditions, ESC could also protect the SH-SY5Y cells from the OS and neuronal death evoked by oligomers of Aβ1–42 peptides. Further, the use of the inhibitors PD98059 and LY294002 also showed that Erk1/2 and Akt signaling pathways were involved in the neuroprotection mediated by ESC. These results encourage further research in AD models to explore the efficacy and safety profile of ESC as a novel neuroprotective agent.
Collapse
|
13
|
Morroni F, Sita G, Graziosi A, Ravegnini G, Molteni R, Paladini MS, Dias KST, dos Santos AF, Viegas C, Camps I, Pruccoli L, Tarozzi A, Hrelia P. PQM130, a Novel Feruloyl-Donepezil Hybrid Compound, Effectively Ameliorates the Cognitive Impairments and Pathology in a Mouse Model of Alzheimer's Disease. Front Pharmacol 2019; 10:658. [PMID: 31244664 PMCID: PMC6581760 DOI: 10.3389/fphar.2019.00658] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/21/2019] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is the most frequent type of dementia in older people. The complex nature of AD calls for the development of multitarget agents addressing key pathogenic processes. Donepezil, an acetylcholinesterase inhibitor, is a first-line acetylcholinesterase inhibitor used for the treatment of AD. Although several studies have demonstrated the symptomatic efficacy of donepezil treatment in AD patients, the possible effects of donepezil on the AD process are not yet known. In this study, a novel feruloyl-donepezil hybrid compound (PQM130) was synthesized and evaluated as a multitarget drug candidate against the neurotoxicity induced by Aβ1-42 oligomer (AβO) injection in mice. Interestingly, PQM130 had already shown anti-inflammatory activity in different in vivo models and neuroprotective activity in human neuronal cells. The intracerebroventricular (i.c.v.) injection of AβO in mice caused the increase of memory impairment, oxidative stress, neurodegeneration, and neuroinflammation. Instead, PQM130 (0.5-1 mg/kg) treatment after the i.c.v. AβO injection reduced oxidative damage and neuroinflammation and induced cell survival and protein synthesis through the modulation of glycogen synthase kinase 3β (GSK3β) and extracellular signal-regulated kinases (ERK1/2). Moreover, PQM130 increased brain plasticity and protected mice against the decline in spatial cognition. Even more interesting is that PQM130 modulated different pathways compared to donepezil, and it is much more effective in counteracting AβO damage. Therefore, our findings highlighted that PQM130 is a potent multi-functional agent against AD and could act as a promising neuroprotective compound for anti-AD drug development.
Collapse
Affiliation(s)
- Fabiana Morroni
- Department of Pharmacy and BioTechnology–FaBiT, Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Giulia Sita
- Department of Pharmacy and BioTechnology–FaBiT, Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Agnese Graziosi
- Department of Pharmacy and BioTechnology–FaBiT, Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Gloria Ravegnini
- Department of Pharmacy and BioTechnology–FaBiT, Alma Mater Studiorum–University of Bologna, Bologna, Italy
| | - Raffaella Molteni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Maria Serena Paladini
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | | | | | - Claudio Viegas
- Institute of Chemistry, Federal University of Alfenas, Alfenas, MG, Brazil
| | - Ihosvany Camps
- Institute of Exact Sciences, Federal University of Alfenas, Alfenas, MG, Brazil
| | - Letizia Pruccoli
- Department for Life Quality Studies-QuVi, Alma Mater Studiorum-University of Bologna, Rimini, Italy
| | - Andrea Tarozzi
- Department for Life Quality Studies-QuVi, Alma Mater Studiorum-University of Bologna, Rimini, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and BioTechnology–FaBiT, Alma Mater Studiorum–University of Bologna, Bologna, Italy
| |
Collapse
|
14
|
Maezawa I, Nguyen HM, Di Lucente J, Jenkins DP, Singh V, Hilt S, Kim K, Rangaraju S, Levey AI, Wulff H, Jin LW. Kv1.3 inhibition as a potential microglia-targeted therapy for Alzheimer's disease: preclinical proof of concept. Brain 2019; 141:596-612. [PMID: 29272333 DOI: 10.1093/brain/awx346] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 10/30/2017] [Indexed: 12/14/2022] Open
Abstract
Microglia significantly contribute to the pathophysiology of Alzheimer's disease but an effective microglia-targeted therapeutic approach is not yet available clinically. The potassium channels Kv1.3 and Kir2.1 play important roles in regulating immune cell functions and have been implicated by in vitro studies in the 'M1-like pro-inflammatory' or 'M2-like anti-inflammatory' state of microglia, respectively. We here found that amyloid-β oligomer-induced expression of Kv1.3 and Kir2.1 in cultured primary microglia. Likewise, ex vivo microglia acutely isolated from the Alzheimer's model 5xFAD mice co-expressed Kv1.3 and Kir2.1 as well as markers traditionally associated with M1 and M2 activation suggesting that amyloid-β oligomer induces a microglial activation state that is more complex than previously thought. Using the orally available, brain penetrant small molecule Kv1.3 blocker PAP-1 as a tool, we showed that pro-inflammatory and neurotoxic microglial responses induced by amyloid-β oligomer required Kv1.3 activity in vitro and in hippocampal slices. Since we further observed that Kv1.3 was highly expressed in microglia of transgenic Alzheimer's mouse models and human Alzheimer's disease brains, we hypothesized that pharmacological Kv1.3 inhibition could mitigate the pathology induced by amyloid-β aggregates. Indeed, treating APP/PS1 transgenic mice with a 5-month oral regimen of PAP-1, starting at 9 months of age, when the animals already manifest cognitive deficits and amyloid pathology, reduced neuroinflammation, decreased cerebral amyloid load, enhanced hippocampal neuronal plasticity, and improved behavioural deficits. The observed decrease in cerebral amyloid deposition was consistent with the in vitro finding that PAP-1 enhanced amyloid-β uptake by microglia. Collectively, these results provide proof-of-concept data to advance Kv1.3 blockers to Alzheimer's disease clinical trials.
Collapse
Affiliation(s)
- Izumi Maezawa
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, 2805 50th Street, Sacramento, CA 95817, USA
| | - Hai M Nguyen
- Department of Pharmacology, University of California Davis, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Jacopo Di Lucente
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, 2805 50th Street, Sacramento, CA 95817, USA
| | - David Paul Jenkins
- Department of Pharmacology, University of California Davis, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Vikrant Singh
- Department of Pharmacology, University of California Davis, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Silvia Hilt
- Department of Biochemistry and Molecular Medicine, University of California Davis, 2700 Stockton Blvd, Sacramento, CA 95817, USA
| | - Kyoungmi Kim
- Department of Public Health Sciences, University of California Davis, One Shields Avenue, Med Sci 1-C, Davis, CA 95616, USA
| | - Srikant Rangaraju
- Department of Neurology and Alzheimer's Disease Research Center, Emory University, 201 Dowman Drive, Atlanta, GA 30322, USA
| | - Allan I Levey
- Department of Neurology and Alzheimer's Disease Research Center, Emory University, 201 Dowman Drive, Atlanta, GA 30322, USA
| | - Heike Wulff
- Department of Pharmacology, University of California Davis, 451 Health Sciences Drive, Davis, CA 95616, USA
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, 2805 50th Street, Sacramento, CA 95817, USA.,Alzheimer's Disease Center, University of California Davis Medical Center, 4860 Y Street, Suite 3900, Sacramento, CA 95817, USA
| |
Collapse
|
15
|
He L, Jiang Y, Green J, Blevins H, Zhang S. Development of bivalent compounds as potential neuroprotectants for Alzheimer's disease. Bioorg Med Chem Lett 2019; 29:1957-1961. [PMID: 31153803 DOI: 10.1016/j.bmcl.2019.05.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/16/2019] [Accepted: 05/17/2019] [Indexed: 01/05/2023]
Abstract
In our efforts to further investigate the impact of the spacer and membrane anchor to the neuroprotective activities, a series of bivalent compounds that contain cholesterol and extended spacers were designed, synthesized and biologically characterized. Our results support previous studies that incorporation of a piperazine ring into the spacer significantly improved the protective potency of bivalent compounds in MC65 cell model. Spacer length beyond 21 atoms does not add further benefits with 21MO being the most potent one with an EC50 of 81.86 ± 11.91 nM. Our results also demonstrated that bivalent compound 21MO suppressed the production of mitochondria reactive oxygen species. Furthermore, our results confirmed that both of the spacer and membrane anchor moiety are essential to metal binding. Collectively, the results provide further evidence and information to guide optimization of such bivalent compounds as potential neuroprotectants for Alzheimer's disease.
Collapse
Affiliation(s)
- Liu He
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Yuqi Jiang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Jakob Green
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Hallie Blevins
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
16
|
Jin L, Lucente JD, Nguyen HM, Singh V, Singh L, Chavez M, Bushong T, Wulff H, Maezawa I. Repurposing the KCa3.1 inhibitor senicapoc for Alzheimer's disease. Ann Clin Transl Neurol 2019; 6:723-738. [PMID: 31019997 PMCID: PMC6469250 DOI: 10.1002/acn3.754] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 02/06/2019] [Accepted: 02/10/2019] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE Microglia play a pivotal role in the initiation and progression of Alzheimer's disease (AD). We here tested the therapeutic hypothesis that the Ca2+-activated potassium channel KCa3.1 constitutes a potential target for treating AD by reducing neuroinflammation. METHODS To determine if KCa3.1 is relevant to AD, we tested if treating cultured microglia or hippocampal slices with Aβ oligomer (AβO) activated KCa3.1 in microglia, and if microglial KCa3.1 was upregulated in 5xFAD mice and in human AD brains. The expression/activity of KCa3.1 was examined by qPCR, Western blotting, immunohistochemistry, and whole-cell patch-clamp. To investigate the role of KCa3.1 in AD pathology, we resynthesized senicapoc, a clinically tested KCa3.1 blocker, and determined its pharmacokinetic properties and its effect on microglial activation, Aβ deposition and hippocampal long-term potentiation (hLTP) in 5xFAD mice. RESULTS We found markedly enhanced microglial KCa3.1 expression/activity in brains of both 5xFAD mice and AD patients. In hippocampal slices, microglial KCa3.1 expression/activity was increased by AβO treatment, and its inhibition diminished the proinflammatory and hLTP-impairing activities of AβO. Senicapoc exhibited excellent brain penetrance and oral availability, and in 5xFAD mice, reduced neuroinflammation, decreased cerebral amyloid load, and enhanced hippocampal neuronal plasticity. INTERPRETATION Our results prompt us to propose repurposing senicapoc for AD clinical trials, as senicapoc has excellent pharmacological properties and was safe and well-tolerated in a prior phase-3 clinical trial for sickle cell anemia. Such repurposing has the potential to expedite the urgently needed new drug discovery for AD.
Collapse
Affiliation(s)
- Lee‐Way Jin
- Department of Pathology and Laboratory MedicineUniversity of California Davis Medical CenterSacramentoCalifornia
| | - Jacopo Di Lucente
- Department of Pathology and Laboratory MedicineUniversity of California Davis Medical CenterSacramentoCalifornia
| | - Hai M. Nguyen
- Department of PharmacologyUniversity of California DavisDavisCalifornia
| | - Vikrant Singh
- Department of PharmacologyUniversity of California DavisDavisCalifornia
| | - Latika Singh
- Department of PharmacologyUniversity of California DavisDavisCalifornia
| | - Monique Chavez
- Department of Pathology and Laboratory MedicineUniversity of California Davis Medical CenterSacramentoCalifornia
| | - Trevor Bushong
- Department of Pathology and Laboratory MedicineUniversity of California Davis Medical CenterSacramentoCalifornia
| | - Heike Wulff
- Department of PharmacologyUniversity of California DavisDavisCalifornia
| | - Izumi Maezawa
- Department of Pathology and Laboratory MedicineUniversity of California Davis Medical CenterSacramentoCalifornia
| |
Collapse
|
17
|
Carmona V, Martín-Aragón S, Goldberg J, Schubert D, Bermejo-Bescós P. Several targets involved in Alzheimer’s disease amyloidogenesis are affected by morin and isoquercitrin. Nutr Neurosci 2018; 23:575-590. [DOI: 10.1080/1028415x.2018.1534793] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Vanesa Carmona
- Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
- Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Sagrario Martín-Aragón
- Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
| | - Joshua Goldberg
- Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - David Schubert
- Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Paloma Bermejo-Bescós
- Departamento de Farmacología, Facultad de Farmacia, Universidad Complutense, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain
| |
Collapse
|
18
|
A Bifunctional Anti-Amyloid Blocks Oxidative Stress and the Accumulation of Intraneuronal Amyloid-Beta. Molecules 2018; 23:molecules23082010. [PMID: 30103547 PMCID: PMC6222334 DOI: 10.3390/molecules23082010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Revised: 08/04/2018] [Accepted: 08/08/2018] [Indexed: 12/30/2022] Open
Abstract
There is growing recognition regarding the role of intracellular amyloid beta (Aβ) in the Alzheimer’s disease process, which has been linked with aberrant signaling and the disruption of protein degradation mechanisms. Most notably, intraneuronal Aβ likely underlies the oxidative stress and mitochondrial dysfunction that have been identified as key elements of disease progression. In this study, we employed fluorescence imaging to explore the ability of a bifunctional small molecule to reduce aggregates of intracellular Aβ and attenuate oxidative stress. Structurally, this small molecule is comprised of a nitroxide spin label linked to an amyloidophilic fluorene and is known as spin-labeled fluorene (SLF). The effect of the SLF on intracellular Aβ accumulation and oxidative stress was measured in MC65 cells, a human neuronal cell line with inducible expression of the amyloid precursor protein and in the N2a neuronal cell line treated with exogenous Aβ. Super-resolution microscopy imaging showed SLF decreases the accumulation of intracellular Aβ. Confocal microscopy imaging of MC65 cells treated with a reactive oxygen species (ROS)-sensitive dye demonstrated SLF significantly reduces the intracellular Aβ-induced ROS signal. In order to determine the contributions of the separate SLF moieties to these protective activities, experiments were also carried out on cells with nitroxides lacking the Aβ targeting domain or fluorene derivatives lacking the nitroxide functionality. The findings support a synergistic effect of SLF in counteracting both the conformational toxicity of both endogenous and exogenous Aβ, its promotion of ROS, and Aβ metabolism. Furthermore, these studies demonstrate an intimate link between ROS production and Aβ oligomer formation.
Collapse
|
19
|
Morroni F, Sita G, Graziosi A, Turrini E, Fimognari C, Tarozzi A, Hrelia P. Neuroprotective Effect of Caffeic Acid Phenethyl Ester in A Mouse Model of Alzheimer's Disease Involves Nrf2/HO-1 Pathway. Aging Dis 2018; 9:605-622. [PMID: 30090650 PMCID: PMC6065293 DOI: 10.14336/ad.2017.0903] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/03/2017] [Indexed: 12/15/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive pathology, where dementia symptoms gradually worsen over a number of years. The hallmarks of AD, such as amyloid β-peptide (Aβ) in senile plaque and neurofibrillary tangles, are strongly intertwined with oxidative stress, which is considered one of the common effectors of the cascade of degenerative events. The endogenous nuclear factor erythroid 2-related factor 2 (Nrf2) is the "master regulator" of the antioxidant response and it is known as an indicator and regulator of oxidative stress. The present study aimed to determine the potential neuroprotective activity of caffeic acid phenethyl ester (CAPE), a polyphenolic compound abundant in honeybee, against the neurotoxicity of Aβ1-42 oligomers (AβO) in mice. An intracerebroventricular (i.c.v.) injection of AβO into the mouse brain triggered increased reactive oxygen species levels, neurodegeneration, neuroinflammation, and memory impairment. In contrast, the intraperitoneal administration of CAPE (10 mg/kg) after i.c.v. AβO-injection counteracted oxidative stress accompanied by an induction of Nrf2 and heme oxygenase-1 via the modulation of glycogen synthase kinase 3β in the hippocampus of mice. Additionally, CAPE treatment decreased AβO-induced neuronal apoptosis and neuroinflammation, and improved learning and memory, protecting mice against the decline in spatial cognition. Our findings demonstrate that CAPE could potentially be considered as a promising neuroprotective agent against progressive neurodegenerative diseases such as AD.
Collapse
Affiliation(s)
- Fabiana Morroni
- 1Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Giulia Sita
- 1Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Agnese Graziosi
- 1Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Eleonora Turrini
- 2Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 47900 Rimini, Italy
| | - Carmela Fimognari
- 2Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 47900 Rimini, Italy
| | - Andrea Tarozzi
- 2Department for Life Quality Studies, Alma Mater Studiorum, University of Bologna, 47900 Rimini, Italy
| | - Patrizia Hrelia
- 1Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
20
|
Morroni F, Sita G, Graziosi A, Turrini E, Fimognari C, Tarozzi A, Hrelia P. Protective Effects of 6-(Methylsulfinyl)hexyl Isothiocyanate on Aβ 1-42-Induced Cognitive Deficit, Oxidative Stress, Inflammation, and Apoptosis in Mice. Int J Mol Sci 2018; 19:E2083. [PMID: 30021941 PMCID: PMC6073905 DOI: 10.3390/ijms19072083] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/10/2018] [Accepted: 07/16/2018] [Indexed: 01/12/2023] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia among older people. Although soluble amyloid species are recognized triggers of the disease, no therapeutic approach is able to stop it. 6-(Methylsulfinyl)hexyl isothiocyanate (6-MSITC) is a major bioactive compound in Wasabia japonica, which is a typical Japanese pungent spice. Recently, in vivo and in vitro studies demonstrated that 6-MSITC has several biological properties. The aim of the present study was to investigate the neuroprotective activity of 6-MSITC in a murine AD model, induced by intracerebroventricular injection of β-amyloid oligomers (Aβ1-42O). The treatment with 6-MSITC started 1 h after the surgery for the next 10 days. Behavioral analysis showed that 6-MSITC ameliorated Aβ1-42O-induced memory impairments. The decrease of glutathione levels and increase of reactive oxygen species in hippocampal tissues following Aβ1-42O injection were reduced by 6-MSITC. Moreover, activation of caspases, increase of inflammatory factors, and phosphorylation of ERK and GSK3 were inhibited by 6-MSITC. These results highlighted an interesting neuroprotective activity of 6-MSITC, which was able to restore a physiological oxidative status, interfere positively with Nrf2-pathway, decrease apoptosis and neuroinflammation and contribute to behavioral recovery. Taken together, these findings demonstrated that 6-MSITC could be a promising complement for AD therapy.
Collapse
Affiliation(s)
- Fabiana Morroni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Irnerio 48, 40126 Bologna, Italy.
| | - Giulia Sita
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Irnerio 48, 40126 Bologna, Italy.
| | - Agnese Graziosi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Irnerio 48, 40126 Bologna, Italy.
| | - Eleonora Turrini
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d'Augusto, 237, 47900 Rimini, Italy.
| | - Carmela Fimognari
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d'Augusto, 237, 47900 Rimini, Italy.
| | - Andrea Tarozzi
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d'Augusto, 237, 47900 Rimini, Italy.
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Irnerio 48, 40126 Bologna, Italy.
| |
Collapse
|
21
|
He L, Boice A, Liu K, Yan X, Jiang Y, Zhang S. Design and characterization of bivalent compounds as potential neuroprotectants for Alzheimer's disease: Impact of the spacer on biological activity. Bioorg Med Chem Lett 2018; 28:1030-1036. [PMID: 29475586 DOI: 10.1016/j.bmcl.2018.02.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/10/2018] [Accepted: 02/13/2018] [Indexed: 11/15/2022]
Abstract
In our continuing efforts to develop bivalent compounds as potential neuroprotectants for Alzheimer's disease, a series of bivalent compounds that contain cholesterylamine and an extended spacer were synthesized and biologically characterized. Our results demonstrated that incorporation of a piperazine ring into the spacer composition significantly improved the protective potency in MC65 cell models. Our results also suggested that the optimal spacer length for such bivalent compounds ranges from 17 to 21 atoms, and further spacer extension beyond 21 atoms results no further optimization. Notably, incorporation of a piperazine ring into the spacer diminished the biometal chelating capacity for these bivalent compounds, thus suggesting structural flexibility of these compounds in interactions with metals. Collectively, the results provided valuable guidance to develop new bivalent compounds as neuroprotectants for Alzheimer's disease.
Collapse
Affiliation(s)
- Liu He
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Ashley Boice
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Kai Liu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Xing Yan
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Yuqi Jiang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA
| | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
22
|
He L, Jiang Y, Liu K, Gomez-Murcia V, Ma X, Torrecillas A, Chen Q, Zhu X, Lesnefsky E, Gomez-Fernandez JC, Xu B, Zhang S. Insights into the Impact of a Membrane-Anchoring Moiety on the Biological Activities of Bivalent Compounds As Potential Neuroprotectants for Alzheimer's Disease. J Med Chem 2018; 61:777-790. [PMID: 29271648 DOI: 10.1021/acs.jmedchem.7b01284] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Bivalent compounds anchoring in different manners to the membrane were designed and biologically characterized to understand the contribution of the anchor moiety to their biological activity as neuroprotectants for Alzheimer's disease. Our results established that the anchor moiety is essential, and we identified a preference for diosgenin, as evidenced by 17MD. Studies in primary neurons and mouse brain mitochondria also identified 17MD as exhibiting activity on neuritic outgrowth and the state 3 oxidative rate of glutamate while preserving the coupling capacity of the mitochondria. Significantly, our studies demonstrated that the integrated bivalent structure is essential to the observed biological activities. Further studies employing bivalent compounds as probes in a model membrane also revealed the influence of the anchor moiety on how they interact with the membrane. Collectively, our results suggest diosgenin to be an optimal anchor moiety, providing bivalent compounds with promising pharmacology that have potential applications for Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | - Victoria Gomez-Murcia
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary, University of Murcia , Murcia, 30080, Spain
| | - Xiaopin Ma
- Department of Pathology, Case Western Reserve University , Cleveland, Ohio 44106, United States
| | - Alejandro Torrecillas
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary, University of Murcia , Murcia, 30080, Spain
| | | | - Xiongwei Zhu
- Department of Pathology, Case Western Reserve University , Cleveland, Ohio 44106, United States
| | | | - Juan C Gomez-Fernandez
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary, University of Murcia , Murcia, 30080, Spain
| | - Bin Xu
- Department of Biochemistry, Virginia Polytechnic Institute and State University , Blacksburg, Virginia 24061, United States
| | | |
Collapse
|
23
|
Abstract
AIM Alzheimer's disease is a still untreatable multifaceted pathology, and drugs able to stop or reverse its progression are urgently needed. In this picture, the recent reformulation of the cholinergic hypothesis renewed the interest for acetylcholinesterase inhibitors. In this paper, a series of naturally inspired chalcone-based carbamates was designed to target cholinesterase enzymes and possibly generate fragments endowed with neuroprotective activity in situ. Results & methodology: All compounds presented in this study showed nanomolar potency for cholinesterase inhibition. Notably, fragment 11d also displayed an interesting neuroprotective profile. CONCLUSION These new derivatives are able to simultaneously modulate different key targets involved in Alzheimer's disease, and could be regarded as promising starting points for the development of disease-modifying drug candidates. [Formula: see text].
Collapse
|
24
|
Maezawa I, Zou B, Di Lucente J, Cao WS, Pascual C, Weerasekara S, Zhang M, Xie XS, Hua DH, Jin LW. The Anti-Amyloid-β and Neuroprotective Properties of a Novel Tricyclic Pyrone Molecule. J Alzheimers Dis 2017; 58:559-574. [PMID: 28482635 PMCID: PMC5438482 DOI: 10.3233/jad-161175] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/15/2017] [Indexed: 12/17/2022]
Abstract
There is an urgent unmet need for new therapeutics for Alzheimer's disease (AD), the most common cause of dementia in the elderly. Therapeutic approaches targeting amyloid-β (Aβ) and its downstream toxicities have become major strategies in AD drug development. We have taken a rational design approach and synthesized a class of tricyclic pyrone (TP) compounds that show anti-Aβ and other neuroprotective actions. The in vivo efficacy of a lead TP named CP2 to ameliorate AD-like pathologies has been shown in mouse models. Here we report the selection and initial characterization of a new lead TP70, which exhibited an anti-Aβ therapeutic index even higher than CP2. Moreover, TP70 was able to reduce oxidative stress, inhibit acyl-coenzyme A:cholesterol acyltransferase (ACAT), and upregulate the expression of ATP-binding cassette subfamily A, member 1 (ABCA1), actions considered neuroprotective in AD. TP70 further showed excellent pharmacokinetic properties, including brain penetration and oral availability. When administered to 5xFAD mice via intraperitoneal or oral route, TP70 enhanced the overall solubility and decreased the level of cerebral Aβ, including both fibrillary and soluble Aβ species. Interestingly, TP70 enhanced N-methyl-D-aspartate (NMDA) receptor-mediated excitatory post-synaptic potential (EPSP) in the hippocampal CA1 area, increased the magnitude of NMDA-dependent hippocampal long-term potentiation (LTP), a cellular model of learning and memory, and prevented the Aβ oligomer-impaired LTP. Significantly, a single dose of TP70 administered to aged 5xFAD mice was effective in mitigating the impaired LTP induction, recorded at 24 h after administration. Our results support a potential of TP70 in clinical development for AD in view of its synergistic neuroprotective actions, ability to positively modulate NMDA receptor-mediated hippocampal plasticity, and favorable pharmacokinetic properties in rodents.
Collapse
Affiliation(s)
- Izumi Maezawa
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, CA, USA
| | - Bende Zou
- AfaSci Research Laboratory, Redwood City, CA, USA
| | - Jacopo Di Lucente
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, CA, USA
| | | | | | | | - Man Zhang
- Department of Chemistry, Kansas State University, Manhattan, KS, USA
| | | | - Duy H. Hua
- Department of Chemistry, Kansas State University, Manhattan, KS, USA
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, CA, USA
- Alzheimer’s Disease Center, University of California Davis Medical Center, Sacramento, CA, USA
| |
Collapse
|
25
|
Morroni F, Sita G, Tarozzi A, Rimondini R, Hrelia P. Early effects of Aβ1-42 oligomers injection in mice: Involvement of PI3K/Akt/GSK3 and MAPK/ERK1/2 pathways. Behav Brain Res 2016; 314:106-15. [PMID: 27498145 DOI: 10.1016/j.bbr.2016.08.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/07/2016] [Accepted: 08/02/2016] [Indexed: 01/24/2023]
Abstract
Neuronal and synaptic loss are the best pathological correlates for memory decline in Alzheimer's disease (AD). Soluble beta-amyloid oligomers (AβO) are considered to putatively play a crucial role in the early synapse loss and cognitive impairment observed in AD. Evidence suggests that oxidative stress and apoptosis are involved in the mechanism of Aβ-induced neurotoxicity and AD pathogenesis. This study aimed to explore the molecular mechanisms that contribute to the early memory deficits induced by intracerebroventricular injection of AβO in mice. Ten days after a single AβO injection memory impairments were observed, as measured by Morris water maze and novel object recognition tests. Cognitive decline was associated with increased oxidative stress, caspase-9 activation, and decreased hippocampal synaptophysin immunoreactivity. Furthermore, GSH levels were significantly higher in AβO-injected mice than in sham mice, showing that a protective mechanism might develop due to oxidative stress. Additionally, AβO-induced toxicity was aligned with an increment of the activation of Akt and ERK1/2, and reduced activity of GSK3. These findings suggest that AβO injection triggers a cascade of events that mimic the key neuropathological hallmarks of AD. Aβ acute injection helps to better understand how this peptide impairs specific signaling pathways leading to synaptic and memory dysfunctions. Thus, this model is a valid tool for investigating AD and may suggest a new way to develop neuroprotective therapies at such early stages of the disease.
Collapse
Affiliation(s)
- Fabiana Morroni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Irnerio 48, 40126 Bologna, Italy.
| | - Giulia Sita
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Andrea Tarozzi
- Department for Life Quality Studies, Alma Mater Studiorum-University of Bologna, Corso d'Augusto, 237, 47900 Rimini, Italy
| | - Roberto Rimondini
- Department of Medical and Clinical Sciences, Alma Mater Studiorum-University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| | - Patrizia Hrelia
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, via Irnerio 48, 40126 Bologna, Italy
| |
Collapse
|
26
|
Uthaya Kumar US, Jothy SL, Kavitha N, Chen Y, Kanwar JR, Sasidharan S. Genoprotection and Cytotoxicity of Cassia surattensis Seed Extract on Vero Cell Evaluated by Comet and Cytotoxicity Assays. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/s40011-016-0761-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
27
|
Altman R, Ly S, Hilt S, Petrlova J, Maezawa I, Kálai T, Hideg K, Jin LW, Laurence TA, Voss JC. Protective spin-labeled fluorenes maintain amyloid beta peptide in small oligomers and limit transitions in secondary structure. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2015; 1854:1860-1870. [PMID: 26374940 DOI: 10.1016/j.bbapap.2015.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 08/20/2015] [Accepted: 09/11/2015] [Indexed: 10/23/2022]
Abstract
Alzheimer's disease is characterized by the presence of extracellular plaques comprised of amyloid beta (Aβ) peptides. Soluble oligomers of the Aβ peptide underlie a cascade of neuronal loss and dysfunction associated with Alzheimer's disease. Single particle analyses of Aβ oligomers in solution by fluorescence correlation spectroscopy (FCS) were used to provide real-time descriptions of how spin-labeled fluorenes (SLFs; bi-functional small molecules that block the toxicity of Aβ) prevent and disrupt oligomeric assemblies of Aβ in solution. Furthermore, the circular dichroism (CD) spectrum of untreated Aβ shows a continuous, progressive change over a 24-hour period, while the spectrum of Aβ treated with SLF remains relatively constant following initial incubation. These findings suggest the conformation of Aβ within the oligomer provides a complementary determinant of Aβ toxicity in addition to oligomer growth and size. Although SLF does not produce a dominant state of secondary structure in Aβ, it does induce a net reduction in beta secondary content compared to untreated samples of Aβ. The FCS results, combined with electron paramagnetic resonance spectroscopy and CD spectroscopy, demonstrate SLFs can inhibit the growth of Aβ oligomers and disrupt existing oligomers, while retaining Aβ as a population of smaller, yet largely disordered oligomers.
Collapse
Affiliation(s)
- Robin Altman
- Department of Biochemistry & Molecular Medicine, University of California Davis, Davis CA 95616, USA
| | - Sonny Ly
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore CA 94550, USA
| | - Silvia Hilt
- Department of Biochemistry & Molecular Medicine, University of California Davis, Davis CA 95616, USA
| | - Jitka Petrlova
- Department of Biochemistry & Molecular Medicine, University of California Davis, Davis CA 95616, USA
| | - Izumi Maezawa
- M.I.N.D. Institute and Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento CA 95817, USA
| | - Tamás Kálai
- Institute of Organic and Medicinal Chemistry, University of Pécs, H-7624 Pécs, Szigeti st. 12. Pécs, Hungary
| | - Kálmán Hideg
- Institute of Organic and Medicinal Chemistry, University of Pécs, H-7624 Pécs, Szigeti st. 12. Pécs, Hungary
| | - Lee-Way Jin
- M.I.N.D. Institute and Department of Pathology and Laboratory Medicine, University of California Davis, Sacramento CA 95817, USA
| | - Ted A Laurence
- Physical and Life Science Directorate, Lawrence Livermore National Laboratory, Livermore CA 94550, USA
| | - John C Voss
- Department of Biochemistry & Molecular Medicine, University of California Davis, Davis CA 95616, USA.
| |
Collapse
|
28
|
Kai T, Zhang L, Wang X, Jing A, Zhao B, Yu X, Zheng J, Zhou F. Tabersonine inhibits amyloid fibril formation and cytotoxicity of Aβ(1-42). ACS Chem Neurosci 2015; 6:879-88. [PMID: 25874995 DOI: 10.1021/acschemneuro.5b00015] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The misfolding and aggregation of amyloid beta (Aβ) peptides into amyloid fibrils are key events in the amyloid cascade hypothesis for the etiology of Alzheimer's disease (AD). Using thioflavin-T (ThT) fluorescence assay, atomic force microscopy, circular dichroism, size exclusion chromatography, surface plasmon resonance (SPR), and cytotoxicity tests, we demonstrate that tabersonine, an ingredient extracted from the bean of Voacanga africana, disrupts Aβ(1-42) aggregation and ameliorates Aβ aggregate-induced cytotoxicity. A small amount of tabersonine (e.g., 10 μM) can effectively inhibit the formation of Aβ(1-42) (e.g., 80 μM) fibrils or convert mature fibrils into largely innocuous amorphous aggregates. SPR results indicate that tabersonine binds to Aβ(1-42) oligomers in a dose-dependent way. Molecular dynamics (MD) simulations further confirm that tabersonine can bind to oligomers such as the pentamer of Aβ(1-42). Tabersonine preferentially interact with the β-sheet grooves of Aβ(1-42) containing aromatic and hydrophobic residues. The various binding sites and modes explain the diverse inhibitory effects of tabersonine on Aβ aggregation. Given that tabersonine is a natural product and a precursor for vincristine used in cancer chemotherapy, the biocompatibility and small size essential for permeating the blood-brain barrier make it a potential therapeutic drug candidate for treating AD.
Collapse
Affiliation(s)
- Tianhan Kai
- College
of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, P. R. China
| | - Lin Zhang
- College
of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan 410083, P. R. China
| | - Xiaoying Wang
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, California 90032, United States
| | - Aihua Jing
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, California 90032, United States
- College of Medical Technology and Engineering, Henan University of Science and Technology, Luoyang, Henan 471003, P. R. China
| | - Bingqing Zhao
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, California 90032, United States
| | - Xiang Yu
- Department
of Chemical and Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Jie Zheng
- Department
of Chemical and Biomolecular Engineering, The University of Akron, Akron, Ohio 44325, United States
| | - Feimeng Zhou
- Department of Chemistry and Biochemistry, California State University, Los Angeles, Los Angeles, California 90032, United States
| |
Collapse
|
29
|
Izzo NJ, Staniszewski A, To L, Fa M, Teich AF, Saeed F, Wostein H, Walko T, Vaswani A, Wardius M, Syed Z, Ravenscroft J, Mozzoni K, Silky C, Rehak C, Yurko R, Finn P, Look G, Rishton G, Safferstein H, Miller M, Johanson C, Stopa E, Windisch M, Hutter-Paier B, Shamloo M, Arancio O, LeVine H, Catalano SM. Alzheimer's therapeutics targeting amyloid beta 1-42 oligomers I: Abeta 42 oligomer binding to specific neuronal receptors is displaced by drug candidates that improve cognitive deficits. PLoS One 2014; 9:e111898. [PMID: 25390368 PMCID: PMC4229098 DOI: 10.1371/journal.pone.0111898] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 10/02/2014] [Indexed: 01/09/2023] Open
Abstract
Synaptic dysfunction and loss caused by age-dependent accumulation of synaptotoxic beta amyloid (Abeta) 1-42 oligomers is proposed to underlie cognitive decline in Alzheimer's disease (AD). Alterations in membrane trafficking induced by Abeta oligomers mediates reduction in neuronal surface receptor expression that is the basis for inhibition of electrophysiological measures of synaptic plasticity and thus learning and memory. We have utilized phenotypic screens in mature, in vitro cultures of rat brain cells to identify small molecules which block or prevent the binding and effects of Abeta oligomers. Synthetic Abeta oligomers bind saturably to a single site on neuronal synapses and induce deficits in membrane trafficking in neuronal cultures with an EC50 that corresponds to its binding affinity. The therapeutic lead compounds we have found are pharmacological antagonists of Abeta oligomers, reducing the binding of Abeta oligomers to neurons in vitro, preventing spine loss in neurons and preventing and treating oligomer-induced deficits in membrane trafficking. These molecules are highly brain penetrant and prevent and restore cognitive deficits in mouse models of Alzheimer's disease. Counter-screening these compounds against a broad panel of potential CNS targets revealed they are highly potent and specific ligands of the sigma-2/PGRMC1 receptor. Brain concentrations of the compounds corresponding to greater than 80% receptor occupancy at the sigma-2/PGRMC1 receptor restore cognitive function in transgenic hAPP Swe/Ldn mice. These studies demonstrate that synthetic and human-derived Abeta oligomers act as pharmacologically-behaved ligands at neuronal receptors--i.e. they exhibit saturable binding to a target, they exert a functional effect related to their binding and their displacement by small molecule antagonists blocks their functional effect. The first-in-class small molecule receptor antagonists described here restore memory to normal in multiple AD models and sustain improvement long-term, representing a novel mechanism of action for disease-modifying Alzheimer's therapeutics.
Collapse
Affiliation(s)
- Nicholas J. Izzo
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Agnes Staniszewski
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Lillian To
- Stanford University Medical School Behavioral and Functional Neuroscience Laboratory, Palo Alto, California, United States of America
| | - Mauro Fa
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Andrew F. Teich
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Faisal Saeed
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Harrison Wostein
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Thomas Walko
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Anisha Vaswani
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Meghan Wardius
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Zanobia Syed
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Jessica Ravenscroft
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Kelsie Mozzoni
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Colleen Silky
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Courtney Rehak
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Raymond Yurko
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Patricia Finn
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Gary Look
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Gilbert Rishton
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Hank Safferstein
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Miles Miller
- Department of Pathology and Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Conrad Johanson
- Department of Pathology and Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Edward Stopa
- Department of Pathology and Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | | | | | - Mehrdad Shamloo
- Stanford University Medical School Behavioral and Functional Neuroscience Laboratory, Palo Alto, California, United States of America
| | - Ottavio Arancio
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Harry LeVine
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Susan M. Catalano
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
30
|
Prior M, Chiruta C, Currais A, Goldberg J, Ramsey J, Dargusch R, Maher PA, Schubert D. Back to the future with phenotypic screening. ACS Chem Neurosci 2014; 5:503-13. [PMID: 24902068 DOI: 10.1021/cn500051h] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
There are no disease-modifying drugs for any old age associated neurodegenerative disease or stroke. This is at least in part due to the failure of drug developers to recognize that the vast majority of neurodegenerative diseases arise from a confluence of multiple toxic insults that accumulate during normal aging and interact with genetic and environmental risk factors. Thus, it is unlikely that the current single target approach based upon rare dominant mutations or even a few preselected targets is going to yield useful drugs for these conditions. Therefore, the identification of drug candidates for neurodegeneration should be based upon their efficacy in phenotypic screening assays that reflect the biology of the aging brain, not a single, preselected target. It is argued here that this approach to drug discovery is the most likely to produce safe and effective drugs for neurodegenerative diseases.
Collapse
Affiliation(s)
- Marguerite Prior
- Cellular
Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037-1002, United States
| | - Chandramouli Chiruta
- Cellular
Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037-1002, United States
| | - Antonio Currais
- Cellular
Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037-1002, United States
| | - Josh Goldberg
- Cellular
Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037-1002, United States
| | - Justin Ramsey
- Cellular
Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037-1002, United States
| | - Richard Dargusch
- Cellular
Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037-1002, United States
| | - Pamela A. Maher
- Cellular
Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037-1002, United States
| | - David Schubert
- Cellular
Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037-1002, United States
| |
Collapse
|
31
|
Abstract
The accumulation of intracellular β amyloid (Aβ) may be one of the factors leading to neuronal cell death in Alzheimer's disease (AD). Using a pyrazole called CNB-001, which was selected for its ability to reduce intracellular Aβ, we show that the activation of the eIF2α/ATF4 arm of the unfolded protein response is sufficient to degrade aggregated intracellular Aβ. CNB-001 is a potent inhibitor of 5-lipoxygenase (5-LOX), decreases 5-LOX expression, and increases proteasome activity. 5-LOX inhibition induces eIF2α and PERK (protein kinase R-like extracellular signal-regulated kinase) phosphorylation, and HSP90 and ATF4 levels. When fed to AD transgenic mice, CNB-001 also increases eIF2α phosphorylation and HSP90 and ATF4 levels, and limits the accumulation of soluble Aβ and ubiquitinated aggregated proteins. Finally, CNB-001 maintains the expression of synapse-associated proteins and improves memory. Therefore, 5-LOX metabolism is a key element in the promotion of endoplasmic reticulum dysfunction, and its inhibition under conditions of stress is sufficient to reduce proteotoxicity both in vivo and in vitro.
Collapse
|
32
|
Pokhrel L, Maezawa I, Nguyen TDT, Chang KO, Jin LW, Hua DH. Inhibition of Acyl-CoA: cholesterol acyltransferase (ACAT), overexpression of cholesterol transporter gene, and protection of amyloid β (Aβ) oligomers-induced neuronal cell death by tricyclic pyrone molecules. J Med Chem 2012; 55:8969-73. [PMID: 23025824 DOI: 10.1021/jm3012189] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A major effort in Alzheimer's disease therapeutic development has targeted Aβ and downstream events. We have synthesized a small library of tricyclic pyrone compounds. Their protective action in MC65 cells and inhibition of ACAT along with the upregulation of cholesterol transporter gene were investigated. Five active compounds exhibited potencies in the nanomolar ranges. The multiple effects of the compounds on Aβ and cellular cholesterol pathways could be potential mechanisms underlying the protective effects in vivo.
Collapse
Affiliation(s)
- Laxman Pokhrel
- Department of Chemistry, 213 CBC Building, Kansas State University, Manhattan, Kansas 66503, USA
| | | | | | | | | | | |
Collapse
|
33
|
Chang CC, Chen CN, Liao WL, Lo TY, Lei SL, Mai FD. Observation of the absorption of the In-DTPA-hexa-lactoside (IDHL) and its distribution in vitro
by ICP-MS and TOF-SIMS. SURF INTERFACE ANAL 2012. [DOI: 10.1002/sia.5030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Chun-Chao Chang
- Division of Gastroenterology and Hepatology, Department of Internal Medicine; Taipei Medical University Hospital; Taipei 11031 Taiwan
- Department of Internal Medicine, School of Medicine; Taipei Medical University; Taipei 11031 Taiwan
| | - Chun-Nan Chen
- Division of Gastroenterology and Hepatology; Department of Internal Medicine, Wan Fang Hospital; Taipei 11031 Taiwan
| | - Wan-Lu Liao
- Graduate Institute of Medical Science; Taipei Medical University; Taipei 11031 Taiwan
| | - Tsui-Yun Lo
- Department of Biochemistry, School of Medicine; Taipei Medical University; Taipei 11031 Taiwan
- Biomedical Mass Imaging Research Center; Taipei Medical University; Taipei 11031 Taiwan
| | - Shiou-Ling Lei
- Department of Chemistry; National Tsing Hua University; Hsinchu 30047 Taiwan
| | - Fu-Der Mai
- Graduate Institute of Medical Science; Taipei Medical University; Taipei 11031 Taiwan
- Department of Biochemistry, School of Medicine; Taipei Medical University; Taipei 11031 Taiwan
- Biomedical Mass Imaging Research Center; Taipei Medical University; Taipei 11031 Taiwan
| |
Collapse
|
34
|
Petrlova J, Kálai T, Maezawa I, Altman R, Harishchandra G, Hong HS, Bricarello DA, Parikh AN, Lorigan GA, Jin LW, Hideg K, Voss JC. The influence of spin-labeled fluorene compounds on the assembly and toxicity of the aβ peptide. PLoS One 2012; 7:e35443. [PMID: 22558151 PMCID: PMC3340382 DOI: 10.1371/journal.pone.0035443] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 03/16/2012] [Indexed: 11/18/2022] Open
Abstract
Background The deposition and oligomerization of amyloid β (Aβ) peptide plays a key role in the pathogenesis of Alzheimer's disease (AD). Aβ peptide arises from cleavage of the membrane-associated domain of the amyloid precursor protein (APP) by β and γ secretases. Several lines of evidence point to the soluble Aβ oligomer (AβO) as the primary neurotoxic species in the etiology of AD. Recently, we have demonstrated that a class of fluorene molecules specifically disrupts the AβO species. Methodology/Principal Findings To achieve a better understanding of the mechanism of action of this disruptive ability, we extend the application of electron paramagnetic resonance (EPR) spectroscopy of site-directed spin labels in the Aβ peptide to investigate the binding and influence of fluorene compounds on AβO structure and dynamics. In addition, we have synthesized a spin-labeled fluorene (SLF) containing a pyrroline nitroxide group that provides both increased cell protection against AβO toxicity and a route to directly observe the binding of the fluorene to the AβO assembly. We also evaluate the ability of fluorenes to target multiple pathological processes involved in the neurodegenerative cascade, such as their ability to block AβO toxicity, scavenge free radicals and diminish the formation of intracellular AβO species. Conclusions Fluorene modified with pyrroline nitroxide may be especially useful in counteracting Aβ peptide toxicity, because they posses both antioxidant properties and the ability to disrupt AβO species.
Collapse
Affiliation(s)
- Jitka Petrlova
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, California, United States of America
| | - Tamás Kálai
- Institute of Organic and Medicinal Chemistry, University of Pécs, Pécs, Hungary
| | - Izumi Maezawa
- Laboratory Medicine, Department of Pathology, M.I.N.D. Institute, Miami University, Oxford, Ohio, United States of America
| | - Robin Altman
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, California, United States of America
| | - Ghimire Harishchandra
- Department of Chemistry and Biochemistry, Miami University, Oxford, Ohio, United States of America
| | - Hyun-Seok Hong
- Laboratory Medicine, Department of Pathology, M.I.N.D. Institute, Miami University, Oxford, Ohio, United States of America
| | - Daniel A. Bricarello
- Department of Applied Science, University of California Davis, Davis, California, United States of America
| | - Atul N. Parikh
- Department of Applied Science, University of California Davis, Davis, California, United States of America
| | - Gary A. Lorigan
- Department of Chemistry and Biochemistry, Miami University, Oxford, Ohio, United States of America
| | - Lee-Way Jin
- Laboratory Medicine, Department of Pathology, M.I.N.D. Institute, Miami University, Oxford, Ohio, United States of America
| | - Kálmán Hideg
- Institute of Organic and Medicinal Chemistry, University of Pécs, Pécs, Hungary
| | - John C. Voss
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, California, United States of America
- * E-mail:
| |
Collapse
|
35
|
Horiuchi M, Maezawa I, Itoh A, Wakayama K, Jin LW, Itoh T, DeCarli C. Amyloid β1-42 oligomer inhibits myelin sheet formation in vitro. Neurobiol Aging 2012; 33:499-509. [PMID: 20594620 PMCID: PMC3013291 DOI: 10.1016/j.neurobiolaging.2010.05.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 04/30/2010] [Accepted: 05/09/2010] [Indexed: 01/26/2023]
Abstract
Accumulating evidence indicates that white matter degeneration contributes to the neural disconnections that underlie Alzheimer's disease pathophysiology. Although this white matter degeneration is partly attributable to axonopathy associated with neuronal degeneration, amyloid β (Aβ) protein-mediated damage to oligodendrocytes could be another mechanism. To test this hypothesis, we studied effects of soluble Aβ in oligomeric form on survival and differentiation of cells of the oligodendroglial lineage using highly purified oligodendroglial cultures from rats at different developmental stages. Aβ oligomer at 10 μM or higher reduced survival of mature oligodendrocytes, whereas oligodendroglial progenitor cells (OPCs) were relatively resistant to the Aβ oligomer-mediated cytotoxicity. Further study revealed that Aβ oligomer even at 1 μM accelerated 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) formazan exocytosis in mature oligodendrocytes, and, more significantly, inhibited myelin sheet formation after induction of in vitro differentiation of OPCs. These results imply a novel pathogenetic mechanism underlying Aβ oligomer-mediated white matter degeneration, which could impair myelin maintenance and remyelination by adult OPCs, resulting in accumulating damage to myelinating axons thereby contributing to neural disconnections.
Collapse
Affiliation(s)
- Makoto Horiuchi
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA, United States
| | - Izumi Maezawa
- M.I.N.D. Institute and Department of Pathology, Department of Internal Medicine, University of California Davis Cancer Center, University of California Davis, Sacramento, CA, United States
| | - Aki Itoh
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA, United States
| | - Kouji Wakayama
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA, United States
| | - Lee-Way Jin
- M.I.N.D. Institute and Department of Pathology, Department of Internal Medicine, University of California Davis Cancer Center, University of California Davis, Sacramento, CA, United States
| | - Takayuki Itoh
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, CA, United States
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA, United States
| | - Charles DeCarli
- Department of Neurology, University of California Davis, School of Medicine, Sacramento, CA, United States
| |
Collapse
|
36
|
Lü L, Zhang L, Wai MSM, Yew DTW, Xu J. Exocytosis of MTT formazan could exacerbate cell injury. Toxicol In Vitro 2012; 26:636-44. [PMID: 22401948 DOI: 10.1016/j.tiv.2012.02.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2010] [Revised: 10/29/2011] [Accepted: 02/20/2012] [Indexed: 01/01/2023]
Abstract
MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] method is one of the most widely used methods to analyze cell proliferation and viability. It is taken up through endocytosis and is reduced by mitochondrial enzymes as well as endosomal/lysosomal compartments, then is transported to cell surfaces to form needle-like MTT formazans; however the effect of MTT itself still remains elusive. Our objective was to investigate the direct effects of MTT on in vitro SH-SY5Y cells. Results showed that the endocytosis of MTT did not cause obvious lesion and induce cell death, but the metabolism and exocytosis of MTT could dramatically damage cells. Our results also indicated that MTT could activate apoptosis related factors such as caspase-8, caspase-3 or accelerate the leakage of cell contents after the appearance of MTT formazan crystals. The present data suggest MTT method should be carefully chosen; otherwise the cell viability would be underestimated and incomparable.
Collapse
Affiliation(s)
- Lanhai Lü
- Department of Anatomy, Zhongshan School of Medicine, Sun Yat-sen University, Northern Campus, Guangzhou, Guangdong, China
| | | | | | | | | |
Collapse
|
37
|
LeVine H, Lampe L, Abdelmoti L, Augelli-Szafran CE. Dihydroxybenzoic acid isomers differentially dissociate soluble biotinyl-Aβ(1-42) oligomers. Biochemistry 2011; 51:307-15. [PMID: 22129351 DOI: 10.1021/bi201288x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Polyphenolic compounds including a number of natural products such as resveratrol, curcumin, catechin derivatives, and nordihydroguaiaretic acid have effects on the assembly of Aβ fibrils and oligomers as well as on fibril morphology. Based on a lead structure obtained from a screen of a small molecule diversity library, simple benzoic acid derivatives distinguished by the number and position of hydroxyls on the aromatic ring displayed different abilities to dissociate preformed biotinyl-Aβ(1-42) oligomers. The 2,3-, 2,5-, and 3,4-dihydroxybenzoic acid (DHBA) isomers were active oligomer dissociators. The remaining DHBA isomers and the monohydroxy and unsubstituted benzoic acids were inactive and did not compete with the active compounds to block oligomer dissociation. None of the compounds blocked oligomer assembly, indicating that they do not interact with monomeric Aβ to shift the oligomer-monomer equilibrium. Dissociating activity was not associated with quinone redox cycling capacity of the compounds. Gallic acid (3,4,5-trihydroxybenzoic acid) stabilized biotinyl-Aβ(1-42) oligomers against intrinsic dissociation and blocked the effects of the active dissociators, independent of the concentration of dissociator. A model for the mechanism of action of the DHBA dissociators proposes that these compounds destabilize oligomer structure promoting progressive monomer dissociation rather than fissioning oligomers into smaller, but still macromolecular, species. Gallic acid blocks dissociation by stabilizing oligomers against this process.
Collapse
Affiliation(s)
- Harry LeVine
- Department of Cellular and Molecular Biochemistry, Center on Aging, Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40506, United States.
| | | | | | | |
Collapse
|
38
|
Abstract
Amyloid-beta (Aβ) aggregates are the main constituent of senile plaques, the histological hallmark of Alzheimer's disease. Aβ molecules form β-sheet containing structures that assemble into a variety of polymorphic oligomers, protofibers, and fibers that exhibit a range of lifetimes and cellular toxicities. This polymorphic nature of Aβ has frustrated its biophysical characterization, its structural determination, and our understanding of its pathological mechanism. To elucidate Aβ polymorphism in atomic detail, we determined eight new microcrystal structures of fiber-forming segments of Aβ. These structures, all of short, self-complementing pairs of β-sheets termed steric zippers, reveal a variety of modes of self-association of Aβ. Combining these atomic structures with previous NMR studies allows us to propose several fiber models, offering molecular models for some of the repertoire of polydisperse structures accessible to Aβ. These structures and molecular models contribute fundamental information for understanding Aβ polymorphic nature and pathogenesis.
Collapse
|
39
|
Landau M, Sawaya MR, Faull KF, Laganowsky A, Jiang L, Sievers SA, Liu J, Barrio JR, Eisenberg D. Towards a pharmacophore for amyloid. PLoS Biol 2011; 9:e1001080. [PMID: 21695112 PMCID: PMC3114762 DOI: 10.1371/journal.pbio.1001080] [Citation(s) in RCA: 164] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Accepted: 04/29/2011] [Indexed: 12/30/2022] Open
Abstract
Diagnosing and treating Alzheimer's and other diseases associated with amyloid fibers remains a great challenge despite intensive research. To aid in this effort, we present atomic structures of fiber-forming segments of proteins involved in Alzheimer's disease in complex with small molecule binders, determined by X-ray microcrystallography. The fiber-like complexes consist of pairs of β-sheets, with small molecules binding between the sheets, roughly parallel to the fiber axis. The structures suggest that apolar molecules drift along the fiber, consistent with the observation of nonspecific binding to a variety of amyloid proteins. In contrast, negatively charged orange-G binds specifically to lysine side chains of adjacent sheets. These structures provide molecular frameworks for the design of diagnostics and drugs for protein aggregation diseases. The devastating and incurable dementia known as Alzheimer's disease affects the thinking, memory, and behavior of dozens of millions of people worldwide. Although amyloid fibers and oligomers of two proteins, tau and amyloid-β, have been identified in association with this disease, the development of diagnostics and therapeutics has proceeded to date in a near vacuum of information about their structures. Here we report the first atomic structures of small molecules bound to amyloid. These are of the dye orange-G, the natural compound curcumin, and the Alzheimer's diagnostic compound DDNP bound to amyloid-like segments of tau and amyloid-β. The structures reveal the molecular framework of small-molecule binding, within cylindrical cavities running along the β-spines of the fibers. Negatively charged orange-G wedges into a specific binding site between two sheets of the fiber, combining apolar binding with electrostatic interactions, whereas uncharged compounds slide along the cavity. We observed that different amyloid polymorphs bind different small molecules, revealing that a cocktail of compounds may be required for future amyloid therapies. The structures described here start to define the amyloid pharmacophore, opening the way to structure-based design of improved diagnostics and therapeutics.
Collapse
Affiliation(s)
- Meytal Landau
- Howard Hughes Medical Institute, UCLA-DOE Institute for Genomics and Proteomics, Departments of Biological Chemistry and Chemistry and Biochemistry, University of California, Los Angeles, California, United States of America
| | - Michael R. Sawaya
- Howard Hughes Medical Institute, UCLA-DOE Institute for Genomics and Proteomics, Departments of Biological Chemistry and Chemistry and Biochemistry, University of California, Los Angeles, California, United States of America
| | - Kym F. Faull
- The Pasarow Mass Spectrometry Laboratory, The NPI-Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Arthur Laganowsky
- Howard Hughes Medical Institute, UCLA-DOE Institute for Genomics and Proteomics, Departments of Biological Chemistry and Chemistry and Biochemistry, University of California, Los Angeles, California, United States of America
| | - Lin Jiang
- Howard Hughes Medical Institute, UCLA-DOE Institute for Genomics and Proteomics, Departments of Biological Chemistry and Chemistry and Biochemistry, University of California, Los Angeles, California, United States of America
| | - Stuart A. Sievers
- Howard Hughes Medical Institute, UCLA-DOE Institute for Genomics and Proteomics, Departments of Biological Chemistry and Chemistry and Biochemistry, University of California, Los Angeles, California, United States of America
| | - Jie Liu
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Jorge R. Barrio
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - David Eisenberg
- Howard Hughes Medical Institute, UCLA-DOE Institute for Genomics and Proteomics, Departments of Biological Chemistry and Chemistry and Biochemistry, University of California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
40
|
Petrlova J, Hong HS, Bricarello DA, Harishchandra G, Lorigan GA, Jin LW, Voss JC. A differential association of Apolipoprotein E isoforms with the amyloid-β oligomer in solution. Proteins 2011; 79:402-16. [PMID: 21069870 DOI: 10.1002/prot.22891] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The molecular pathogenesis of disorders arising from protein misfolding and aggregation is difficult to elucidate, involving a complex ensemble of intermediates, whose toxicity depends upon their state of progression along distinct processing pathways. To address the complex misfolding and aggregation that initiates the toxic cascade resulting in Alzheimer's disease (AD), we have developed a 2,2,6,6-tetramethylpiperidine-1-oxyl-4-amino-4-carboxylic acid spin-labeled amyloid-β (Aβ) peptide to observe its isoform-dependent interaction with the apoE protein. Although most individuals carry the E3 isoform of apoE, ∼15% of humans carry the E4 isoform, which is recognized as the most significant genetic determinant for Alzheimer's. ApoE is consistently associated with the amyloid plaque marker for AD. A vital question centers on the influence of the two predominant isoforms, E3 and E4, on Aβ peptide processing and hence Aβ toxicity. We used electron paramagnetic resonance (EPR) spectroscopy of incorporated spin labels to investigate the interaction of apoE with the toxic oligomeric species of Aβ in solution. EPR spectra of the spin-labeled side chain report on side chain and backbone dynamics as well as the spatial proximity of spins in an assembly. Our results indicate oligomer binding involves the C-terminal domain of apoE, with apoE3 reporting a much greater response through this conformational marker. Coupled with SPR binding measurements, apoE3 displays a higher affinity and capacity for the toxic Aβ oligomer. These findings support the hypothesis that apoE polymorphism and Alzheimer's risk can largely be attributed to the reduced ability of apoE4 to function as a clearance vehicle for the toxic form of Aβ.
Collapse
Affiliation(s)
- Jitka Petrlova
- Department of Biochemistry and Molecular Medicine, University of California Davis, California, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Anekonda TS, Wadsworth TL, Sabin R, Frahler K, Harris C, Petriko B, Ralle M, Woltjer R, Quinn JF. Phytic acid as a potential treatment for alzheimer's pathology: evidence from animal and in vitro models. J Alzheimers Dis 2011; 23:21-35. [PMID: 20930278 DOI: 10.3233/jad-2010-101287] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Alzheimer's disease (AD) causes progressive, age-dependent cortical and hippocampal dysfunction leading to abnormal intellectual capacity and memory. We propose a novel protective treatment for AD pathology with phytic acid (inositol hexakisphosphate), a phytochemical found in food grains and a key signaling molecule in mammalian cells. We evaluated the protective and beneficial effects of phytic acid against amyloid-β (Aβ) pathology in MC65 cells and the Tg2576 mouse model. In MC65 cells, 48-72-hour treatment with phytic acid provided complete protection against amyloid precursor protein-C-terminal fragment-induced cytotoxicity by attenuating levels of increased intracellular calcium, hydrogen peroxide, superoxide, Aβ oligomers, and moderately upregulated the expression of autophagy (beclin-1) protein. In a tolerance paradigm, wild type mice were treated with 2% phytic acid in drinking water for 70 days. Phytic acid was well tolerated. Ceruloplasmin activity, brain copper and iron levels, and brain superoxide dismutase and ATP levels were unaffected by the treatment. There was a significant increase in brain levels of cytochrome oxidase and a decrease in lipid peroxidation with phytic acid administration. In a treatment paradigm, 12-month old Tg2576 and wild type mice were treated with 2% phytic acid or vehicle for 6 months. Brain levels of copper, iron, and zinc were unaffected. The effects of phytic acid were modest on the expression of AβPP trafficking-associated protein AP180, autophagy-associated proteins (beclin-1, LC3B), sirtuin 1, the ratio of phosphorylated AMP-activated protein kinase (PAMPK) to AMPK, soluble Aβ1-40, and insoluble Aβ1-42. These results suggest that phytic acid may provide a viable treatment option for AD.
Collapse
Affiliation(s)
- Thimmappa S Anekonda
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Multitargeted drugs discovery: Balancing anti-amyloid and anticholinesterase capacity in a single chemical entity. Bioorg Med Chem Lett 2011; 21:2655-8. [DOI: 10.1016/j.bmcl.2010.12.093] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2010] [Revised: 12/17/2010] [Accepted: 12/20/2010] [Indexed: 01/06/2023]
|
43
|
Weidner AM, Housley M, Murphy MP, Levine H. Purified high molecular weight synthetic Aβ(1-42) and biological Aβ oligomers are equipotent in rapidly inducing MTT formazan exocytosis. Neurosci Lett 2011; 497:1-5. [PMID: 21504780 DOI: 10.1016/j.neulet.2011.03.082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 03/21/2011] [Accepted: 03/28/2011] [Indexed: 11/28/2022]
Abstract
Synthetic soluble Aβ oligomers are often used as a surrogate for biologic material in a number of model systems. We compared the activity of Aβ oligomers (synthetic and cell culture media derived) on the human SH-SY5Y neuroblastoma and C2C12 mouse myoblast cell lines in a novel, modified MTT assay. Separating oligomers from monomeric peptide by size exclusion chromatography produced effects at peptide concentrations approaching physiologic levels (10-100 nM). Purified oligomers, but not monomers or fibrils, elicited an increase of a detergent-insoluble form of MTT formazan within 2h as opposed to a control toxin (H(2)O(2)). This effect was comparable for biological and synthetic peptide in both cell types. Monomeric Aβ attenuated the effect of soluble oligomers. This study suggests that the activities of biological and synthetic oligomers are indistinguishable during early stages of Aβ oligomer-cell interaction.
Collapse
Affiliation(s)
- Adam M Weidner
- Department of Cellular and Molecular Biochemistry, Center on Aging, Center for Structural Biology, University of Kentucky, 800 S. Limestone Street, Lexington, KY 40536-0230, United States.
| | | | | | | |
Collapse
|
44
|
Anekonda TS, Quinn JF, Harris C, Frahler K, Wadsworth TL, Woltjer RL. L-type voltage-gated calcium channel blockade with isradipine as a therapeutic strategy for Alzheimer's disease. Neurobiol Dis 2011; 41:62-70. [PMID: 20816785 PMCID: PMC2982927 DOI: 10.1016/j.nbd.2010.08.020] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 08/18/2010] [Accepted: 08/25/2010] [Indexed: 12/31/2022] Open
Abstract
There is strong evidence that intracellular calcium dysregulation plays an important pathological role in Alzheimer's disease, and specifically that beta amyloid may induce increases in intracellular calcium and lead to neuronal cell dysfunction and death. Here we investigated the feasibility of modifying Alzheimer's pathology with the L-type voltage-gated calcium channel blockers verapamil, diltiazem, isradipine and nimodipine. All four compounds protected MC65 neuroblastoma cells from amyloid beta protein precursor C-terminal fragment (APP CTF)-induced neurotoxicity. Isradipine was the most potent blocker, preventing APP CTF neurotoxicity at nanomolar concentrations. Intracellular beta amyloid expression was associated with increased expression of Cav 1.2 calcium channels and increased intracellular calcium influx from the extracellular space. Despite the cytoprotection afforded by calcium channel blockers, amyloid beta oligomer formation was not suppressed. The mechanism of cell death in MC65 cells is appeared to be caspase-3 independent. With the goal of determining if there is sufficient experimental support to move forward with animal trials of isradipine, we determined its bioavailability in the triple transgenic mouse model of AD. Subcutaneous implantation of carrier-bound isradipine (3 μg/g/day) for 60 days resulted in nanomolar concentrations in both the plasma and brain. Taken together, our in vitro results support the theory that calcium blockers exert protective effects downstream of the effects of beta amyloid. Isradipine's neuroprotective effect at concentrations that are clinically relevant and achievable in vitro and in vivo suggests that this particular calcium blocking agent may have therapeutic value in the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Thimmappa S Anekonda
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Maezawa I, Zimin PI, Wulff H, Jin LW. Amyloid-beta protein oligomer at low nanomolar concentrations activates microglia and induces microglial neurotoxicity. J Biol Chem 2010; 286:3693-706. [PMID: 20971854 DOI: 10.1074/jbc.m110.135244] [Citation(s) in RCA: 223] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Neuroinflammation and associated neuronal dysfunction mediated by activated microglia play an important role in the pathogenesis of Alzheimer disease (AD). Microglia are activated by aggregated forms of amyloid-β protein (Aβ), usually demonstrated in vitro by stimulating microglia with micromolar concentrations of fibrillar Aβ, a major component of amyloid plaques in AD brains. Here we report that amyloid-β oligomer (AβO), at 5-50 nm, induces a unique pattern of microglia activation that requires the activity of the scavenger receptor A and the Ca(2+)-activated potassium channel KCa3.1. AβO treatment induced an activated morphological and biochemical profile of microglia, including activation of p38 MAPK and nuclear factor κB. Interestingly, although increasing nitric oxide (NO) production, AβO did not increase several proinflammatory mediators commonly induced by lipopolyliposaccharides or fibrillar Aβ, suggesting that AβO stimulates both common and divergent pathways of microglia activation. AβO at low nanomolar concentrations, although not neurotoxic, induced indirect, microglia-mediated damage to neurons in dissociated cultures and in organotypic hippocampal slices. The indirect neurotoxicity was prevented by (i) doxycycline, an inhibitor of microglia activation; (ii) TRAM-34, a selective KCa3.1 blocker; and (iii) two inhibitors of inducible NO synthase, indicating that KCa3.1 activity and excessive NO release are required for AβO-induced microglial neurotoxicity. Our results suggest that AβO, generally considered a neurotoxin, may more potently cause neuronal damage indirectly by activating microglia in AD.
Collapse
Affiliation(s)
- Izumi Maezawa
- Medical Investigation of Neurodevelopmental Disorders Institute, University of California, Davis, California 95618, USA
| | | | | | | |
Collapse
|
46
|
Kreutzmann P, Wolf G, Kupsch K. Minocycline recovers MTT-formazan exocytosis impaired by amyloid beta peptide. Cell Mol Neurobiol 2010; 30:979-84. [PMID: 20455019 PMCID: PMC11498743 DOI: 10.1007/s10571-010-9528-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 04/26/2010] [Indexed: 01/09/2023]
Abstract
Minocycline, a tetracycline antibiotic, has been reported to exert beneficial effects in models of Alzheimer's disease (AD). To characterize the mechanisms underlying the putative minocycline-related neuroprotection, we studied its effect in an in vitro model of AD. Primary hippocampal cultures were treated with β-amyloid peptide (Aβ) and cell viability was assessed by standard MTT-assay. Incubation with 10 μM Aβ for 24 h significantly inhibits cellular MTT-reduction without inducing morphological signs of enhanced cell death or increase in release of lactate dehydrogenase. This indicates that cell viability was not affected. The inhibition of MTT-reduction by Aβ was due to an acceleration of MTT-formazan exocytosis. Intriguingly, the Aβ-triggered increase in MTT-formazan exocytosis was abolished by co-treatment with minocycline. In vehicle-treated cells minocycline had no effect on formazan exocytosis. This hitherto unrecognized property of minocycline has to be noticed in the elucidation of the underlying mechanism of this promising neuroprotectant.
Collapse
Affiliation(s)
- Peter Kreutzmann
- Institute of Medical Neurobiology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, Magdeburg, Germany.
| | | | | |
Collapse
|
47
|
Zovo K, Helk E, Karafin A, Tõugu V, Palumaa P. Label-Free High-Throughput Screening Assay for Inhibitors of Alzheimer’s Amyloid-β Peptide Aggregation Based on MALDI MS. Anal Chem 2010; 82:8558-65. [DOI: 10.1021/ac101583q] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Kairit Zovo
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, Tallinn 12618, Estonia
| | - Eneken Helk
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, Tallinn 12618, Estonia
| | - Ann Karafin
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, Tallinn 12618, Estonia
| | - Vello Tõugu
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, Tallinn 12618, Estonia
| | - Peep Palumaa
- Department of Gene Technology, Tallinn University of Technology, Akadeemia tee 15, Tallinn 12618, Estonia
| |
Collapse
|
48
|
Single-cell mechanics provides a sensitive and quantitative means for probing amyloid-beta peptide and neuronal cell interactions. Proc Natl Acad Sci U S A 2010; 107:13872-7. [PMID: 20643929 DOI: 10.1073/pnas.1008341107] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
By using a highly sensitive technique of atomic force microscopy-based single-cell compression, the rigidity of cultured N2a and HT22 neuronal cells was measured as a function of amyloid-beta42 (Abeta42) protein treatment. Abeta42 oligomers led to significant cellular stiffening; for example, 90-360% higher force was required to reach 80% deformation for N2a cells. Disaggregated or fibrillar forms of Abeta42 showed much less change. These observations were explained by a combination of two factors: (i) incorporation of oligomer into cellular membrane, which resulted in an increase in the Young's modulus of the membrane from 0.9+/-0.4 to 1.85+/-0.75 MPa for N2a cells and from 1.73+/-0.90 to 5.5+/-1.4 MPa for HT22 cells, and (ii) an increase in intracellular osmotic pressure (e.g., from 7 to 40 Pa for N2a cells) through unregulated ion influx. These findings and measurements provide a deeper, more characteristic, and quantitative insight into interactions between cells and Abeta42 oligomers, which have been considered the prime suspect for initiating neuronal dysfunction in Alzheimer's disease.
Collapse
|
49
|
Tarozzi A, Morroni F, Merlicco A, Bolondi C, Teti G, Falconi M, Cantelli-Forti G, Hrelia P. Neuroprotective effects of cyanidin 3-O-glucopyranoside on amyloid beta (25–35) oligomer-induced toxicity. Neurosci Lett 2010; 473:72-6. [DOI: 10.1016/j.neulet.2010.02.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Revised: 01/19/2010] [Accepted: 02/02/2010] [Indexed: 11/26/2022]
|
50
|
Bulic B, Pickhardt M, Mandelkow EM, Mandelkow E. Tau protein and tau aggregation inhibitors. Neuropharmacology 2010; 59:276-89. [PMID: 20149808 DOI: 10.1016/j.neuropharm.2010.01.016] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 01/20/2010] [Accepted: 01/26/2010] [Indexed: 12/13/2022]
Abstract
Alzheimer disease is characterized by pathological aggregation of two proteins, tau and Abeta-amyloid, both of which are considered to be toxic to neurons. In this review we summarize recent advances on small molecule inhibitors of protein aggregation with emphasis on tau, with activities mediated by the direct interference of self-assembly. The inhibitors can be clustered in several compound classes according to their chemical structure, with subsequent description of the structure-activity relationships, showing that hydrophobic interactions are prevailing. The description is extended to the pharmacological profile of the compounds in order to evaluate their drug-likeness, with special attention to toxicity and bioavailability. The collected data indicate that following the improvements of the in vitro inhibitory potencies, the consideration of the in vivo pharmacokinetics is an absolute prerequisite for the development of compounds suitable for a transfer from bench to bedside.
Collapse
Affiliation(s)
- Bruno Bulic
- Center for Advanced European Studies and Research, Bonn, Germany.
| | | | | | | |
Collapse
|