1
|
Góral I, Wichur T, Sługocka E, Grygier P, Głuch-Lutwin M, Mordyl B, Honkisz-Orzechowska E, Szałaj N, Godyń J, Panek D, Zaręba P, Sarka A, Żmudzki P, Latacz G, Pustelny K, Bucki A, Czarna A, Menezes F, Więckowska A. Exploring Novel GSK-3β Inhibitors for Anti-Neuroinflammatory and Neuroprotective Effects: Synthesis, Crystallography, Computational Analysis, and Biological Evaluation. ACS Chem Neurosci 2024; 15:3181-3201. [PMID: 39158934 PMCID: PMC11378298 DOI: 10.1021/acschemneuro.4c00365] [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: 06/12/2024] [Revised: 07/31/2024] [Accepted: 07/31/2024] [Indexed: 08/20/2024] Open
Abstract
In the pathogenesis of Alzheimer's disease, the overexpression of glycogen synthase kinase-3β (GSK-3β) stands out due to its multifaced nature, as it contributes to the promotion of amyloid β and tau protein accumulation, as well as neuroinflammatory processes. Therefore, in the present study, we have designed, synthesized, and evaluated a new series of GSK-3β inhibitors based on the N-(pyridin-2-yl)cyclopropanecarboxamide scaffold. We identified compound 36, demonstrating an IC50 of 70 nM against GSK-3β. Subsequently, through crystallography studies and quantum mechanical analysis, we elucidated its binding mode and identified the structural features crucial for interactions with the active site of GSK-3β, thereby understanding its inhibitory potency. Compound 36 was effective in the cellular model of hyperphosphorylated tau-induced neurodegeneration, where it restored cell viability after okadaic acid treatment and showed anti-inflammatory activity in the LPS model, significantly reducing NO, IL-6, and TNF-α release. In ADME-tox in vitro studies, we confirmed the beneficial profile of 36, including high permeability in PAMPA (Pe equals 9.4) and high metabolic stability in HLMs as well as lack of significant interactions with isoforms of the CYP enzymes and lack of considerable cytotoxicity on selected cell lines (IC50 > 100 μM on HT-22 cells and 89.3 μM on BV-2 cells). Based on promising pharmacological activities and favorable ADME-tox properties, compound 36 may be considered a promising candidate for in vivo research as well as constitute a reliable starting point for further studies.
Collapse
Affiliation(s)
- Izabella Góral
- Department
of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
- Doctoral
School of Medical and Health Sciences, Jagiellonian
University Medical College, 16 Lazarza St., Krakow 31-530, Poland
| | - Tomasz Wichur
- Department
of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| | - Emilia Sługocka
- Department
of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
- Doctoral
School of Medical and Health Sciences, Jagiellonian
University Medical College, 16 Lazarza St., Krakow 31-530, Poland
- Malopolska
Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, Krakow 30-387, Poland
| | - Przemysław Grygier
- Malopolska
Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, Krakow 30-387, Poland
- Doctoral
School of Exact and Natural Sciences, Jagiellonian
University, Lojasiewicza 11, Krakow 30-348, Poland
| | - Monika Głuch-Lutwin
- Department
of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| | - Barbara Mordyl
- Department
of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| | - Ewelina Honkisz-Orzechowska
- Department
of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| | - Natalia Szałaj
- Department
of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| | - Justyna Godyń
- Department
of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| | - Dawid Panek
- Department
of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| | - Paula Zaręba
- Department
of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| | - Anna Sarka
- Department
of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| | - Paweł Żmudzki
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| | - Gniewomir Latacz
- Department
of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| | - Katarzyna Pustelny
- Department
of Physical Biochemistry, Faculty of Biochemistry, Biophysics and
Biotechnology, Jagiellonian University, Gronostajowa 7 St., Krakow 30-387, Poland
| | - Adam Bucki
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| | - Anna Czarna
- Malopolska
Centre of Biotechnology, Jagiellonian University, Gronostajowa 7a, Krakow 30-387, Poland
| | - Filipe Menezes
- Helmholtz
Munich, Molecular Targets and Therapeutics Center, Institute of Structural
Biology, Ingolstädter
Landstr. 1, Neuherberg 85764, Germany
| | - Anna Więckowska
- Department
of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., Krakow 30-688, Poland
| |
Collapse
|
2
|
Góral I, Wichur T, Sługocka E, Godyń J, Szałaj N, Zaręba P, Głuch-Lutwin M, Mordyl B, Panek D, Więckowska A. Connecting GSK-3β Inhibitory Activity with IKK-β or ROCK-1 Inhibition to Target Tau Aggregation and Neuroinflammation in Alzheimer's Disease-Discovery, In Vitro and In Cellulo Activity of Thiazole-Based Inhibitors. Molecules 2024; 29:2616. [PMID: 38893493 PMCID: PMC11173485 DOI: 10.3390/molecules29112616] [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: 04/29/2024] [Revised: 05/26/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
GSK-3β, IKK-β, and ROCK-1 kinases are implicated in the pathomechanism of Alzheimer's disease due to their involvement in the misfolding and accumulation of amyloid β (Aβ) and tau proteins, as well as inflammatory processes. Among these kinases, GSK-3β plays the most crucial role. In this study, we present compound 62, a novel, remarkably potent, competitive GSK-3β inhibitor (IC50 = 8 nM, Ki = 2 nM) that also exhibits additional ROCK-1 inhibitory activity (IC50 = 2.3 µM) and demonstrates anti-inflammatory and neuroprotective properties. Compound 62 effectively suppresses the production of nitric oxide (NO) and pro-inflammatory cytokines in the lipopolysaccharide-induced model of inflammation in the microglial BV-2 cell line. Furthermore, it shows neuroprotective effects in an okadaic-acid-induced tau hyperphosphorylation cell model of neurodegeneration. The compound also demonstrates the potential for further development, characterized by its chemical and metabolic stability in mouse microsomes and fair solubility.
Collapse
Affiliation(s)
- Izabella Góral
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Lazarza St., 31-530 Krakow, Poland
| | - Tomasz Wichur
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Emilia Sługocka
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Lazarza St., 31-530 Krakow, Poland
| | - Justyna Godyń
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Natalia Szałaj
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Paula Zaręba
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Monika Głuch-Lutwin
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (M.G.-L.); (B.M.)
| | - Barbara Mordyl
- Department of Pharmacobiology, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (M.G.-L.); (B.M.)
| | - Dawid Panek
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| | - Anna Więckowska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna St., 30-688 Krakow, Poland; (I.G.); (T.W.); (E.S.); (J.G.); (N.S.); (P.Z.); (D.P.)
| |
Collapse
|
3
|
Fan B, Lu F, Du WJ, Chen J, An XG, Wang RF, Li W, Song YL, Zha DJ, Chen FQ. PTEN inhibitor bisperoxovanadium protects against noise-induced hearing loss. Neural Regen Res 2023; 18:1601-1606. [PMID: 36571368 PMCID: PMC10075117 DOI: 10.4103/1673-5374.358606] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Studies have shown that phosphatase and tensin homolog deleted on chromosome ten (PTEN) participates in the regulation of cochlear hair cell survival. Bisperoxovanadium protects against neurodegeneration by inhibiting PTEN expression. However, whether bisperoxovanadium can protect against noise-induced hearing loss and the underlying mechanism remains unclear. In this study, we established a mouse model of noise-induced hearing loss by exposure to 105 dB sound for 2 hours. We found that PTEN expression was increased in the organ of Corti, including outer hair cells, inner hair cells, and lateral wall tissues. Intraperitoneal administration of bisperoxovanadium decreased the auditory threshold and the loss of cochlear hair cells and inner hair cell ribbons. In addition, noise exposure decreased p-PI3K and p-Akt levels. Bisperoxovanadium preconditioning or PTEN knockdown upregulated the activity of PI3K-Akt. Bisperoxovanadium also prevented H2O2-induced hair cell death by reducing mitochondrial reactive oxygen species generation in cochlear explants. These findings suggest that bisperoxovanadium reduces noise-induced hearing injury and reduces cochlear hair cell loss.
Collapse
Affiliation(s)
- Bei Fan
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Fei Lu
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Wei-Jia Du
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Jun Chen
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Xiao-Gang An
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Ren-Feng Wang
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Wei Li
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Yong-Li Song
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Ding-Jun Zha
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Fu-Quan Chen
- Department of Otolaryngology Head and Neck Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| |
Collapse
|
4
|
Hedna R, Kovacic H, Pagano A, Peyrot V, Robin M, Devred F, Breuzard G. Tau Protein as Therapeutic Target for Cancer? Focus on Glioblastoma. Cancers (Basel) 2022; 14:5386. [PMID: 36358803 PMCID: PMC9653627 DOI: 10.3390/cancers14215386] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 08/27/2023] Open
Abstract
Despite being extensively studied for several decades, the microtubule-associated protein Tau has not finished revealing its secrets. For long, Tau has been known for its ability to promote microtubule assembly. A less known feature of Tau is its capability to bind to cancer-related protein kinases, suggesting a possible role of Tau in modulating microtubule-independent cellular pathways that are associated with oncogenesis. With the intention of finding new therapeutic targets for cancer, it appears essential to examine the interaction of Tau with these kinases and their consequences. This review aims at collecting the literature data supporting the relationship between Tau and cancer with a particular focus on glioblastoma tumors in which the pathological significance of Tau remains largely unexplored. We will first treat this subject from a mechanistic point of view showing the pivotal role of Tau in oncogenic processes. Then, we will discuss the involvement of Tau in dysregulating critical pathways in glioblastoma. Finally, we will outline promising strategies to target Tau protein for the therapy of glioblastoma.
Collapse
Affiliation(s)
- Rayane Hedna
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Hervé Kovacic
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Alessandra Pagano
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Vincent Peyrot
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Maxime Robin
- Faculté de Pharmacie, Institut Méditerranéen de Biodiversité et Ecologie marine et continentale (IMBE), UMR 7263, CNRS, IRD 237, Aix-Marseille Université, 13005 Marseille, France
| | - François Devred
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| | - Gilles Breuzard
- Faculté des Sciences Médicales et Paramédicales, Institut de Neurophysiopathologie (INP), UMR 7051, CNRS, Aix Marseille Université, 13005 Marseille, France
| |
Collapse
|
5
|
Liu Y, Xu S, Cai Q, Li D, Li H, Yang W. In Vitro Interactions between Okadaic Acid and Rat Gut Microbiome. Mar Drugs 2022; 20:556. [PMID: 36135745 PMCID: PMC9500940 DOI: 10.3390/md20090556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Okadaic acid (OA) is a marine biotoxin associated with diarrhetic shellfish poisoning (DSP), posing some threat to human beings. The oral toxicity of OA is complex, and the mechanism of toxicity is not clear. The interaction between OA and gut microbiota may provide a reasonable explanation for the complex toxicity of OA. Due to the complex environment in vivo, an in vitro study may be better for the interactions between OA and gut microbiome. Here, we conducted an in vitro fermentation experiment of gut bacteria in the presence of 0-1000 nM OA. The remolding ability of OA on bacterial composition was investigated by 16S rDNA sequencing, and differential metabolites in fermentation system with different concentration of OA was detected by LC-MS/MS. We found that OA inhibited some specific bacterial genera but promoted others. In addition, eight possible metabolites of OA, including dinophysistoxin-2 (DTX-2), were detected in the fermentation system. The abundance of Faecalitalea was strongly correlated with the possible metabolites of OA, suggesting that Faecalitalea may be involved in the metabolism of OA in vitro. Our findings confirmed the direct interaction between OA and gut bacteria, which helps to reveal the metabolic process of OA and provide valuable evidence for elucidating the complex toxicity of OA.
Collapse
Affiliation(s)
| | | | | | - Dawei Li
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | | | - Weidong Yang
- Key Laboratory of Aquatic Eutrophication and Control of Harmful Algal Blooms of Guangdong Higher Education Institute, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
6
|
Tsamou M, Carpi D, Pistollato F, Roggen EL. Sporadic Alzheimer's Disease- and Neurotoxicity-Related microRNAs Affecting Key Events of Tau-Driven Adverse Outcome Pathway Toward Memory Loss. J Alzheimers Dis 2022; 86:1427-1457. [PMID: 35213375 DOI: 10.3233/jad-215434] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND A complex network of aging-related homeostatic pathways that are sensitive to further deterioration in the presence of genetic, systemic, and environmental risk factors, and lifestyle, is implicated in the pathogenesis of progressive neurodegenerative diseases, such as sporadic (late-onset) Alzheimer's disease (sAD). OBJECTIVE Since sAD pathology and neurotoxicity share microRNAs (miRs) regulating common as well as overlapping pathological processes, environmental neurotoxic compounds are hypothesized to exert a risk for sAD initiation and progression. METHODS Literature search for miRs associated with human sAD and environmental neurotoxic compounds was conducted. Functional miR analysis using PathDip was performed to create miR-target interaction networks. RESULTS The identified miRs were successfully linked to the hypothetical starting point and key events of the earlier proposed tau-driven adverse outcome pathway toward memory loss. Functional miR analysis confirmed most of the findings retrieved from literature and revealed some interesting findings. The analysis identified 40 miRs involved in both sAD and neurotoxicity that dysregulated processes governing the plausible adverse outcome pathway for memory loss. CONCLUSION Creating miR-target interaction networks related to pathological processes involved in sAD initiation and progression, and environmental chemical-induced neurotoxicity, respectively, provided overlapping miR-target interaction networks. This overlap offered an opportunity to create an alternative picture of the mechanisms underlying sAD initiation and early progression. Looking at initiation and progression of sAD from this new angle may open for new biomarkers and novel drug targets for sAD before the appearance of the first clinical symptoms.
Collapse
Affiliation(s)
- Maria Tsamou
- ToxGenSolutions (TGS), Maastricht, The Netherlands
| | - Donatella Carpi
- European Commission, Joint Research Centre (JRC), Ispra VA, Italy
| | | | | |
Collapse
|
7
|
Shati AA, El-Kott AF, Alkhateeb MA. Resolvin D1 prevents cadmium chloride-induced memory loss and hippocampal damage in rats by activation/upregulation of PTEN-induced suppression of PI3K/Akt/mTOR signaling pathway. Clin Exp Pharmacol Physiol 2021; 49:275-290. [PMID: 34570918 DOI: 10.1111/1440-1681.13596] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/16/2021] [Accepted: 09/24/2021] [Indexed: 12/14/2022]
Abstract
This study evaluated the protective effect of resolvin D1 (RVD1) against cadmium chloride (CdCl2 )-induced hippocampal damage and memory loss in rats and investigated if such protection is mediated by modulating the PTEN/PI3K/Akt/mTOR pathway. Adult male Wistar rats (n = 18/group) were divided as control, control + RVD1, CdCl2 , CdCl2 + RVD1 and CdCl2 + RVD1 + bpV(pic), a PTEN inhibitor. All treatments were conducted for 4 weeks. Resolvin D1 improved the memory function as measured by Morris water maze (MWM), preserved the structure of CA1 area of the hippocampus, and increased hippocampal levels of RVD1 in the CdCl2 -treated rats. Resolvin D1 also suppressed the generation of reactive oxygen species (ROS), tumour necrosis factor-α and interleukine-6 (IL-6), inhibited nuclear factor κB (NF-κB) p65, stimulated levels of glutathione (GSH), manganese superoxide dismutase (MnSOD), and Bcl2 but reduced the expression of Bax and cleaved caspase 3 in hippocampi of CdCl2 -treated rats. Concomitantly, it stimulated levels and activity of PTEN and reduced the phosphorylation (activation) of PI3K, Akt and mTOR in hippocampi of CdCl2 -treated rats. In conclusion, RVD1 attenuates CdCl2 -induced memory loss and hippocampal damage in rats mainly by activating PTEN-induced suppression of PI3K/Akt/mTOR, an effect that seems secondary to its' anti-oxidant and anti-inflammatory potential.
Collapse
Affiliation(s)
- Ali A Shati
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Attalla F El-Kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia.,Department of Zoology, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Mahmoud A Alkhateeb
- Department of Basic Medical Sciences, College of Medicine, King Saud University for Health Sciences, Riyadh, Saudi Arabia
| |
Collapse
|
8
|
Henriques AD, Machado-Silva W, Leite RE, Suemoto CK, Leite KR, Srougi M, Pereira AC, Jacob-Filho W, Nóbrega OT. Genome-wide profiling and predicted significance of post-mortem brain microRNA in Alzheimer’s disease. Mech Ageing Dev 2020; 191:111352. [DOI: 10.1016/j.mad.2020.111352] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 12/12/2022]
|
9
|
An N, Bassil K, Al Jowf GI, Steinbusch HWM, Rothermel M, de Nijs L, Rutten BPF. Dual-specificity phosphatases in mental and neurological disorders. Prog Neurobiol 2020; 198:101906. [PMID: 32905807 DOI: 10.1016/j.pneurobio.2020.101906] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 08/26/2020] [Accepted: 09/01/2020] [Indexed: 01/01/2023]
Abstract
The dual-specificity phosphatase (DUSP) family includes a heterogeneous group of protein phosphatases that dephosphorylate both phospho-tyrosine and phospho-serine/phospho-threonine residues within a single substrate. These protein phosphatases have many substrates and modulate diverse neural functions, such as neurogenesis, differentiation, and apoptosis. DUSP genes have furthermore been associated with mental disorders such as depression and neurological disorders such as Alzheimer's disease. Herein, we review the current literature on the DUSP family of genes concerning mental and neurological disorders. This review i) outlines the structure and general functions of DUSP genes, and ii) overviews the literature on DUSP genes concerning mental and neurological disorders, including model systems, while furthermore providing perspectives for future research.
Collapse
Affiliation(s)
- Ning An
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; European Graduate School of Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Katherine Bassil
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; European Graduate School of Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Ghazi I Al Jowf
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; College of Applied Medical Sciences, Department of Public Health, King Faisal University, Al-Ahsa, Saudi Arabia; European Graduate School of Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Harry W M Steinbusch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; European Graduate School of Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Markus Rothermel
- European Graduate School of Neuroscience, Maastricht University, Maastricht, the Netherlands; Department of Chemosensation - AG Neuromodulation, RWTH Aachen University, Aachen, Germany
| | - Laurence de Nijs
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; European Graduate School of Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Bart P F Rutten
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, the Netherlands; European Graduate School of Neuroscience, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
10
|
Rastogi M, Singh SK. Modulation of Type-I Interferon Response by hsa-miR-374b-5p During Japanese Encephalitis Virus Infection in Human Microglial Cells. Front Cell Infect Microbiol 2019; 9:291. [PMID: 31448245 PMCID: PMC6695837 DOI: 10.3389/fcimb.2019.00291] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/29/2019] [Indexed: 12/24/2022] Open
Abstract
Japanese Encephalitis virus (JEV) is a neurotropic ssRNA virus, belonging to the Flaviviridae family. JEV is one of the leading causes of the viral encephalitis in Southeast-Asian countries. JEV primarily infects neurons however, the microglial activation has been reported to further enhance the neuroinflammation and promote neuronal death. The PI3K/AKT pathway has been reported to play an important role in type-I interferon response via IRF3. Phosphatase and tensin homolog (PTEN), a negative regulator of PI3K/AKT pathway, participates in microglial polarization and neuroinflammation. The microRNAs are small non-coding endogenously expressed RNAs, which regulate the gene expression by binding at 3′ UTR of target gene. The human microglial cells were infected with JEV (JaOArS982 strain) and up-regulation of microRNA; hsa-miR-374b-5p was confirmed by qRT-PCR. The genes in PI3K/AKT pathway, over-expression and knock-down studies of hsa-miR-374b-5p with and without JEV infection were analyzed through immuno blotting. The regulatory role of hsa-miR-374b-5p on the expression of type-I interferon was determined by luciferase assays. JEV infection modulated the expression of hsa-miR-374b-5p and PI3K/AKT pathway via PTEN. The over-expression of hsa-miR-374b-5p suppressed the PTEN while up-regulated the AKT and IRF3 proteins, whereas, the knockdown rescued the PTEN expression and suppressed the AKT and IRF3 proteins. The modulation of hsa-miR-374b-5p regulated the type-I interferon response during JEV infection. In present study, we have shown the modulation of PTEN by hsa-miR-374b-5p, which regulated the PI3K/AKT/IRF3 axis in JEV infected microglial cells.
Collapse
Affiliation(s)
- Meghana Rastogi
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Sunit K Singh
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
11
|
Kuo YC, Rajesh R. Challenges in the treatment of Alzheimer’s disease: recent progress and treatment strategies of pharmaceuticals targeting notable pathological factors. Expert Rev Neurother 2019; 19:623-652. [DOI: 10.1080/14737175.2019.1621750] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yung-Chih Kuo
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan, Republic of China
| | - Rajendiran Rajesh
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan, Republic of China
| |
Collapse
|
12
|
Nitulescu GM, Van De Venter M, Nitulescu G, Ungurianu A, Juzenas P, Peng Q, Olaru OT, Grădinaru D, Tsatsakis A, Tsoukalas D, Spandidos DA, Margina D. The Akt pathway in oncology therapy and beyond (Review). Int J Oncol 2018; 53:2319-2331. [PMID: 30334567 PMCID: PMC6203150 DOI: 10.3892/ijo.2018.4597] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023] Open
Abstract
Protein kinase B (Akt), similar to many other protein kinases, is at the crossroads of cell death and survival, playing a pivotal role in multiple interconnected cell signaling mechanisms implicated in cell metabolism, growth and division, apoptosis suppression and angiogenesis. Akt protein kinase displays important metabolic effects, among which are glucose uptake in muscle and fat cells or the suppression of neuronal cell death. Disruptions in the Akt-regulated pathways are associated with cancer, diabetes, cardiovascular and neurological diseases. The regulation of the Akt signaling pathway renders Akt a valuable therapeutic target. The discovery process of Akt inhibitors using various strategies has led to the identification of inhibitors with great selectivity, low side-effects and toxicity. The usefulness of Akt emerges beyond cancer therapy and extends to other major diseases, such as diabetes, heart diseases, or neurodegeneration. This review presents key features of Akt structure and functions, and presents the progress of Akt inhibitors in regards to drug development, and their preclinical and clinical activity in regards to therapeutic efficacy and safety for patients.
Collapse
Affiliation(s)
- George Mihai Nitulescu
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Maryna Van De Venter
- Department of Biochemistry and Microbiology, Nelson Mandela University, Port Elizabeth 6031, South Africa
| | - Georgiana Nitulescu
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Anca Ungurianu
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Petras Juzenas
- Department of Pathology, Radiumhospitalet, Oslo University Hospital, 0379 Oslo, Norway
| | - Qian Peng
- Department of Pathology, Radiumhospitalet, Oslo University Hospital, 0379 Oslo, Norway
| | - Octavian Tudorel Olaru
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Daniela Grădinaru
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| | - Aristides Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Dimitris Tsoukalas
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Denisa Margina
- Faculty of Pharmacy, 'Carol Davila' University of Medicine and Pharmacy, 020956 Bucharest, Romania
| |
Collapse
|
13
|
Kumar M, Bansal N. Fasudil hydrochloride ameliorates memory deficits in rat model of streptozotocin-induced Alzheimer’s disease: Involvement of PI3-kinase, eNOS and NFκB. Behav Brain Res 2018; 351:4-16. [DOI: 10.1016/j.bbr.2018.05.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 05/07/2018] [Accepted: 05/24/2018] [Indexed: 12/13/2022]
|
14
|
Gandini A, Bartolini M, Tedesco D, Martinez-Gonzalez L, Roca C, Campillo NE, Zaldivar-Diez J, Perez C, Zuccheri G, Miti A, Feoli A, Castellano S, Petralla S, Monti B, Rossi M, Moda F, Legname G, Martinez A, Bolognesi ML. Tau-Centric Multitarget Approach for Alzheimer’s Disease: Development of First-in-Class Dual Glycogen Synthase Kinase 3β and Tau-Aggregation Inhibitors. J Med Chem 2018; 61:7640-7656. [DOI: 10.1021/acs.jmedchem.8b00610] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Annachiara Gandini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, I-34136 Trieste, Italy
| | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Daniele Tedesco
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | | | - Carlos Roca
- Centro de Investigaciones Biologica, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Nuria E. Campillo
- Centro de Investigaciones Biologica, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Josefa Zaldivar-Diez
- Centro de Investigaciones Biologica, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Concepción Perez
- Instituto de Quimica Medica, CSIC, Calle Juan de la Cierva 3, 28006 Madrid, Spain
| | - Giampaolo Zuccheri
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
- S3 Center of the Institute of Nanosciences, Italian National Research Council (CNR), I-41125 Modena, Italy
| | - Andrea Miti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
- S3 Center of the Institute of Nanosciences, Italian National Research Council (CNR), I-41125 Modena, Italy
| | - Alessandra Feoli
- EpigeneticMedChemLab, Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, I-84084 Fisciano, Italy
| | - Sabrina Castellano
- EpigeneticMedChemLab, Department of Pharmacy, University of Salerno, Via Giovanni Paolo II 132, I-84084 Fisciano, Italy
| | - Sabrina Petralla
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Barbara Monti
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| | - Martina Rossi
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, I-34136 Trieste, Italy
| | - Fabio Moda
- Fondazione IRCCS Istituto Neurologico Carlo Besta, via Celoria 11, I-20133 Milan, Italy
| | - Giuseppe Legname
- Laboratory of Prion Biology, Department of Neuroscience, Scuola Internazionale Superiore di Studi Avanzati (SISSA), Via Bonomea 265, I-34136 Trieste, Italy
| | - Ana Martinez
- Centro de Investigaciones Biologica, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Maria Laura Bolognesi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum—University of Bologna, Via Belmeloro 6, I-40126 Bologna, Italy
| |
Collapse
|
15
|
Javidnia M, Hebron ML, Xin Y, Kinney NG, Moussa CEH. Pazopanib Reduces Phosphorylated Tau Levels and Alters Astrocytes in a Mouse Model of Tauopathy. J Alzheimers Dis 2018; 60:461-481. [PMID: 28869476 DOI: 10.3233/jad-170429] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hyperphosphorylation and aggregation of tau protein is a critical factor in many neurodegenerative diseases. These diseases are increasing in prevalence, and there are currently no cures. Previous work from our group and others has shown that tyrosine kinase inhibitors (TKIs) can stimulate autophagy, decrease pathological proteins, and improve symptoms in models of neurodegeneration. Here we examined the role of pazopanib in mouse models that express either human mutant P301L tau (TauP301L) or triple mutant amyloid precursor protein (3x-AβPP). The TauP301L mouse expresses P301L tau under the control of a prion promoter in both neurons and astrocytes, reminiscent of some human tauopathies. Pazopanib crosses the blood-brain barrier with no detectable peripheral off-side effects, and decreases p-tau in TauP301L mice. Pazopanib reaches a brain concentration sufficient for inhibition of several tyrosine kinases, including vascular endothelial growth factor receptors (VEGFRs). Further, pazopanib does not affect microglia but reduces astrocyte levels toward nontransgenic controls in TauP301L mice. Pazopanib does not alter amyloid beta levels or astrocytes in 3x-AβPP mice but modulates a number of inflammatory markers (IP-10, MIP-1α, MIP-1β, and RANTES). These data suggest that pazopanib may be involved in p-tau clearance and modulation of astrocytic activity in models of tauopathies.
Collapse
Affiliation(s)
- Monica Javidnia
- Department of Neurology, Laboratory for Dementiaand Parkinsonism, Translational Neurotherapeutics Program, Washington, DC, USA.,Department of Pharmacologyand Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Michaeline L Hebron
- Department of Neurology, Laboratory for Dementiaand Parkinsonism, Translational Neurotherapeutics Program, Washington, DC, USA
| | - Yue Xin
- Department of Neurology, Laboratory for Dementiaand Parkinsonism, Translational Neurotherapeutics Program, Washington, DC, USA
| | - Nikolas G Kinney
- Department of Neurology, Laboratory for Dementiaand Parkinsonism, Translational Neurotherapeutics Program, Washington, DC, USA
| | - Charbel E-H Moussa
- Department of Neurology, Laboratory for Dementiaand Parkinsonism, Translational Neurotherapeutics Program, Washington, DC, USA
| |
Collapse
|
16
|
Pulido R. PTEN Inhibition in Human Disease Therapy. Molecules 2018; 23:molecules23020285. [PMID: 29385737 PMCID: PMC6017825 DOI: 10.3390/molecules23020285] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 01/26/2018] [Accepted: 01/28/2018] [Indexed: 12/19/2022] Open
Abstract
The tumor suppressor PTEN is a major homeostatic regulator, by virtue of its lipid phosphatase activity against phosphatidylinositol 3,4,5-trisphosphate [PI(3,4,5)P3], which downregulates the PI3K/AKT/mTOR prosurvival signaling, as well as by its protein phosphatase activity towards specific protein targets. PTEN catalytic activity is crucial to control cell growth under physiologic and pathologic situations, and it impacts not only in preventing tumor cell survival and proliferation, but also in restraining several cellular regeneration processes, such as those associated with nerve injury recovery, cardiac ischemia, or wound healing. In these conditions, inhibition of PTEN catalysis is being explored as a potentially beneficial therapeutic intervention. Here, an overview of human diseases and conditions in which PTEN inhibition could be beneficial is presented, together with an update on the current status of specific small molecule inhibitors of PTEN enzymatic activity, their use in experimental models, and their limitations as research or therapeutic drugs.
Collapse
Affiliation(s)
- Rafael Pulido
- Biomarkers in Cancer Unit, Biocruces Health Research Institute, 48903 Barakaldo, Spain.
- IKERBASQUE, Basque Foundation for Science, 48013 Bilbao, Spain.
| |
Collapse
|
17
|
李 月. Research Progress on the Secondary Metabolites from Dinoflagellate Prorocentrum Spp. INTERNATIONAL JOURNAL OF ECOLOGY 2018. [DOI: 10.12677/ije.2018.74023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
18
|
Liu XY, Zhang LJ, Chen Z, Liu LB. The PTEN inhibitor bpV(pic) promotes neuroprotection against amyloid β-peptide (25-35)-induced oxidative stress and neurotoxicity. Neurol Res 2017; 39:758-765. [PMID: 28436304 DOI: 10.1080/01616412.2017.1317916] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Xiao-Ying Liu
- Department of Endocrinology, Fujian Institute of Endocrinology, Union Hospital of Fujian Medical University, Fuzhou, China
| | - Li-Jing Zhang
- Zhejiang Department of Pharmacology, Pharmaceutical college, Ningbo, China
| | - Zhou Chen
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Li-Bin Liu
- Department of Endocrinology, Fujian Institute of Endocrinology, Union Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
19
|
O'Rourke C, Lee-Reeves C, Drake RA, Cameron GW, Loughlin AJ, Phillips JB. Adapting tissue-engineered in vitro CNS models for high-throughput study of neurodegeneration. J Tissue Eng 2017; 8:2041731417697920. [PMID: 28507726 PMCID: PMC5415290 DOI: 10.1177/2041731417697920] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 02/14/2017] [Indexed: 11/21/2022] Open
Abstract
Neurodegenerative conditions remain difficult to treat, with the continuing failure to see therapeutic research successfully advance to clinical trials. One of the obstacles that must be overcome is to develop enhanced models of disease. Tissue engineering techniques enable us to create organised artificial central nervous system tissue that has the potential to improve the drug development process. This study presents a replicable model of neurodegenerative pathology through the use of engineered neural tissue co-cultures that can incorporate cells from various sources and allow degeneration and protection of neurons to be observed easily and measured, following exposure to neurotoxic compounds – okadaic acid and 1-methyl-4-phenylpyridinium. Furthermore, the technology has been miniaturised through development of a mould with 6 mm length that recreates the advantageous features of engineered neural tissue co-cultures at a scale suitable for commercial research and development. Integration of human-derived induced pluripotent stem cells aids more accurate modelling of human diseases, creating new possibilities for engineered neural tissue co-cultures and their use in drug screening.
Collapse
Affiliation(s)
- Caitriona O'Rourke
- Department of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK.,Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - Charlotte Lee-Reeves
- Department of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
| | | | | | - A Jane Loughlin
- Department of Life, Health and Chemical Sciences, The Open University, Milton Keynes, UK
| | - James B Phillips
- Department of Biomaterials and Tissue Engineering, UCL Eastman Dental Institute, University College London, London, UK
| |
Collapse
|
20
|
Tan L, Chen X, Wang W, Zhang J, Li S, Zhao Y, Wang J, Luo A. Pharmacological inhibition of PTEN attenuates cognitive deficits caused by neonatal repeated exposures to isoflurane via inhibition of NR2B-mediated tau phosphorylation in rats. Neuropharmacology 2017; 114:135-145. [PMID: 27836791 DOI: 10.1016/j.neuropharm.2016.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 10/30/2016] [Accepted: 11/07/2016] [Indexed: 10/20/2022]
Abstract
Evidence has shown that children exposed to repeated anesthesia in early childhood display long-term cognitive disabilities. However, the underlying mechanisms remain largely unclear. Our previous study has indicated the involvement of phosphatase and tensin homolog deleted on chromosome 10 (PTEN) in isoflurane-induced decrease of self-renewal capacity in hippocampal neural precursor cells. Additionally, it is demonstrated by others that PTEN inhibition could protect against cognitive impairment via reduction of tau phosphorylation in the alzheimer's disease model. Therefore, in the present in vivo study, we aimed to examine the effects of PTEN inhibition on the cognitive dysfunction and tau hyperphosphorylation caused by neonatal repeated exposures to isoflurane. Our results showed that the neonatal repeated exposures to isoflurane resulted in the activation of PTEN in the hippocampus. The treatment of PTEN inhibitor BPV (pic) restored PSD-95 synthesis, and attenuated tau phosphorylation as well as the cognitive dysfunction caused by the repeated isoflurane exposures. In addition, BPV (pic) treatment reversed the activation of NR2B-containing NMDARs induced by repeated isoflurane exposures, while in turn, the antagonism of NR2B subunit with ifenprodil alleviated tau phosphorylation, indicating a possible role of NR2B as the downstream of PTEN in mediating tau phosphorylation in the neonatal rats repeatedly exposed to isoflurane. In conclusion, our results reveal a novel role of PTEN in mediating tau phosphorylation and cognitive deficits caused by neonatal repeated exposures to isoflurane, implying that targeting on PTEN may be a potential therapeutic approach for the anesthetic-related cognitive decline in the developing brain.
Collapse
Affiliation(s)
- Lei Tan
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Chen
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, Xi'an 710068, China
| | - Wei Wang
- Department of Anesthesiology, First Hospital of Xian Jiaotong University, Xi'an 710061, China
| | - Jianfang Zhang
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shiyong Li
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yilin Zhao
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jintao Wang
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ailin Luo
- Department of Anesthesiology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
21
|
Wu Q, Ye X, Xiong Y, Zhu H, Miao J, Zhang W, Wan J. The Protective Role of microRNA-200c in Alzheimer's Disease Pathologies Is Induced by Beta Amyloid-Triggered Endoplasmic Reticulum Stress. Front Mol Neurosci 2016; 9:140. [PMID: 28008308 PMCID: PMC5143617 DOI: 10.3389/fnmol.2016.00140] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 11/24/2016] [Indexed: 12/26/2022] Open
Abstract
MicroRNAs are small non-coding RNAs that repress the expression of their target proteins. The roles of microRNAs in the development of Alzheimer's disease (AD) are not clear. In this study we show that miR-200c represses the expression of PTEN protein. PTEN downregulation by miR-200c supports the survival and differentiation of cultured neurons. AD is a progressive neurodegenerative disease signified by beta amyloid (Aβ) peptide aggregation and deposition. In a mouse model of AD that is induced by APPswe and PS1ΔE9 double transgenes, we found Aβ deposition results in neuronal ER stress that induces miR200c. Pharmacological blockade of ER stress inhibited Aβ-induced miR-200c overexpression in AD brains. MiR-200c was detected in the serum of both AD mice and human AD patients. These findings suggest that miR-200c functions as part of the neuronal cell-intrinsic adaptive machinery, and supports neuronal survival and differentiation in response to Aβ induced ER-stress by downregulating PTEN.
Collapse
Affiliation(s)
- Qi Wu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center Shenzhen, China
| | - Xiaoyang Ye
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center Shenzhen, China
| | - Yi Xiong
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center Shenzhen, China
| | - Haili Zhu
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center Shenzhen, China
| | - Jianting Miao
- Department of Neurology, Tangdu Hospital, Fourth Military Medical University Xi'an City, China
| | - Wei Zhang
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center Shenzhen, China
| | - Jun Wan
- Shenzhen Key Laboratory for Neuronal Structural Biology, Biomedical Research Institute, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical CenterShenzhen, China; Division of Life Science, The Hong Kong University of Science and TechnologyHong Kong, China
| |
Collapse
|
22
|
Josephy-Hernandez S, Jmaeff S, Pirvulescu I, Aboulkassim T, Saragovi HU. Neurotrophin receptor agonists and antagonists as therapeutic agents: An evolving paradigm. Neurobiol Dis 2016; 97:139-155. [PMID: 27546056 DOI: 10.1016/j.nbd.2016.08.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 08/10/2016] [Accepted: 08/16/2016] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative disorders are prevalent, complex and devastating conditions, with very limited treatment options currently available. While they manifest in many forms, there are commonalities that link them together. In this review, we will focus on neurotrophins - a family of related factors involved in neuronal development and maintenance. Neurodegenerative diseases often present with a neurotrophin imbalance, in which there may be decreases in trophic signaling through Trk receptors for example, and/or increases in pro-apoptotic activity through p75. Clinical trials with neurotrophins have continuously failed due to their poor pharmacological properties as well as the unavoidable activation of p75. Thus, there is a need for drugs without such setbacks. Small molecule neurotrophin mimetics are favorable options since they can selectively activate Trks or inactivate p75. In this review, we will initially present a brief outline of how these molecules are synthesized and their mechanisms of action; followed by an update in the current state of neurotrophins and small molecules in major neurodegenerative diseases. Although there has been significant progress in the development of potential therapeutics, more studies are needed to establish clear mechanisms of action and target specificity in order to transition from animal models to the assessment of safety and use in humans.
Collapse
Affiliation(s)
- Sylvia Josephy-Hernandez
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Sean Jmaeff
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Iulia Pirvulescu
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Tahar Aboulkassim
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - H Uri Saragovi
- Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada; Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
23
|
Zhao J, Chen Y, Xu Y, Pi G. Effects of PTEN inhibition on the regulation of Tau phosphorylation in rat cortical neuronal injury after oxygen and glucose deprivation. Brain Inj 2016; 30:1150-9. [PMID: 27245882 DOI: 10.3109/02699052.2016.1161828] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
OBJECTIVE This report investigated the involvement of the PTEN pathway in the regulation of Tau phosphorylation using an oxygen and glucose deprivation (OGD) model with rat cortical neurons. METHODS Primary cortical neurons were used to establish the oxygen and glucose deprivation (OGD) model in vitro. These were randomly divided into control, OGD, bpV+OGD, As+OGD, Se+OGD and Mock treatment groups. The neuron viability was assessed by MTT, the cell apoptosis was detected using TUNEL staining. The expression of Phospho-PTEN/PTEN, Phospho-Tau/Tau, Phospho-Akt/Akt and Phospho-GSK-3β/GSK-3β were detected by Western blotting. RESULTS OGD induced Tau phosphorylation through PTEN and glycogen synthase kinase-3β (GSK-3β) activation, together with a decrease in AKT activity. Pre-treatment with bpv, a potent PTEN inhibitor, and PTEN antisense nucleotides decreased PTEN and GSK-3β activity and caused alterations in Tau phosphorylation. Neuronal apoptosis was also reduced. CONCLUSIONS The PTEN/Akt/GSK-3β/Tau pathway is involved in the regulation of neuronal injury, providing a novel route for protecting neurons following neonatal HI.
Collapse
Affiliation(s)
- Jing Zhao
- a Department of Neonatology , Affiliated Hospital of North Sichuan Medical College , Nanchong , PR China.,b Department of Pediatrics , North Sichuan Medical College , Nanchong , PR China
| | - Yurong Chen
- a Department of Neonatology , Affiliated Hospital of North Sichuan Medical College , Nanchong , PR China.,b Department of Pediatrics , North Sichuan Medical College , Nanchong , PR China
| | - Yuxia Xu
- b Department of Pediatrics , North Sichuan Medical College , Nanchong , PR China
| | - Guanghuan Pi
- b Department of Pediatrics , North Sichuan Medical College , Nanchong , PR China
| |
Collapse
|
24
|
Qi L, Ke L, Liu X, Liao L, Ke S, Liu X, Wang Y, Lin X, Zhou Y, Wu L, Chen Z, Liu L. Subcutaneous administration of liraglutide ameliorates learning and memory impairment by modulating tau hyperphosphorylation via the glycogen synthase kinase-3β pathway in an amyloid β protein induced alzheimer disease mouse model. Eur J Pharmacol 2016; 783:23-32. [PMID: 27131827 DOI: 10.1016/j.ejphar.2016.04.052] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/18/2016] [Accepted: 04/26/2016] [Indexed: 12/21/2022]
Abstract
Type 2 diabetes mellitus is a risk factor for Alzheimer's disease (AD). The glucagon-like peptide-1 analog liraglutide, a novel long-lasting incretin hormone, has been used to treat type 2 diabetes mellitus. In addition, liraglutide has been shown to be neurotrophic and neuroprotective. Here, we investigated the effects of liraglutide on amyloid β protein (Aβ)-induced AD in mice and explored its mechanism of action. The results showed that subcutaneous administration of liraglutide (25nmol/day), once daily for 8 weeks, prevented memory impairments in the Y Maze and Morris Water Maze following Aβ1-42 intracerebroventricular injection, and alleviated the ultra-structural changes of pyramidal neurons and chemical synapses in the hippocampal CA1 region. Furthermore, liraglutide reduced Aβ1-42-induced tau phosphorylation via the protein kinase B and glycogen synthase kinase-3β pathways. Thus liraglutide may alleviate cognitive impairment in AD by at least decreasing the phosphorylation of tau.
Collapse
Affiliation(s)
- Liqin Qi
- Department of Endocrinology, Union Hospital, Fujian Institute of Endocrinology, The Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Linfang Ke
- Department of Endocrinology, Union Hospital, Fujian Institute of Endocrinology, The Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Xiaohong Liu
- Department of Endocrinology, Union Hospital, Fujian Institute of Endocrinology, The Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Lianming Liao
- Department of Endocrinology, Union Hospital, Fujian Institute of Endocrinology, The Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Sujie Ke
- Department of Endocrinology, Union Hospital, Fujian Institute of Endocrinology, The Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Xiaoying Liu
- Department of Endocrinology, Union Hospital, Fujian Institute of Endocrinology, The Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Yanping Wang
- Department of Endocrinology, Union Hospital, Fujian Institute of Endocrinology, The Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Xiaowei Lin
- Department of Endocrinology, Union Hospital, Fujian Institute of Endocrinology, The Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Yu Zhou
- Department of Endocrinology, Union Hospital, Fujian Institute of Endocrinology, The Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Lijuan Wu
- Department of Endocrinology, Union Hospital, Fujian Institute of Endocrinology, The Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Zhou Chen
- Department of Pharmacology, College of Pharmacy, Fujian Medical University, Fuzhou, Fujian, People's Republic of China.
| | - Libin Liu
- Department of Endocrinology, Union Hospital, Fujian Institute of Endocrinology, The Union Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China.
| |
Collapse
|
25
|
Pulido R. PTEN: a yin-yang master regulator protein in health and disease. Methods 2016; 77-78:3-10. [PMID: 25843297 DOI: 10.1016/j.ymeth.2015.02.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Accepted: 02/19/2015] [Indexed: 01/16/2023] Open
Abstract
The PTEN gene is a tumor suppressor gene frequently mutated in human tumors, which encodes a ubiquitous protein whose major activity is to act as a lipid phosphatase that counteracts the action of the oncogenic PI3K. In addition, PTEN displays protein phosphatase- and catalytically-independent activities. The physiologic control of PTEN function, and its inactivation in cancer and other human diseases, including some neurodevelopmental disorders, is upon the action of multiple regulatory mechanisms. This provides a wide spectrum of potential therapeutic approaches to reconstitute PTEN activity. By contrast, inhibition of PTEN function may be beneficial in a different group of human diseases, such as type 2 diabetes or neuroregeneration-related pathologies. This makes PTEN a functionally dual yin-yang protein with high potential in the clinics. Here, a brief overview on PTEN and its relation with human disease is presented.
Collapse
Affiliation(s)
- Rafael Pulido
- BioCruces Health Research Institute, Barakaldo, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
26
|
Targeting PTEN using small molecule inhibitors. Methods 2015; 77-78:63-8. [DOI: 10.1016/j.ymeth.2015.02.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 02/15/2015] [Accepted: 02/16/2015] [Indexed: 12/22/2022] Open
|
27
|
The role of TLR4-mediated PTEN/PI3K/AKT/NF-κB signaling pathway in neuroinflammation in hippocampal neurons. Neuroscience 2014; 269:93-101. [DOI: 10.1016/j.neuroscience.2014.03.039] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 03/19/2014] [Accepted: 03/19/2014] [Indexed: 11/18/2022]
|
28
|
Valdiglesias V, Prego-Faraldo MV, Pásaro E, Méndez J, Laffon B. Okadaic acid: more than a diarrheic toxin. Mar Drugs 2013; 11:4328-49. [PMID: 24184795 PMCID: PMC3853731 DOI: 10.3390/md11114328] [Citation(s) in RCA: 167] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Revised: 10/08/2013] [Accepted: 10/23/2013] [Indexed: 01/04/2023] Open
Abstract
Okadaic acid (OA) is one of the most frequent and worldwide distributed marine toxins. It is easily accumulated by shellfish, mainly bivalve mollusks and fish, and, subsequently, can be consumed by humans causing alimentary intoxications. OA is the main representative diarrheic shellfish poisoning (DSP) toxin and its ingestion induces gastrointestinal symptoms, although it is not considered lethal. At the molecular level, OA is a specific inhibitor of several types of serine/threonine protein phosphatases and a tumor promoter in animal carcinogenesis experiments. In the last few decades, the potential toxic effects of OA, beyond its role as a DSP toxin, have been investigated in a number of studies. Alterations in DNA and cellular components, as well as effects on immune and nervous system, and even on embryonic development, have been increasingly reported. In this manuscript, results from all these studies are compiled and reviewed to clarify the role of this toxin not only as a DSP inductor but also as cause of alterations at the cellular and molecular levels, and to highlight the relevance of biomonitoring its effects on human health. Despite further investigations are required to elucidate OA mechanisms of action, toxicokinetics, and harmful effects, there are enough evidences illustrating its toxicity, not related to DSP induction, and, consequently, supporting a revision of the current regulation on OA levels in food.
Collapse
Affiliation(s)
- Vanessa Valdiglesias
- Toxicology Unit, Department of Psychobiology, University of A Coruña, A Coruña E15071, Spain; E-Mails: (E.P.); (B.L.)
- Department of Cellular and Molecular Biology, University of A Coruna, A Coruña E15071, Spain; E-Mails: (M.V.P.-F.); (J.M.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +34-981167000; Fax: +34-981167172
| | - María Verónica Prego-Faraldo
- Department of Cellular and Molecular Biology, University of A Coruna, A Coruña E15071, Spain; E-Mails: (M.V.P.-F.); (J.M.)
| | - Eduardo Pásaro
- Toxicology Unit, Department of Psychobiology, University of A Coruña, A Coruña E15071, Spain; E-Mails: (E.P.); (B.L.)
| | - Josefina Méndez
- Department of Cellular and Molecular Biology, University of A Coruna, A Coruña E15071, Spain; E-Mails: (M.V.P.-F.); (J.M.)
| | - Blanca Laffon
- Toxicology Unit, Department of Psychobiology, University of A Coruña, A Coruña E15071, Spain; E-Mails: (E.P.); (B.L.)
| |
Collapse
|
29
|
Patil SP, Tran N, Geekiyanage H, Liu L, Chan C. Curcumin-induced upregulation of the anti-tau cochaperone BAG2 in primary rat cortical neurons. Neurosci Lett 2013; 554:121-5. [PMID: 24035895 PMCID: PMC3825752 DOI: 10.1016/j.neulet.2013.09.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 06/20/2013] [Accepted: 09/04/2013] [Indexed: 01/12/2023]
Abstract
Alzheimer's disease (AD) is a progressive, neurodegenerative disease characterized by extracellular deposits of amyloid beta (Aβ) protein and intracellular neurofibrillary tangles of hyperphosphorylated tau protein. Various studies suggest that the tau tangle pathology, which lies downstream to Aβ pathology, is essential to produce AD-associated clinical phenotype and thus treatments targeting tau pathology may prevent or delay disease progression effectively. In this context, our present study examined three polyphenol compounds (curcumin, EGCG and resveratrol) for their possible activity against two endogenous proteins (BAG2 and LAMP1) that are shown to play a vital role in clearing tau tangles from neurons. Human epidemiological and animal data suggest potential positive effects of these polyphenols against AD. Here, primary rat cortical neurons treated with these polyphenols significantly up-regulated BAG2 levels at different concentrations, while only EGCG upregulated LAMP1 levels, although at higher concentrations. Importantly, curcumin doubled BAG2 levels at low micromolar concentrations that are clinically relevant. In addition, curcumin also downregulated levels of phosphorylated tau, which may be potentially attributed to the curcumin-induced upregulation in BAG2 levels in the neurons. The present results demonstrate novel activity of polyphenol curcumin in up-regulating an anti-tau cochaperone BAG2 and thus, suggest probable benefit of curcumin against AD-associated tauopathy.
Collapse
Affiliation(s)
- Sachin P. Patil
- Cellular and Biomolecular Laboratory, Department of Chemical Engineering and Material Science, Michigan State University, East Lansing, MI 48824
| | - Nhung Tran
- Cellular and Biomolecular Laboratory, Department of Chemical Engineering and Material Science, Michigan State University, East Lansing, MI 48824
| | - Hirosha Geekiyanage
- Cellular and Biomolecular Laboratory, Department of Chemical Engineering and Material Science, Michigan State University, East Lansing, MI 48824
| | - Li Liu
- Cellular and Biomolecular Laboratory, Department of Chemical Engineering and Material Science, Michigan State University, East Lansing, MI 48824
| | - Christina Chan
- Cellular and Biomolecular Laboratory, Department of Chemical Engineering and Material Science, Michigan State University, East Lansing, MI 48824
| |
Collapse
|
30
|
Medina M, Avila J, Villanueva N. Use of okadaic acid to identify relevant phosphoepitopes in pathology: a focus on neurodegeneration. Mar Drugs 2013; 11:1656-68. [PMID: 23697949 PMCID: PMC3707166 DOI: 10.3390/md11051656] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 04/02/2013] [Accepted: 04/16/2013] [Indexed: 12/24/2022] Open
Abstract
Protein phosphorylation is involved in the regulation of a wide variety of physiological processes and is the result of a balance between protein kinase and phosphatase activities. Biologically active marine derived compounds have been shown to represent an interesting source of novel compounds that could modify that balance. Among them, the marine toxin and tumor promoter, okadaic acid (OA), has been shown as an inhibitor of two of the main cytosolic, broad-specificity protein phosphatases, PP1 and PP2A, thus providing an excellent cell-permeable probe for examining the role of protein phosphorylation, and PP1 and PP2A in particular, in any physiological or pathological process. In the present work, we review the use of okadaic acid to identify specific phosphoepitopes mainly in proteins relevant for neurodegeneration. We will specifically highlight those cases of highly dynamic phosphorylation-dephosphorylation events and the ability of OA to block the high turnover phosphorylation, thus allowing the detection of modified residues that could be otherwise difficult to identify. Finally, its effect on tau hyperhosphorylation and its relevance in neurodegenerative pathologies such as Alzheimer’s disease and related dementia will be discussed.
Collapse
Affiliation(s)
- Miguel Medina
- CIBERNED (Center for Networked Biomedical Research in Neurodegenerative Diseases), Valderrebollo 5, Madrid 28041, Spain; E-Mail:
| | - Jesús Avila
- CIBERNED (Center for Networked Biomedical Research in Neurodegenerative Diseases), Valderrebollo 5, Madrid 28041, Spain; E-Mail:
- Center of Molecular Biology “Severo Ochoa” CSIC-UAM, Nicolás Cabrera 1, Madrid 28049, Spain
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +34-91-196-4803; Fax: +34-91-196-4715
| | - Nieves Villanueva
- National Center of Microbiology (CNM), Carlos III Institute of Health (ISCIII), Crta. Majadahonda-Pozuelo km 2, Majadahonda, Madrid 28220, Spain; E-Mail:
| |
Collapse
|