1
|
Jones T, Raman R, Puhl AC, Lane TR, Riabova O, Kazakova E, Makarov V, Ekins S. Discovery of Dual Targeting GSK-3β/HIV-1 Reverse Transcriptase Inhibitors as Neuroprotective Antiviral Agents. ACS Chem Neurosci 2024. [PMID: 39663760 DOI: 10.1021/acschemneuro.4c00725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024] Open
Abstract
Glycogen synthase kinase-3 beta (GSK-3β or GSK-3B) is a serine-threonine kinase involved in various pathways and cellular processes. Alteration in GSK-3β activity is associated with several neurological diseases including Alzheimer's disease (AD), bipolar disorder, and rare diseases like Rett syndrome. GSK-3β is also implicated in HIV-associated dementia (HAD), as it is upregulated in HIV-1-infected cells and plays a role in neuronal dysfunction. Therefore, a small molecule that can inhibit both GSK-3β and HIV-1 reverse transcriptase could offer neuroprotective therapy for patients suffering from HIV-1. Despite this, there are no known GSK-3β inhibitors currently approved, thus prompting us to screen our panel of various antiviral compounds against this kinase to better understand its structure-activity relationship. We show for the first time that the approved drugs, etravirine and rilpivirine, possess GSK-3β activity (IC50 619 nM and 502 nM, respectively). We have also identified 3 lead molecules exhibiting IC50 < 1 μM (11726169, 12326205, and 12326207), with compound 11726169 being the most potent in vitro GSK-3β inhibitor (IC50 = 12.1 nM). We also describe the generation of machine learning models for GSK-3β inhibition and their validation with our data as an external test set and propose their use for the future optimization of such inhibitors.
Collapse
Affiliation(s)
- Thane Jones
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Renuka Raman
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Ana C Puhl
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Thomas R Lane
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| | - Olga Riabova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, Moscow 119071, Russia
| | - Elena Kazakova
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, Moscow 119071, Russia
| | - Vadim Makarov
- Research Center of Biotechnology RAS, Leninsky Prospekt 33-2, Moscow 119071, Russia
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, North Carolina 27606, United States
| |
Collapse
|
2
|
Di Lorenzo D. Tau Protein and Tauopathies: Exploring Tau Protein-Protein and Microtubule Interactions, Cross-Interactions and Therapeutic Strategies. ChemMedChem 2024; 19:e202400180. [PMID: 39031682 DOI: 10.1002/cmdc.202400180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/22/2024]
Abstract
Tau, a microtubule-associated protein (MAP), is essential to maintaining neuronal stability and function in the healthy brain. However, aberrant modifications and pathological aggregations of Tau are implicated in various neurodegenerative disorders, collectively known as tauopathies. The most common Tauopathy is Alzheimer's Disease (AD) counting nowadays more than 60 million patients worldwide. This comprehensive review delves into the multifaceted realm of Tau protein, puzzling out its intricate involvement in both physiological and pathological roles. Emphasis is put on Tau Protein-Protein Interactions (PPIs), depicting its interaction with tubulin, microtubules and its cross-interaction with other proteins such as Aβ1-42, α-synuclein, and the chaperone machinery. In the realm of therapeutic strategies, an overview of diverse possibilities is presented with their relative clinical progresses. The focus is mostly addressed to Tau protein aggregation inhibitors including recent small molecules, short peptides and peptidomimetics with specific focus on compounds that showed a double anti aggregative activity on both Tau protein and Aβ amyloid peptide. This review amalgamates current knowledge on Tau protein and evolving therapeutic strategies, providing a comprehensive resource for researchers seeking to deepen their understanding of the Tau protein and for scientists involved in the development of new peptide-based anti-aggregative Tau compounds.
Collapse
Affiliation(s)
- Davide Di Lorenzo
- Department of Chemistry, Organic and Bioorganic Chemistry, Bielefeld University, Universitätsstraße 25, D-33615, Bielefeld, Germany
| |
Collapse
|
3
|
Mokhtar HI, Zaitone SA, El-Sayed K, Lashine RM, Ahmed N, Moursi SMM, Shehata SA, Aldahish AA, Helal MA, El-Kherbetawy MK, Fawzy MS, Abd El-Fadeal NM. Molecular Docking, Bioinformatic Analysis, and Experimental Verification for the Effect of Naringin on ADHD: Possible Inhibition of GSK-3β and HSP90. Pharmaceuticals (Basel) 2024; 17:1436. [PMID: 39598348 PMCID: PMC11597433 DOI: 10.3390/ph17111436] [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: 09/22/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: One of the most abundant and growing neurodevelopmental disorders in recent decades is attention deficit hyperactivity disorder (ADHD). Many trials have been performed on using drugs for the improvement of ADHD signs. This study aimed to detect the possible interaction of naringin with Wnt/β-catenin signaling and its putative anti-inflammatory and protective effects in the mouse ADHD model based on bioinformatic, behavioral, and molecular investigations. Furthermore, molecular docking was applied to investigate possible interactions with the GSK-3β and HSP90 proteins. Methods: Male Swiss albino mice were divided into four groups, a normal control group, monosodium glutamate (SGL) control, SGL + naringin 50 mg/kg, and SGL + naringin 100 mg/kg. The psychomotor activity of the mice was assessed using the self-grooming test, rope crawling test, and attentional set-shifting task (ASST). In addition, biochemical analyses were performed using brain samples. Results: The results of the SGL group showed prolonged grooming time (2.47-folds), a lower percentage of mice with successful crawling on the rope (only 16.6%), and a higher number of trials for compound discrimination testing in the ASST (12.83 ± 2.04 trials versus 5.5 ± 1.88 trials in the normal group). Treatment with naringin (50 or 100 mg per kg) produced significant shortening in the grooming time (31% and 27% reductions), as well as a higher percentage of mice succeeding in crawling with the rope (50% and 83%, respectively). Moreover, the ELISA assays indicated decreased dopamine levels (0.36-fold) and increased TNF-α (2.85-fold) in the SGL control group compared to the normal mice, but an improvement in dopamine level was observed in the naringin (50 or 100 mg per kg)-treated groups (1.58-fold and 1.97-fold). Similarly, the PCR test showed significant declines in the expression of the Wnt (0.36), and β-catenin (0.33) genes, but increased caspase-3 (3.54-fold) and BAX (5.36-fold) genes in the SGL group; all these parameters were improved in the naringin 50 or 100 mg/kg groups. Furthermore, molecular docking indicated possible inhibition for HSP90 and GSK-3β. Conclusions: Overall, we can conclude that naringin is a promising agent for alleviating ADHD symptoms, and further investigations are required to elucidate its mechanism of action.
Collapse
Affiliation(s)
- Hatem I. Mokhtar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Sinai University-Kantara Branch, Ismailia 41636, Egypt;
| | - Sawsan A. Zaitone
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, University of Tabuk, Tabuk 47713, Saudi Arabia
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Karima El-Sayed
- Medical Physiology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Rehab M. Lashine
- Clinical Pharmacology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Nada Ahmed
- Department of Clinical Pathology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Suzan M. M. Moursi
- Medical Physiology Department, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Shaimaa A. Shehata
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Afaf A. Aldahish
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 61441, Saudi Arabia;
| | - Mohamed A. Helal
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, October Gardens, 6th of October, Giza 12587, Egypt;
- Medicinal Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | | | - Manal S. Fawzy
- Department of Biochemistry, Faculty of Medicine, Northern Border University, Arar 91431, Saudi Arabia;
| | - Noha M. Abd El-Fadeal
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt
- Biochemistry Department, Ibn Sina National College for Medical Studies, Jeddah 22421, Saudi Arabia
| |
Collapse
|
4
|
Xiao Z, Li Y, Haider A, Pfister SK, Rong J, Chen J, Zhao C, Zhou X, Song Z, Gao Y, Patel JS, Collier TL, Ran C, Zhai C, Yuan H, Liang SH. Radiosynthesis and evaluation of a novel 18F-labeled tracer for PET imaging of glycogen synthase kinase 3. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2024; 14:327-336. [PMID: 39583910 PMCID: PMC11578811 DOI: 10.62347/obzs8887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 09/10/2024] [Indexed: 11/26/2024]
Abstract
Glycogen synthase kinase 3 (GSK3) is a multifunctional serine/threonine kinase family that regulates diverse biological processes including glucose metabolism, insulin activity and energy homeostasis. Dysregulation of GSK3 is implicated in the development of several diseases such as type 2 diabetes mellitus, Alzheimer's disease (AD), and various cancer types. In this study, we report the synthesis and evaluation of a novel positron emission tomography (PET) ligand compound 28 (codenamed [18F]GSK3-2209). The PET ligand [18F]28 was obtained via copper-mediated radiofluorination in more than 32% radiochemical yields, with high radiochemical purity and high molar activity. In vitro autoradiography studies in rodents demonstrated that this tracer exhibited a high specific binding to GSK3. Furthermore, PET imaging studies of [18F]28 revealed its ability to penetrate the blood-brain barrier (BBB).
Collapse
Affiliation(s)
- Zhiwei Xiao
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Yinlong Li
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Ahmed Haider
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Stefanie K Pfister
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Jian Rong
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Jiahui Chen
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Chunyu Zhao
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Xin Zhou
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
| | - Zhendong Song
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
| | - Yabiao Gao
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
| | - Jimmy S Patel
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Department of Radiation Oncology, Winship Cancer Institute of Emory UniversityAtlanta, GA 30322, USA
| | - Thomas L Collier
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical SchoolBoston, MA 02114, USA
| | - Chuangyan Zhai
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
| | - Hongjie Yuan
- Department of Pharmacology and Chemical Biology, Emory University School of MedicineAtlanta, GA 30322, USA
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory UniversityAtlanta, GA 30322, USA
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Department of Radiology, Harvard Medical SchoolBoston, MA 02114, USA
| |
Collapse
|
5
|
Varshini MS, Reddy RA, Krishnamurthy PT, Wadhwani A. Harmony of Wnt pathway in Alzheimer's: Navigating the multidimensional progression from preclinical to clinical stages. Neurosci Biobehav Rev 2024; 165:105863. [PMID: 39179059 DOI: 10.1016/j.neubiorev.2024.105863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/14/2024] [Accepted: 08/18/2024] [Indexed: 08/26/2024]
Abstract
The Wnt pathway stands out as a pivotal signal transduction pathway, operating through two distinct modes of signaling: the canonical/β-catenin pathway and the non-canonical pathway. Among these, the canonical pathway assumes a paramount role in various physiological and pathological processes within the human body. Particularly in the brain, Wnt exhibits involvement in fundamental physiological events including neuronal differentiation/survival, axonogenesis, neural stem cell regulation, synaptic plasticity, and cell cycle modulation. Notably, scientific evidence underscores the critical role of the Wnt pathway in the pathogenesis of Alzheimer's disease (AD), correlating with its involvement in key pathological features such as tau tangles, Amyloid-β plaques, synaptic dysfunction, oxidative stress, mitochondrial dysfunction, cognitive impairments, and disruption of the blood-brain barrier integrity. This review aims to comprehensively explore the involvement and significance of Wnt signaling in Alzheimer's. Furthermore, it delves into recent advancements in research on Wnt signaling, spanning from preclinical investigations to clinical trials.
Collapse
Affiliation(s)
- Magham Sai Varshini
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, TN 643001, India
| | - Ramakkamma Aishwarya Reddy
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, TN 643001, India
| | | | - Ashish Wadhwani
- Department of Pharmaceutical Biotechnology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, TN 643001, India; Faculty of Health Sciences, School of Pharmacy, JSS Academy of Higher Education and Research, Mauritius, Vacoas 73304, Mauritius
| |
Collapse
|
6
|
Jezernik G, Glavač D, Skok P, Krušič M, Potočnik U, Gorenjak M. Discovery of Novel Biomarkers with Extended Non-Coding RNA Interactor Networks from Genetic and Protein Biomarkers. Int J Mol Sci 2024; 25:10210. [PMID: 39337694 PMCID: PMC11432684 DOI: 10.3390/ijms251810210] [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: 08/07/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Curated online interaction databases and gene ontology tools have streamlined the analysis of highly complex gene/protein networks. However, understanding of disease pathogenesis has gradually shifted from a protein-based core to complex interactive networks where non-coding RNA (ncRNA) is thought to play an essential role. As current gene ontology is based predominantly on protein-level information, there is a growing need to analyze networks with ncRNA. In this study, we propose a gene ontology workflow integrating ncRNA using the NPInter V5.0 database. To validate the proposed workflow, we analyzed our previously published curated biomarker datasets for hidden disease susceptibility processes and pharmacogenomics. Our results show a novel involvement of melanogenesis in psoriasis response to biological drugs in general. Hyperpigmentation has been previously observed in psoriasis following treatment with currently indicated biological drugs, thus calling attention to melanogenesis research as a response biomarker in psoriasis. Moreover, our proposed workflow highlights the need to critically evaluate computed ncRNA interactions within databases and a demand for gene ontology analysis of large miRNA blocks.
Collapse
Affiliation(s)
- Gregor Jezernik
- Center for Human Genetics & Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (D.G.); (M.K.); (U.P.); (M.G.)
- National-Level Institute for Sustainable Environmental Solutions, Jadranska cesta 28, 2000 Maribor, Slovenia
| | - Damjan Glavač
- Center for Human Genetics & Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (D.G.); (M.K.); (U.P.); (M.G.)
- Department of Molecular Genetics, Institute of Pathology, Faculty of Medicine, University of Ljubljana, Korytkova 2, 1000 Ljubljana, Slovenia
| | - Pavel Skok
- Department of Gastroenterology, Internal Medicine Clinic, University Medical Centre Maribor, Ljubljanska ulica 8, 2000 Maribor, Slovenia;
- Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia
| | - Martina Krušič
- Center for Human Genetics & Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (D.G.); (M.K.); (U.P.); (M.G.)
| | - Uroš Potočnik
- Center for Human Genetics & Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (D.G.); (M.K.); (U.P.); (M.G.)
- Department for Science and Research, University Medical Centre Maribor, Ljubljanska ulica 8, 2000 Maribor, Slovenia
- Faculty of Chemistry and Chemical Engineering, University of Maribor, Smetanova ulica 17, 2000 Maribor, Slovenia
| | - Mario Gorenjak
- Center for Human Genetics & Pharmacogenomics, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000 Maribor, Slovenia; (D.G.); (M.K.); (U.P.); (M.G.)
| |
Collapse
|
7
|
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
|
8
|
Sai Varshini M, Aishwarya Reddy R, Thaggikuppe Krishnamurthy P. Unlocking hope: GSK-3 inhibitors and Wnt pathway activation in Alzheimer's therapy. J Drug Target 2024; 32:909-917. [PMID: 38838023 DOI: 10.1080/1061186x.2024.2365263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterised by progressive cognitive decline and the accumulation of amyloid-β plaques and tau tangles. The Wnt signalling pathway known for its crucial role in neurodevelopment and adult neurogenesis has emerged as a potential target for therapeutic intervention in AD. Glycogen synthase kinase-3 beta (GSK-3β), a key regulator of the Wnt pathway, plays a pivotal role in AD pathogenesis by promoting tau hyperphosphorylation and neuroinflammation. Several preclinical studies have demonstrated that inhibiting GSK-3β leads to the activation of Wnt pathway thereby promoting neuroprotective effects, and mitigating cognitive deficits in AD animal models. The modulation of Wnt signalling appears to have multifaceted benefits including the reduction of amyloid-β production, tau hyperphosphorylation, enhancement of synaptic plasticity, and inhibition of neuroinflammation. These findings suggest that targeting GSK-3β to activate Wnt pathway may represent a novel approach for slowing or halting the progression of AD. This hypothesis reviews the current state of research exploring the activation of Wnt pathway through the inhibition of GSK-3β as a promising therapeutic strategy in AD.
Collapse
Affiliation(s)
- Magham Sai Varshini
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, India
| | - Ramakkamma Aishwarya Reddy
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, India
| | | |
Collapse
|
9
|
Turkistani A, Al-Kuraishy HM, Al-Gareeb AI, Albuhadily AK, Alexiou A, Papadakis M, Elfiky MM, Saad HM, Batiha GES. Therapeutic Potential Effect of Glycogen Synthase Kinase 3 Beta (GSK-3β) Inhibitors in Parkinson Disease: Exploring an Overlooked Avenue. Mol Neurobiol 2024; 61:7092-7108. [PMID: 38367137 PMCID: PMC11338983 DOI: 10.1007/s12035-024-04003-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 01/20/2024] [Indexed: 02/19/2024]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease of the brain due to degeneration of dopaminergic neurons in the substantia nigra (SN). Glycogen synthase kinase 3 beta (GSK-3β) is implicated in the pathogenesis of PD. Therefore, the purpose of the present review was to revise the mechanistic role of GSK-3β in PD neuropathology, and how GSK-3β inhibitors affect PD neuropathology. GSK-3 is a conserved threonine/serine kinase protein that is intricate in the regulation of cellular anabolic and catabolic pathways by modulating glycogen synthase. Over-expression of GSK-3β is also interconnected with the development of different neurodegenerative diseases. However, the underlying mechanism of GSK-3β in PD neuropathology is not fully clarified. Over-expression of GSK-3β induces the development of PD by triggering mitochondrial dysfunction and oxidative stress in the dopaminergic neurons of the SN. NF-κB and NLRP3 inflammasome are activated in response to dysregulated GSK-3β in PD leading to progressive neuronal injury. Higher expression of GSK-3β in the early stages of PD neuropathology might contribute to the reduction of neuroprotective brain-derived neurotrophic factor (BDNF). Thus, GSK-3β inhibitors may be effective in PD by reducing inflammatory and oxidative stress disorders which are associated with degeneration of dopaminergic in the SN.
Collapse
Affiliation(s)
- Areej Turkistani
- Department of Pharmacology and Toxicology, College of Medicine, Taif University, 21944, Taif, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
- Department of Research & Development, Funogen, Athens, Greece
- Department of Research & Development, AFNP Med, 1030, Vienna, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, Heusnerstrasse 40, University of Witten-Herdecke, 42283, Wuppertal, Germany.
| | - Mohamed M Elfiky
- Anatomy Department, General Medicine Practice Program, Batterjee Medical College, Jeddah, Saudi Arabia
- Anatomy Department, Faculty of Medicine, Menoufia University, Shibin El Kom, Al Minufya, Egypt
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Matrouh, 51744, Egypt
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
10
|
DiCesare SM, Ortega AJ, Collier GE, Daniel S, Thompson KN, McCoy MK, Posner BA, Hulleman JD. GSK3 inhibition reduces ECM production and prevents age-related macular degeneration-like pathology. JCI Insight 2024; 9:e178050. [PMID: 39114980 PMCID: PMC11383595 DOI: 10.1172/jci.insight.178050] [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: 11/30/2023] [Accepted: 06/20/2024] [Indexed: 08/22/2024] Open
Abstract
Malattia Leventinese/Doyne honeycomb retinal dystrophy (ML/DHRD) is an age-related macular degeneration-like (AMD-like) retinal dystrophy caused by an autosomal dominant R345W mutation in the secreted glycoprotein, fibulin-3 (F3). To identify new small molecules that reduce F3 production in retinal pigmented epithelium (RPE) cells, we knocked-in a luminescent peptide tag (HiBiT) into the endogenous F3 locus that enabled simple, sensitive, and high-throughput detection of the protein. The GSK3 inhibitor, CHIR99021 (CHIR), significantly reduced F3 burden (expression, secretion, and intracellular levels) in immortalized RPE and non-RPE cells. Low-level, long-term CHIR treatment promoted remodeling of the RPE extracellular matrix, reducing sub-RPE deposit-associated proteins (e.g., amelotin, complement component 3, collagen IV, and fibronectin), while increasing RPE differentiation factors (e.g., tyrosinase, and pigment epithelium-derived factor). In vivo, treatment of 8-month-old R345W+/+ knockin mice with CHIR (25 mg/kg i.p., 1 mo) was well tolerated and significantly reduced R345W F3-associated AMD-like basal laminar deposit number and size, thereby preventing the main pathological feature in these mice. This is an important demonstration of small molecule-based prevention of AMD-like pathology in ML/DHRD mice and may herald a rejuvenation of interest in GSK3 inhibition for the treatment of retinal degenerative diseases, including potentially AMD itself.
Collapse
Affiliation(s)
- Sophia M DiCesare
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Antonio J Ortega
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Gracen E Collier
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Steffi Daniel
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| | - Krista N Thompson
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Melissa K McCoy
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bruce A Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John D Hulleman
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
11
|
Mehrani R, Mondal J, Ghazanfari D, Goetz DJ, McCall KD, Bergmeier SC, Sharma S. Capturing the Effects of Single Atom Substitutions on the Inhibition Efficiency of Glycogen Synthase Kinase-3β Inhibitors via Markov State Modeling and Experiments. J Chem Theory Comput 2024; 20:6278-6286. [PMID: 38975986 DOI: 10.1021/acs.jctc.4c00311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024]
Abstract
Small modifications in the chemical structure of ligands are known to dramatically change their ability to inhibit the activity of a protein. Unraveling the mechanisms that govern these dramatic changes requires scrutinizing the dynamics of protein-ligand binding and unbinding at the atomic level. As an exemplary case, we have studied Glycogen Synthase Kinase-3β (GSK-3β), a multifunctional kinase that has been implicated in a host of pathological processes. As such, there is a keen interest in identifying ligands that inhibit GSK-3β activity. One family of compounds that are highly selective and potent inhibitors of GSK-3β is exemplified by a molecule termed COB-187. COB-187 consists of a five-member heterocyclic ring with a thione at C2, a pyridine substituted methyl at N3, and a hydroxyl and phenyl at C4. We have studied the inhibition of GSK-3β by COB-187-related ligands that differ in a single heavy atom from each other (either in the location of nitrogen in their pyridine ring, or with the pyridine ring replaced by a phenyl ring), or in the length of the alkyl group joining the pyridine and the N3. The inhibition experiments show a large range of half-maximal inhibitory concentration (IC50) values from 10 nM to 10 μM, implying that these ligands exhibit vastly different propensities to inhibit GSK-3β. To explain these differences, we perform Markov State Modeling (MSM) using fully atomistic simulations. Our MSM results are in excellent agreement with the experiments in that they accurately capture differences in the binding propensities of the ligands. The simulations show that the binding propensities are related to the ligands' ability to attain a compact conformation where their two aromatic rings are spatially close. We rationalize this result by sampling numerous binding and unbinding events via funnel metadynamics simulations, which show that indeed while approaching the bound state, the ligands prefer to be in their compact conformation. We find that the presence of nitrogen in the aromatic ring increases the probability of attaining the compact conformation. Protein-ligand binding is understood to be dictated by the energetics of interactions and entropic factors, like the release of bound water from the binding pockets. This work shows that changes in the conformational distribution of ligands due to atom-level modifications in the structure play an important role in protein-ligand binding.
Collapse
Affiliation(s)
- Ramin Mehrani
- Department of Mechanical Engineering, Ohio University, Athens, Ohio 45701, United States
| | - Jagannath Mondal
- Center for Interdisciplinary Sciences, Tata Institute of Fundamental Research, Hyderabad 500046, India
| | - Davoud Ghazanfari
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, Ohio 45701, United States
| | - Douglas J Goetz
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, Ohio 45701, United States
- Biomedical Engineering Program, Ohio University, Athens, Ohio 45701, United States
| | - Kelly D McCall
- Biomedical Engineering Program, Ohio University, Athens, Ohio 45701, United States
- Department of Specialty Medicine, Ohio University, Athens, Ohio 45701, United States
- The Diabetes Institute, Ohio University, Athens, Ohio 45701, United States
- Molecular and Cellular Biology Program, Ohio University, Athens, Ohio 45701, United States
- Translational Biomedical Sciences Program, Ohio University, Athens, Ohio 45701, United States
| | - Stephen C Bergmeier
- Biomedical Engineering Program, Ohio University, Athens, Ohio 45701, United States
- Department of Chemistry and Biochemistry, Ohio University, Athens, Ohio 45701, United States
| | - Sumit Sharma
- Department of Chemical and Biomolecular Engineering, Ohio University, Athens, Ohio 45701, United States
| |
Collapse
|
12
|
Singh A, Singh D, Tiwari N, Mittal P, Siddiqui MH, Mittal N. Exploring the therapeutic potential of rosemary compounds against Alzheimer's disease through GC-MS and molecular docking analysis. In Silico Pharmacol 2024; 12:63. [PMID: 39035101 PMCID: PMC11254900 DOI: 10.1007/s40203-024-00238-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024] Open
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disorder that is the leading cause of dementia in elderly individuals. Currently, there is no permanent treatment option available for this disorder, and the existing drug regimens are associated with limited effectiveness and side effects. To evaluate the neuroprotective effect of rosemary compounds, an extensive study was started with gas chromatography-mass spectrometry (GC-MS) analysis. GC-MS was performed to study the composition of rosemary essential oil and a total of 120 volatile compounds were identified. The 36 compounds from GC-MS data of rosemary essential oil having > 1% concentration in the oil were selected along with 3 already reported well-known non-volatile compounds of rosemary. se39 bioactive natural compounds of rosemary were docked against ACE, BACE1, GSK3, and TACE proteins, which are involved in AD progression. The top 3 compounds against each target protein were selected based on their binding energies and a total of 6 compounds were found as best candidates to target the AD; α Amyrin, Rosmanol, Androsta-1,4-dien-3-one,16,17-dihydroxy-(16.beta.,17.beta), Benzenesulfonamide,4-methyl-N-(5-nitro-2-pyridinyl), Methyl abietate, and Rosmarinic acid were the best compounds. The binding energy of α-Amyrin, Rosmanol, and Androsta-1,4-dien-3-one,16,17-dihydroxy-(16.beta.,17.beta) to ACE target is -10 kcal/mol, -9.3 kcal/mol, and - 9.3 kcal/mol, respectively. The best binding affinity was shown by complexes formed between GSK3-α-Amyrin (-9.1 kcal/mol), BACE1- α-Amyrin (-9.9 kcal/mol), and TACE- Benzenesulfonamide,4-methyl-N-(5-nitro-2-pyridinyl) (-9.1 kcal/mol). The comparative analysis between known inhibitors/ drugs of target proteins and the rosemary compound that shows the highest binding affinity against each protein also revealed the higher potential of rosemary natural compounds in terms of binding energy. The drug-likeliness properties like Lipinski's rule of five and the ADME/T analysis of top-selected compounds were screened through PkCSM and Deep-PK tools. The findings from this study suggested that rosemary compounds have the potential as a therapeutic lead for treating AD. This kind of experimental confirmation can lead to novel drug candidates against the pharmacological targets of AD. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00238-9.
Collapse
Affiliation(s)
- Anjali Singh
- Faculty of Biosciences, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Lucknow-Dewa Road, Barabanki, Uttar Pradesh 225003 India
| | - Dhananjay Singh
- Department of Biosciences, Integral University, Kursi Road, Lucknow, Uttar Pradesh 226026 India
| | - Neeraj Tiwari
- Faculty of Biosciences, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Lucknow-Dewa Road, Barabanki, Uttar Pradesh 225003 India
| | - Pooja Mittal
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401 India
| | - Mohammed Haris Siddiqui
- Department of Bioengineering, Integral University, Kursi Road, Lucknow, Uttar Pradesh 226026 India
| | - Nishu Mittal
- Faculty of Biosciences, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Lucknow-Dewa Road, Barabanki, Uttar Pradesh 225003 India
| |
Collapse
|
13
|
Wongjaikam S, Nopparat C, Boontem P, Panmanee J, Thasana N, Shukla M, Govitrapong P. Huperzine A Regulates the Physiological Homeostasis of Amyloid Precursor Protein Proteolysis and Tau Protein Conformation-A Computational and Experimental Investigation. BIOLOGY 2024; 13:518. [PMID: 39056711 PMCID: PMC11273828 DOI: 10.3390/biology13070518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/05/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024]
Abstract
The beneficial actions of the natural compound Huperzine A (Hup A) against age-associated learning and memory deficits promote this compound as a nootropic agent. Alzheimer's disease (AD) pathophysiology is characterized by the accumulation of amyloid beta (Aβ). Toxic Aβ oligomers account for the cognitive dysfunctions much before the pathological lesions are manifested in the brain. In the present study, we investigated the effects of Hup A on amyloid precursor protein (APP) proteolysis in SH-SY5Y neuroblastoma cells. Hup A downregulated the expression of β-site amyloid precursor protein cleaving enzyme 1 (BACE1) and presenilin 1 (PS1) levels but augmented the levels of A disintegrin and metalloproteinase 10 (ADAM10) with significant decrement in the Aβ levels. We herein report for the first time an in silico molecular docking analysis that revealed that Hup A binds to the functionally active site of BACE1. We further analyzed the effect of Hup A on glycogen synthase kinase-3 β (GSK3β) and phosphorylation status of tau. In this scenario, based on the current observations, we propose that Hup A is a potent regulator of APP processing and capable of modulating tau homeostasis under physiological conditions holding immense potential in preventing and treating AD like disorders.
Collapse
Affiliation(s)
- Suwakon Wongjaikam
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok 10210, Thailand; (S.W.)
- Cell and Animal Model Unit, Institute of Nutrition, Mahidol University, Nakhonpathom 73170, Thailand
| | - Chutikorn Nopparat
- Innovative Learning Center, Srinakharinwirot University, Sukhumvit 23, Bangkok 10110, Thailand
| | - Parichart Boontem
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok 10210, Thailand; (S.W.)
| | - Jiraporn Panmanee
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Nakhonpathom 73170, Thailand
| | - Nopporn Thasana
- Program in Chemical Sciences, Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok 10210, Thailand
- Laboratory of Medicinal Chemistry, Chulabhorn Research Institute, Bangkok 10210, Thailand
| | - Mayuri Shukla
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok 10210, Thailand; (S.W.)
| | - Piyarat Govitrapong
- Chulabhorn Graduate Institute, Chulabhorn Royal Academy, Bangkok 10210, Thailand; (S.W.)
| |
Collapse
|
14
|
Sequeira RC, Godad A. Understanding Glycogen Synthase Kinase-3: A Novel Avenue for Alzheimer's Disease. Mol Neurobiol 2024; 61:4203-4221. [PMID: 38064104 DOI: 10.1007/s12035-023-03839-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 11/28/2023] [Indexed: 07/11/2024]
Abstract
Alzheimer's Disease (AD) is the most prevalent form of age-related dementia. Even though a century has passed since the discovery of AD, the exact cause of the disease still remains unknown. As a result, this poses a major hindrance in developing effective therapies for treating AD. Glycogen synthase kinase-3 (GSK-3) is one of the kinases that has been investigated recently as a potential therapeutic target for the treatment of AD. It is also known as human tau protein kinase and is a proline-directed serine-threonine kinase. Since dysregulation of this kinase affects all the major characteristic features of the disease, such as tau phosphorylation, amyloid formation, memory, and synaptic function, it is thought to be a major player in the pathogenesis of AD. In this review, we present the most recent information on the role of this kinase in the onset and progression of AD, as well as significant findings that identify GSK-3 as one of the most important targets for AD therapy. We further discuss the potential of treating AD by targeting GSK-3 and give an overview of the ongoing studies aimed at developing GSK-3 inhibitors in preclinical and clinical investigations.
Collapse
Affiliation(s)
- Ronnita C Sequeira
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Gate No.1, Mithibai College Campus, Vaikunthlal Mehta Rd, Vile Parle West, Mumbai, Maharashtra, 400056, India
| | - Angel Godad
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Gate No.1, Mithibai College Campus, Vaikunthlal Mehta Rd, Vile Parle West, Mumbai, Maharashtra, 400056, India.
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India.
| |
Collapse
|
15
|
Yi LX, Zeng L, Wang Q, Tan EK, Zhou ZD. Reelin links Apolipoprotein E4, Tau, and Amyloid-β in Alzheimer's disease. Ageing Res Rev 2024; 98:102339. [PMID: 38754634 DOI: 10.1016/j.arr.2024.102339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder that affects the cerebral cortex and hippocampus, and is characterised by progressive cognitive decline and memory loss. A recent report of a patient carrying a novel gain-of-function variant of RELN (H3447R, termed RELN-COLBOS) who developed resilience against presenilin-linked autosomal-dominant AD (ADAD) has generated enormous interest. The RELN-COLBOS variant enhances interactions with the apolipoprotein E receptor 2 (ApoER2) and very-low-density lipoprotein receptor (VLDLR), which are associated with delayed AD onset and progression. These findings were validated in a transgenic mouse model. Reelin is involved in neurodevelopment, neurogenesis, and neuronal plasticity. The evidence accumulated thus far has demonstrated that the Reelin pathway links apolipoprotein E4 (ApoE4), amyloid-β (Aβ), and tubulin-associated unit (Tau), which are key proteins that have been implicated in AD pathogenesis. Reelin and key components of the Reelin pathway have been highlighted as potential therapeutic targets and biomarkers for AD.
Collapse
Affiliation(s)
- Ling Xiao Yi
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore 30843, Singapore
| | - Li Zeng
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore 30843, Singapore; Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Eng King Tan
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore 30843, Singapore; Department of Neurology, Singapore General Hospital, Outram Road, Singapore 169608, Singapore; Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore.
| | - Zhi Dong Zhou
- National Neuroscience Institute of Singapore, 11 Jalan Tan Tock Seng, Singapore 30843, Singapore; Signature Research Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore.
| |
Collapse
|
16
|
El Hajjar L, Page A, Bridot C, Cantrelle FX, Landrieu I, Smet-Nocca C. Regulation of Glycogen Synthase Kinase-3β by Phosphorylation and O-β-Linked N-Acetylglucosaminylation: Implications on Tau Protein Phosphorylation. Biochemistry 2024; 63:1513-1533. [PMID: 38788673 DOI: 10.1021/acs.biochem.4c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Glycogen synthase kinase 3 (GSK3) plays a pivotal role in signaling pathways involved in insulin metabolism and the pathogenesis of neurodegenerative disorders. In particular, the GSK3β isoform is implicated in Alzheimer's disease (AD) as one of the key kinases involved in the hyperphosphorylation of tau protein, one of the neuropathological hallmarks of AD. As a constitutively active serine/threonine kinase, GSK3 is inactivated by Akt/PKB-mediated phosphorylation of Ser9 in the N-terminal disordered domain, and for most of its substrates, requires priming (prephosphorylation) by another kinase that targets the substrate to a phosphate-specific pocket near the active site. GSK3 has also been shown to be post-translationally modified by O-linked β-N-acetylglucosaminylation (O-GlcNAcylation), with still unknown functions. Here, we have found that binding of Akt inhibits GSK3β kinase activity on both primed and unprimed tau substrates. Akt-mediated Ser9 phosphorylation restores the GSK3β kinase activity only on primed tau, thereby selectively inactivating GSK3β toward unprimed tau protein. Additionally, we have shown that GSK3β is highly O-GlcNAcylated at multiple sites within the kinase domain and the disordered N- and C-terminal domains, including Ser9. In contrast to Akt-mediated regulation, neither the O-GlcNAc transferase nor O-GlcNAcylation significantly alters GSK3β kinase activity, but high O-GlcNAc levels reduce Ser9 phosphorylation by Akt. Reciprocally, Akt phosphorylation downregulates the overall O-GlcNAcylation of GSK3β, indicating a crosstalk between both post-translational modifications. Our results indicate that specific O-GlcNAc profiles may be involved in the phosphorylation-dependent Akt-mediated regulation of GSK3β kinase activity.
Collapse
Affiliation(s)
- Léa El Hajjar
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| | - Adeline Page
- Protein Science Facility, SFR Biosciences Univ Lyon, ENS de Lyon, CNRS UAR3444, Inserm US8, Université Claude Bernard Lyon 1, 50 Avenue Tony Garnier, Lyon F-69007, France
| | - Clarisse Bridot
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| | - François-Xavier Cantrelle
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| | - Isabelle Landrieu
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| | - Caroline Smet-Nocca
- Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, University of Lille, Lille F-59000, France
- CNRS EMR9002 Integrative Structural Biology, Lille F-59000, France
| |
Collapse
|
17
|
Sindi G, Ismael S, Uddin R, Slepchenko KG, Colvin RA, Lee D. Endogenous tau released from human ReNCell VM cultures by neuronal activity is phosphorylated at multiple sites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.02.597022. [PMID: 38854111 PMCID: PMC11160771 DOI: 10.1101/2024.06.02.597022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Tau is an intracellular protein but also known to be released into the extracellular fluid. Tau release mechanisms have drawn intense attention as these are known to play a key role in Alzheimer's disease (AD) pathology. However, tau can also be released under physiological conditions although its physiological function and release mechanisms have been poorly characterized, especially in human neuronal cells. We investigated endogenous tau release in ReNCell VM, a human neuroprogenitor cell line, under physiological conditions and found that tau is spontaneously released from cells. To study activity-dependent release of endogenous tau, human ReNCell VM culture was stimulated by 100μM AMPA or 50mM KCl for one-hour, tau was actively released to the culture medium. The released tau was highly phosphorylated at nine phosphorylation sites (pSites) detected by phospho-specific tau antibodies including AT270 (T175/T181), AT8 (S202/T205), AT100 (T212/S214), AT180 (T231), and PHF-1 (S396/S404), showing that these pSites are important for activity-dependent tau release from human ReNCell VM. Intracellular tau showed various phosphorylation status across these sites, with AT270 and PHF-1 highly phosphorylated while AT8 and AT180 were minimally phosphorylated, suggesting that AT8 and AT180 pSites exhibit a propensity for secretion rather than being retained intracellularly. This activity-dependent tau release was significantly decreased by inhibition of GSK-3β, demonstrating that GSK3β-dependent phosphorylation of tau plays an important role in its release by neuronal activity. In this study, we showed that ReNCell VM serves as a valuable model for studying endogenous physiological tau release. Further, ReNCell model can be also used to study pathological release of human tau that will contribute to our understanding of the progression of AD and related dementias.
Collapse
Affiliation(s)
| | - Sazan Ismael
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Reaz Uddin
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Kira G. Slepchenko
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Robert A. Colvin
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Daewoo Lee
- Neuroscience Program, Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
18
|
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
|
19
|
Schneider NO, Gilreath K, Burkett DJ, St. Maurice M, Donaldson WA. Synthesis and Evaluation of 5-(Heteroarylmethylene)hydantoins as Glycogen Synthase Kinase-3β Inhibitors. Pharmaceuticals (Basel) 2024; 17:570. [PMID: 38794140 PMCID: PMC11123921 DOI: 10.3390/ph17050570] [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: 03/13/2024] [Revised: 04/21/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase which plays a center role in the phosphorylation of a wide variety of proteins, generally leading to their inactivation. As such, GSK-3 is viewed as a therapeutic target. An ever-increasing number of small organic molecule inhibitors of GSK-3 have been reported. Phenylmethylene hydantoins are known to exhibit a wide range of inhibitory activities including for GSK-3β. A family of fourteen 2-heterocycle substituted methylene hydantoins (14, 17-29) were prepared and evaluated for the inhibition of GSK-3β at 25 μM. The IC50 values of five of these compounds was determined; the two best inhibitors are 5-[(4'-chloro-2-pyridinyl)methylene]hydantoin (IC50 = 2.14 ± 0.18 μM) and 5-[(6'-bromo-2-pyridinyl)methylene]hydantoin (IC50 = 3.39 ± 0.16 μM). The computational docking of the compounds with GSK-3β (pdb 1q41) revealed poses with hydrogen bonding to the backbone at Val135. The 5-[(heteroaryl)methylene]hydantoins did not strongly inhibit other metalloenzymes, demonstrating poor inhibitory activity against matrix metalloproteinase-12 at 25 μM and against human carbonic anhydrase at 200 μM, and were not inhibitors for Staphylococcus aureus pyruvate carboxylase at concentrations >1000 μM.
Collapse
Affiliation(s)
- Nicholas O. Schneider
- Department of Biological Sciences, Marquette University, P.O. Box 1881, Milwaukee, WI 53201, USA
| | - Kendra Gilreath
- Department of Chemistry, Marquette University, P.O. Box 1881, Milwaukee, WI 53201, USA
| | - Daniel J. Burkett
- Department of Chemistry, Marquette University, P.O. Box 1881, Milwaukee, WI 53201, USA
| | - Martin St. Maurice
- Department of Biological Sciences, Marquette University, P.O. Box 1881, Milwaukee, WI 53201, USA
| | - William A. Donaldson
- Department of Chemistry, Marquette University, P.O. Box 1881, Milwaukee, WI 53201, USA
| |
Collapse
|
20
|
Farid HA, Sayed RH, El-Shamarka MES, Abdel-Salam OME, El Sayed NS. PI3K/AKT signaling activation by roflumilast ameliorates rotenone-induced Parkinson's disease in rats. Inflammopharmacology 2024; 32:1421-1437. [PMID: 37541971 PMCID: PMC11006765 DOI: 10.1007/s10787-023-01305-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 07/20/2023] [Indexed: 08/06/2023]
Abstract
Parkinson's disease (PD) is the second most common progressive age-related neurodegenerative disorder. Paramount evidence shed light on the role of PI3K/AKT signaling activation in the treatment of neurodegenerative disorders. PI3K/AKT signaling can be activated via cAMP-dependent pathways achieved by phosphodiesterase 4 (PDE4) inhibition. Roflumilast is a well-known PDE4 inhibitor that is currently used in the treatment of chronic obstructive pulmonary disease. Furthermore, roflumilast has been proposed as a favorable candidate for the treatment of neurological disorders. The current study aimed to unravel the neuroprotective role of roflumilast in the rotenone model of PD in rats. Ninety male rats were allocated into six groups as follows: control, rotenone (1.5 mg/kg/48 h, s.c.), L-dopa (22.5 mg/kg, p.o), and roflumilast (0.2, 0.4 or 0.8 mg/kg, p.o). All treatments were administrated for 21 days 1 h after rotenone injection. Rats treated with roflumilast showed an improvement in motor activity and coordination as well as preservation of dopaminergic neurons in the striatum. Moreover, roflumilast increased cAMP level and activated the PI3K/AKT axis via stimulation of CREB/BDNF/TrkB and SIRT1/PTP1B/IGF1 signaling cascades. Roflumilast also caused an upsurge in mTOR and Nrf2, halted GSK-3β and NF-ĸB, and suppressed FoxO1 and caspase-3. Our study revealed that roflumilast exerted neuroprotective effects in rotenone-induced neurotoxicity in rats. These neuroprotective effects were mediated via the crosstalk between CREB/BDNF/TrkB and SIRT1/PTP1B/IGF1 signaling pathways which activates PI3K/AKT trajectory. Therefore, PDE4 inhibition is likely to offer a reliable persuasive avenue in curing PD via PI3K/AKT signaling activation.
Collapse
Affiliation(s)
- Heba A Farid
- Department of Narcotics, Ergogenic Aids and Poisons, National Research Centre, Cairo, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt.
| | | | - Omar M E Abdel-Salam
- Department of Narcotics, Ergogenic Aids and Poisons, National Research Centre, Cairo, Egypt
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El Aini St., Cairo, 11562, Egypt
| |
Collapse
|
21
|
Diaz AP, Canal CAM, Valdés AJ, Delgado JEG, Varela-M RE. GSK-3 kinase a putative therapeutic target in trypanosomatid parasites. Braz J Infect Dis 2024; 28:103736. [PMID: 38467387 PMCID: PMC10955101 DOI: 10.1016/j.bjid.2024.103736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 02/11/2024] [Accepted: 02/22/2024] [Indexed: 03/13/2024] Open
Abstract
Trypanosomatids are an important group of parasites that predominate in tropical and subtropical areas of the planet, which cause diseases that are classified as forgotten and neglected by the world health organization. In this group of parasites, we find Trypanosoma cruzi, Trypanosoma brucei, Trypanosoma brucei rhodesiense and Leishmania spp, for which there is no vaccine available, and its control has focused mainly on pharmacological treatment. Due to the poverty situation where these diseases are found and the biological complexity of these parasites, there are multiple variables to control, including the diversity of species, the complexity of their life cycles, drug resistance, cytotoxicity, the limited use in pregnant women, the high costs of treatment and the little-known pharmacological mechanisms of action, among others. It is therefore necessary to find new strategies and approaches for the treatment of these parasitic diseases. Among these new approaches is the rational search for new targets based on the allosteric inhibition of protein kinases, which have been little studied in trypanosomatids. Among these kinases, we find Glycogen Synthase Kinase-3 (GSK-3), a kinase of great pharmacological interest, which is under intense basic and clinical research by pharmaceutical companies for the treatment of cancer. This kinase, highly studied in the PI3K/AKT/mTOR pathway signaling in humans, has an orthologous gene in these parasites (GSK-3 s), which has proven to be essential for them in response to different challenges; Therefore, it is notable to increase research in this kinase in order to achieve a broad structural and functional characterization in the different species of trypanosomatids.
Collapse
Affiliation(s)
| | | | | | | | - R E Varela-M
- Laboratory of Parasitology and Tropical Diseases, Faculty of Basic Sciences, Universidad Santiago de Cali, Cali, Colombia.
| |
Collapse
|
22
|
Atienza-Navarro I, Del Marco A, Alves-Martinez P, Garcia-Perez MDLA, Raya-Marin A, Benavente-Fernandez I, Gil C, Martinez A, Lubian-Lopez S, Garcia-Alloza M. Glycogen Synthase Kinase-3β Inhibitor VP3.15 Ameliorates Neurogenesis, Neuronal Loss and Cognitive Impairment in a Model of Germinal Matrix-intraventricular Hemorrhage of the Preterm Newborn. Transl Stroke Res 2024:10.1007/s12975-023-01229-2. [PMID: 38231413 DOI: 10.1007/s12975-023-01229-2] [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: 10/16/2023] [Revised: 11/24/2023] [Accepted: 12/18/2023] [Indexed: 01/18/2024]
Abstract
Advances in neonatology have significantly reduced mortality rates due to prematurity. However, complications of prematurity have barely changed in recent decades. Germinal matrix-intraventricular hemorrhage (GM-IVH) is one of the most severe complications of prematurity, and these children are prone to suffer short- and long-term sequelae, including cerebral palsy, cognitive and motor impairments, or neuropsychiatric disorders. Nevertheless, GM-IVH has no successful treatment. VP3.15 is a small, heterocyclic molecule of the 5-imino-1,2,4-thiadiazole family with a dual action as a phosphodiesterase 7 and glycogen synthase kinase-3β (GSK-3β) inhibitor. VP3.15 reduces neuroinflammation and neuronal loss in other neurodegenerative disorders and might ameliorate complications associated with GM-IVH. We administered VP3.15 to a mouse model of GM-IVH. VP3.15 reduces the presence of hemorrhages and microglia in the short (P14) and long (P110) term. It ameliorates brain atrophy and ventricle enlargement while limiting tau hyperphosphorylation and neuronal and myelin basic protein loss. VP3.15 also improves proliferation and neurogenesis as well as cognition after the insult. Interestingly, plasma gelsolin levels, a feasible biomarker of brain damage, improved after VP3.15 treatment. Altogether, our data support the beneficial effects of VP3.15 in GM-IVH by ameliorating brain neuroinflammatory, vascular and white matter damage, ultimately improving cognitive impairment associated with GM-IVH.
Collapse
Affiliation(s)
- Isabel Atienza-Navarro
- Division of Physiology, School of Medicine, University of Cadiz, C/Dr. Marañon 3, 3rd Floor, 11002, Cadiz, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA) Research Unit, Puerta del Mar University Hospital, Cadiz, Spain
| | - Angel Del Marco
- Division of Physiology, School of Medicine, University of Cadiz, C/Dr. Marañon 3, 3rd Floor, 11002, Cadiz, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA) Research Unit, Puerta del Mar University Hospital, Cadiz, Spain
| | - Pilar Alves-Martinez
- Division of Physiology, School of Medicine, University of Cadiz, C/Dr. Marañon 3, 3rd Floor, 11002, Cadiz, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA) Research Unit, Puerta del Mar University Hospital, Cadiz, Spain
| | | | - Alvaro Raya-Marin
- Biomedical Research and Innovation Institute of Cadiz (INiBICA) Research Unit, Puerta del Mar University Hospital, Cadiz, Spain
| | - Isabel Benavente-Fernandez
- Area of Pediatrics, Department of Child and Mother Health and Radiology, School of Medicine, University of Cadiz, Cadiz, Spain
- Section of Neonatology, Division of Pediatrics, Puerta del Mar University Hospital, Avda. Ana de Viya sn, 11007, Cadiz, Spain
| | - Carmen Gil
- Centro de Investigaciones, Biologicas Margarita Salas-CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Ana Martinez
- Centro de Investigaciones, Biologicas Margarita Salas-CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
- Centro de Investigaciones Biomedicas en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Avda. Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Simon Lubian-Lopez
- Area of Pediatrics, Department of Child and Mother Health and Radiology, School of Medicine, University of Cadiz, Cadiz, Spain.
- Section of Neonatology, Division of Pediatrics, Puerta del Mar University Hospital, Avda. Ana de Viya sn, 11007, Cadiz, Spain.
| | - Monica Garcia-Alloza
- Division of Physiology, School of Medicine, University of Cadiz, C/Dr. Marañon 3, 3rd Floor, 11002, Cadiz, Spain.
- Biomedical Research and Innovation Institute of Cadiz (INiBICA) Research Unit, Puerta del Mar University Hospital, Cadiz, Spain.
| |
Collapse
|
23
|
Mohite R, Gharat S, Doshi G. Unraveling the Role of the Glycogen Synthase Kinase-3β, Bruton's Tyrosine Kinase, and Sphingosine 1 Phosphate Pathways in Multiple Sclerosis. Endocr Metab Immune Disord Drug Targets 2024; 24:1131-1145. [PMID: 38284723 DOI: 10.2174/0118715303261413231117113707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 10/16/2023] [Accepted: 10/20/2023] [Indexed: 01/30/2024]
Abstract
Inflammation, demyelination, and neurodegeneration are symptoms of the central nervous system (CNS) condition known as Multiple sclerosis (MS). Due to its crucial function in controlling immune cell activation and inflammation, the glycogen synthase kinase-3β (GSK- 3β), Bruton's tyrosine kinase (BTK), and Sphingosine 1 phosphate (S1P) signaling pathway have become a viable target for the therapy of MS. The GSK-3β signaling system, which controls several biological target processes, including cell survival, proliferation, and inflammation, depends on the GSK-3β enzyme. In MS animal models and human studies, GSK-3β inhibition has been demonstrated to lessen demyelination and inflammation. Clinical research on MS has demonstrated that BTK inhibitors decrease inflammation and disease activity by preventing B cell activation and the subsequent release of cytokines. Clinical investigations for MS have demonstrated that S1P modulators, such as fingolimod, lower disease activity and inflammation by limiting immune cell migration to the central nervous system and preventing cytokine production. The GSK-3β /BTK/S1P signaling pathway in MS is the subject of this paper's summary and discussion of prospective treatment targets.
Collapse
Affiliation(s)
- Rupali Mohite
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, India
| | - Sankalp Gharat
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, India
| | - Gaurav Doshi
- Department of Pharmacology, Toxicology and Therapeutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V.M. Road, Vile Parle (W), Mumbai, India
| |
Collapse
|
24
|
Lai S, Wang P, Gong J, Zhang S. New insights into the role of GSK-3β in the brain: from neurodegenerative disease to tumorigenesis. PeerJ 2023; 11:e16635. [PMID: 38107562 PMCID: PMC10722984 DOI: 10.7717/peerj.16635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/18/2023] [Indexed: 12/19/2023] Open
Abstract
Glycogen synthase kinase 3 (GSK-3) is a serine/threonine kinase widely expressed in various tissues and organs. Unlike other kinases, GSK-3 is active under resting conditions and is inactivated upon stimulation. In mammals, GSK-3 includes GSK-3 α and GSK-3β isoforms encoded by two homologous genes, namely, GSK3A and GSK3B. GSK-3β is essential for the control of glucose metabolism, signal transduction, and tissue homeostasis. As more than 100 known proteins have been identified as GSK-3β substrates, it is sometimes referred to as a moonlighting kinase. Previous studies have elucidated the regulation modes of GSK-3β. GSK-3β is involved in almost all aspects of brain functions, such as neuronal morphology, synapse formation, neuroinflammation, and neurological disorders. Recently, several comparatively specific small molecules have facilitated the chemical manipulation of this enzyme within cellular systems, leading to the discovery of novel inhibitors for GSK-3β. Despite these advancements, the therapeutic significance of GSK-3β as a drug target is still complicated by uncertainties surrounding the potential of inhibitors to stimulate tumorigenesis. This review provides a comprehensive overview of the intricate mechanisms of this enzyme and evaluates the existing evidence regarding the therapeutic potential of GSK-3β in brain diseases, including Alzheimer's disease, Parkinson's disease, mood disorders, and glioblastoma.
Collapse
Affiliation(s)
- Shenjin Lai
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Peng Wang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| | - Jingru Gong
- Department of Pharmacy, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Shuaishuai Zhang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- Institute for Brain Research and Rehabilitation, South China Normal University, Guangzhou, China
| |
Collapse
|
25
|
Na D, Lim DH, Hong JS, Lee HM, Cho D, Yu MS, Shaker B, Ren J, Lee B, Song JG, Oh Y, Lee K, Oh KS, Lee MY, Choi MS, Choi HS, Kim YH, Bui JM, Lee K, Kim HW, Lee YS, Gsponer J. A multi-layered network model identifies Akt1 as a common modulator of neurodegeneration. Mol Syst Biol 2023; 19:e11801. [PMID: 37984409 DOI: 10.15252/msb.202311801] [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: 06/05/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/22/2023] Open
Abstract
The accumulation of misfolded and aggregated proteins is a hallmark of neurodegenerative proteinopathies. Although multiple genetic loci have been associated with specific neurodegenerative diseases (NDs), molecular mechanisms that may have a broader relevance for most or all proteinopathies remain poorly resolved. In this study, we developed a multi-layered network expansion (MLnet) model to predict protein modifiers that are common to a group of diseases and, therefore, may have broader pathophysiological relevance for that group. When applied to the four NDs Alzheimer's disease (AD), Huntington's disease, and spinocerebellar ataxia types 1 and 3, we predicted multiple members of the insulin pathway, including PDK1, Akt1, InR, and sgg (GSK-3β), as common modifiers. We validated these modifiers with the help of four Drosophila ND models. Further evaluation of Akt1 in human cell-based ND models revealed that activation of Akt1 signaling by the small molecule SC79 increased cell viability in all models. Moreover, treatment of AD model mice with SC79 enhanced their long-term memory and ameliorated dysregulated anxiety levels, which are commonly affected in AD patients. These findings validate MLnet as a valuable tool to uncover molecular pathways and proteins involved in the pathophysiology of entire disease groups and identify potential therapeutic targets that have relevance across disease boundaries. MLnet can be used for any group of diseases and is available as a web tool at http://ssbio.cau.ac.kr/software/mlnet.
Collapse
Affiliation(s)
- Dokyun Na
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Do-Hwan Lim
- College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- School of Systems Biomedical Science, Soongsil University, Seoul, Republic of Korea
| | - Jae-Sang Hong
- College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Hyang-Mi Lee
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Daeahn Cho
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Myeong-Sang Yu
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Bilal Shaker
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Jun Ren
- Department of Biomedical Engineering, Chung-Ang University, Seoul, Republic of Korea
| | - Bomi Lee
- College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Jae Gwang Song
- College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Yuna Oh
- Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Kyungeun Lee
- Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Kwang-Seok Oh
- Information-based Drug Research Center, Korea Research Institute of Chemical Technology, Deajeon, Republic of Korea
| | - Mi Young Lee
- Information-based Drug Research Center, Korea Research Institute of Chemical Technology, Deajeon, Republic of Korea
| | - Min-Seok Choi
- College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Han Saem Choi
- College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Yang-Hee Kim
- College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Jennifer M Bui
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Kangseok Lee
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| | - Hyung Wook Kim
- College of Life Sciences, Sejong University, Seoul, Republic of Korea
| | - Young Sik Lee
- College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jörg Gsponer
- Department of Biochemistry and Molecular Biology, Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
26
|
Tomita K, Kuwahara Y, Igarashi K, Kitanaka J, Kitanaka N, Takashi Y, Tanaka KI, Roudkenar MH, Roushandeh AM, Kurimasa A, Nishitani Y, Sato T. Therapeutic potential for KCC2-targeted neurological diseases. JAPANESE DENTAL SCIENCE REVIEW 2023; 59:431-438. [PMID: 38022385 PMCID: PMC10665825 DOI: 10.1016/j.jdsr.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/01/2023] [Accepted: 11/05/2023] [Indexed: 12/01/2023] Open
Abstract
Patients with neurological diseases, such as schizophrenia, tend to show low K+-Cl- co-transporter 2 (KCC2) levels in the brain. The cause of these diseases has been associated with stress and neuroinflammation. However, since the pathogenesis of these diseases is not yet fully investigated, drug therapy is still limited to symptomatic therapy. Targeting KCC2, which is mainly expressed in the brain, seems to be an appropriate approach in the treatment of these diseases. In this review, we aimed to discuss about stress and inflammation, KCC2 and Gamma-aminobutyric acid (GABA) function, diseases which decrease the KCC2 levels in the brain, factors that regulate KCC2 activity, and the possibility to overcome neuronal dysfunction targeting KCC2. We also aimed to discuss the relationships between neurological diseases and LPS caused by Porphyromonas gingivalis (P. g), which is a type of oral bacterium. Clinical trials on oxytocin, sirtuin 1 (SIRT1) activator, and transient receptor potential cation channel subfamily V Member 1 activator have been conducted to develop effective treatment methods. We believe that KCC2 modulators that regulate mitochondria, such as oxytocin, glycogen synthase kinase 3β (GSK3β), and SIRT1, can be potential targets for neurological diseases.
Collapse
Affiliation(s)
- Kazuo Tomita
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890–8544, Japan
- Division of Pharmacology, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, Hyogo 650–8530, Japan
| | - Yoshikazu Kuwahara
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890–8544, Japan
- Division of Radiation Biology and Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Miyagi, 983-8536, Japan
| | - Kento Igarashi
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890–8544, Japan
- Division of Pharmacology, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, Hyogo 650–8530, Japan
| | - Junichi Kitanaka
- Laboratory of Drug Addiction and Experimental Therapeutics, Schoolof Pharmacy, Hyogo Medical University, Hyogo 650-8530, Japan
| | - Nobue Kitanaka
- Laboratory of Drug Addiction and Experimental Therapeutics, Schoolof Pharmacy, Hyogo Medical University, Hyogo 650-8530, Japan
- Department of Pharmacology, School of Medicine, Hyogo Medical University, Hyogo 663-8501, Japan
| | - Yuko Takashi
- Department of Restorative Dentistry and Endodontology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890–8544, Japan
| | - Koh-ichi Tanaka
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890–8544, Japan
- Division of Pharmacology, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, Hyogo 650–8530, Japan
| | - Mehryar Habibi Roudkenar
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890–8544, Japan
- Burn and Regenerative Medicine Research Center, Velayat Hospital, School of Medicine, Guilan University of Medical Sciences, Rasht 41937–13194, Iran
| | - Amaneh Mohammadi Roushandeh
- Department of Anatomy, School of Biomedical Sciences, Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Akihiro Kurimasa
- Division of Radiation Biology and Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Miyagi, 983-8536, Japan
| | - Yoshihiro Nishitani
- Department of Restorative Dentistry and Endodontology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890–8544, Japan
| | - Tomoaki Sato
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890–8544, Japan
| |
Collapse
|
27
|
Borde S, Matosevic S. Metabolic adaptation of NK cell activity and behavior in tumors: challenges and therapeutic opportunities. Trends Pharmacol Sci 2023; 44:832-848. [PMID: 37770314 DOI: 10.1016/j.tips.2023.08.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/30/2023]
Abstract
The adaptation of natural killer (NK) cells to conditions in the microenvironment of tumors is deeply affected by their metabolic activity, itself a result of nutrient availability and the metabolism of the cancer cells themselves. Elevated rates of glycolysis and lipid metabolism in cancers not only lead to the accumulation of immunosuppressive byproducts but also contribute to an environment of elevated concentrations of extracellular metabolites. This results in altered NK cell bioenergetics through changes in transcriptional and translational profiles, ultimately affecting their pharmacology and impairing NK cell responses. However, understanding the metabolic processes that drive alterations in immunological signaling on NK cells remains both difficult and vastly underexplored. We discuss the varied and complex drivers of NK cell metabolism in homeostasis and the tumor microenvironment (TME), challenges associated with their targetability, and unexplored therapeutic opportunities.
Collapse
Affiliation(s)
- Shambhavi Borde
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, USA
| | - Sandro Matosevic
- Department of Industrial and Physical Pharmacy, Purdue University, West Lafayette, IN, USA; Center for Cancer Research, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
28
|
Saleh JS, Abd El Hadi SR, Ibrahim HS, Elrazaz EZ, Abouzid KAM. Fragment merging approach for design, synthesis, and biological assessment of urea/acetyl hydrazide clubbed thienopyrimidine derivatives as GSK-3β inhibitors. BMC Chem 2023; 17:127. [PMID: 37759329 PMCID: PMC10538245 DOI: 10.1186/s13065-023-01026-w] [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: 03/29/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
New thienopyrimidine derivatives were designed and synthesized as GSK-3β inhibitors based on the structure of active binding site of GSK-3β enzyme. In this study, compounds 6b and 6a were found to be moderate GSK-3β inhibitors with IC50s 10.2 and 17.3 μM, respectively. Molecular docking study was carried out by docking the targeted compounds in the binding site of the GSK-3β enzyme using the MOE program. Moreover, ADME study was performed to predict certain pharmacokinetic properties. The results showed that all synthesized compounds may not be able to penetrate the blood brain barrier; so, the chances of CNS side effects are predicted to be low. CYP1D6 is predicted to be inhibited by compounds (5a, 5d, 6a, 9a and 9b), So drug-drug interactions are expected upon administration of these compounds.
Collapse
Affiliation(s)
- Joseph S Saleh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Egyptian Russian University, P.O. Box 11829, Badr City, Cairo, Egypt.
| | - Soha R Abd El Hadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Egyptian Russian University, P.O. Box 11829, Badr City, Cairo, Egypt.
| | - Hany S Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Egyptian Russian University, P.O. Box 11829, Badr City, Cairo, Egypt
| | - Eman Z Elrazaz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University, P.O. Box 11566, Abbassia, Cairo, Egypt
| | - Khaled A M Abouzid
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University, P.O. Box 11566, Abbassia, Cairo, Egypt.
| |
Collapse
|
29
|
Chuang WH, Chou YT, Chen YH, Kuo TH, Liaw WF, Lu TT, Kao CF, Wang YM. Neuroprotective Effect of NO-Delivery Dinitrosyl Iron Complexes (DNICs) on Amyloid Pathology in the Alzheimer's Disease Cell Model. ACS Chem Neurosci 2023; 14:2922-2934. [PMID: 37533298 DOI: 10.1021/acschemneuro.3c00348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive impairment, memory loss, and behavioral deficits. β-amyloid1-42 (Aβ1-42) aggregation is a significant cause of the pathogenesis in AD. Despite the numerous types of research, the current treatment efficacy remains insufficient. Hence, a novel therapeutic strategy is required. Nitric oxide (NO) is a multifunctional gaseous molecule. NO displays a neuroprotective role in the central nervous system by inhibiting the Aβ aggregation and rescuing memory and learning deficit through the NO signaling pathway. Targeting the NO pathway might be a therapeutic option; however, NO has a limited half-life under the biological system. To address this issue, a biomimetic dinitrosyl iron complex [(NO)2Fe(μ-SCH2CH2COOH)2Fe(NO)2] (DNIC-COOH) that could stably deliver NO was explored in the current study. To determine whether DNIC-COOH exerts anti-AD efficacy, DNIC-COOH was added to neuron-like cells and primary cortical neurons along with Aβ1-42. This study found that DNIC-COOH protected neuronal cells from Aβ-induced cytotoxicity, potentiated neuronal functions, and facilitated Aβ1-42 degradation through the NO-sGC-cGMP-AKT-GSK3β-CREB/MMP-9 pathway.
Collapse
Affiliation(s)
- Wen-Han Chuang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yu-Ting Chou
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yi-Hong Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Ting-Han Kuo
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Wen-Feng Liaw
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Tsai-Te Lu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
- Department of Chemistry, Chung Yuan Christian University, Taoyuan 32023, Taiwan
| | - Chih-Fei Kao
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Yun-Ming Wang
- Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- Department of Biomedical Science and Environmental Biology, Department of Dentistry, Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
30
|
Passero M, Zhai T, Huang Z. Investigation of Potential Drug Targets for Cholesterol Regulation to Treat Alzheimer's Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6217. [PMID: 37444065 PMCID: PMC10341567 DOI: 10.3390/ijerph20136217] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023]
Abstract
Despite extensive research and seven approved drugs, the complex interplay of genes, proteins, and pathways in Alzheimer's disease remains a challenge. This implies the intricacies of the mechanism for Alzheimer's disease, which involves the interaction of hundreds of genes, proteins, and pathways. While the major hallmarks of Alzheimer's disease are the accumulation of amyloid plaques and tau protein tangles, excessive accumulation of cholesterol is reportedly correlated with Alzheimer's disease patients. In this work, protein-protein interaction analysis was conducted based upon the genes from a clinical database to identify the top protein targets with most data-indicated involvement in Alzheimer's disease, which include ABCA1, CYP46A1, BACE1, TREM2, GSK3B, and SREBP2. The reactions and pathways associated with these genes were thoroughly studied for their roles in regulating brain cholesterol biosynthesis, amyloid beta accumulation, and tau protein tangle formation. Existing clinical trials for each protein target were also investigated. The research indicated that the inhibition of SREBP2, BACE1, or GSK3B is beneficial to reduce cholesterol and amyloid beta accumulation, while the activation of ABCA1, CYP46A1, or TREM2 has similar effects. In this study, Sterol Regulatory Element-Binding Protein 2 (SREBP2) emerged as the primary protein target. SREBP2 serves a pivotal role in maintaining cholesterol balance, acting as a transcription factor that controls the expression of several enzymes pivotal for cholesterol biosynthesis. Novel studies suggest that SREBP2 performs a multifaceted role in Alzheimer's disease. The hyperactivity of SREBP2 may lead to heightened cholesterol biosynthesis, which suggested association with the pathogenesis of Alzheimer's disease. Lowering SREBP2 levels in an Alzheimer's disease mouse model results in reduced production of amyloid-beta, a major contributor to Alzheimer's disease progression. Moreover, its thoroughly analyzed crystal structure allows for computer-aided screening of potential inhibitors; SREBP2 is thus selected as a prospective drug target. While more protein targets can be added onto the list in the future, this work provides an overview of key proteins involved in the regulation of brain cholesterol biosynthesis that may be further investigated for Alzheimer's disease intervention.
Collapse
Affiliation(s)
| | | | - Zuyi Huang
- Department of Chemical Engineering, Villanova University, Villanova, PA 19085, USA
| |
Collapse
|
31
|
Catalano T, Selvaggi F, Esposito DL, Cotellese R, Aceto GM. Infectious Agents Induce Wnt/β-Catenin Pathway Deregulation in Primary Liver Cancers. Microorganisms 2023; 11:1632. [PMID: 37512809 PMCID: PMC10386003 DOI: 10.3390/microorganisms11071632] [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: 05/25/2023] [Revised: 06/18/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Interaction between infectious agents and liver tissue, as well as repeated and extreme biological events beyond adaptive capacities, may result in pathological conditions predisposing people to development of primary liver cancers (PLCs). In adults, PLCs mainly comprise hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA). Various infectious agents in the hepatic microenvironment can destabilize normal liver cell functions by modulating the Wnt/β-catenin pathway components. Among them, hepatotropic viruses B, C, and D are involved in Wnt/β-catenin signaling dysregulation. Other microbial agents, including oncogenic viruses such as Epstein-Barr virus (EBV) and human papilloma virus (HPV), bacteria, e.g., Mycoplasma hyorhinis and Salmonella Typhi, the protozoan parasite Toxoplasma gondii, the fungus Aspergillus flavus, and liver flukes such as Clonorchissinensis or Opisthorchis viverrini, may induce malignant transformation in hepatocytes or in target cells of the biliary tract through aberrant Wnt signaling activation. This review focuses on new insights into infectious agents implicated in the deregulation of Wnt signaling and PLC development. Since the Wnt/β-catenin pathway is a driver of cancer following viral and bacterial infections, molecules inhibiting the complex axis of Wnt signaling could represent novel therapeutic approaches in PLC treatment.
Collapse
Affiliation(s)
- Teresa Catalano
- Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Federico Selvaggi
- Unit of General Surgery, ASL2 Lanciano-Vasto-Chieti, Ospedale Clinicizzato SS Annunziata, 66100 Chieti, Italy
| | - Diana Liberata Esposito
- Center for Advanced Studies and Technology (CAST), 66100 Chieti, Italy
- Department of Innovative Technologies in Medicine & Dentistry, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Roberto Cotellese
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
- Villa Serena Foundation for Research, 65013 Città Sant'Angelo, Italy
| | - Gitana Maria Aceto
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
32
|
Jia J, Yi L, Xia Z, Yang M, Qiu D, Zhao Z, Peng Z. Development of [18F]Thiazolylacylaminopyridine-Based Glycogen Synthase Kinase-3β Ligands for Positron Emission Tomography Imaging. Bioorg Med Chem Lett 2023; 88:129263. [PMID: 37004924 DOI: 10.1016/j.bmcl.2023.129263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/16/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023]
Abstract
Glycogen synthase kinase-3β (GSK-3β) regulates numerous of CNS-specific signaling pathways, and is particularly implicated in various pathogenetic mechanisms of Alzheimer's disease (AD). A noninvasive method for detecting GSK-3β in AD brains via positron emission tomography (PET) imaging could enhance the understanding of AD pathogenesis and aid in the development of AD therapeutic drugs. In this study, an array of fluorinated thiazolyl acylaminopyridines (FTAAP) targeting GSK-3β were designed and synthesized. These compounds showed moderate to high affinities (IC50 = 6.0 - 426 nM) for GSK-3β in vitro. A potential GSK-3β tracer, [18F]8, was successfully radiolabeled. [18F]8 had unsatisfactory initial brain uptake despite its suitable lipophilicity, molecular size and good stability. Further structural refinement of the lead compound is needed to develop promising [18F]-labeled radiotracers for the detection of GSK-3β in AD brains.
Collapse
Affiliation(s)
- Jianhua Jia
- Department of Radiological Medicine, College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China.
| | - Lan Yi
- Department of Radiological Medicine, College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Zhu Xia
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
| | - Meixian Yang
- Department of Radiological Medicine, College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Dachuan Qiu
- Department of Radiological Medicine, College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Zhenghuan Zhao
- Department of Radiological Medicine, College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China
| | - Zhiping Peng
- Department of Radiological Medicine, College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, PR China.
| |
Collapse
|
33
|
Rathnayake S, Narayan B, Elber R, Wong CF. Milestoning simulation of ligand dissociation from the glycogen synthase kinase 3β. Proteins 2023; 91:209-217. [PMID: 36104870 PMCID: PMC9822852 DOI: 10.1002/prot.26423] [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: 06/04/2022] [Revised: 08/26/2022] [Accepted: 09/06/2022] [Indexed: 01/11/2023]
Abstract
As drug-binding kinetics has become an important factor to be considered in modern drug discovery, this work evaluated the ability of the Milestoning method in computing the absolute dissociation rate of a ligand from the serine-threonine kinase, glycogen synthase kinase 3β, which is a target for designing drugs to treat diseases such as neurodegenerative disorders and diabetes. We found that the Milestoning method gave good agreement with experiment with modest computational costs. Although the time scale for dissociation lasted tens of seconds, the collective molecular dynamics simulations total less than 1μs. Computing the committor function helped to identify the transition states (TSs), in which the ligand moved substantially away from the binding pocket. The glycine-rich loop with a serine residue attaching to its tips was found to undergo large movement from the bound to the TSs and might play a role in controlling drug-dissociation kinetics.
Collapse
Affiliation(s)
- Samith Rathnayake
- Department of Chemistry and Biochemistry, University of Missouri-St. Louis, St. Louis, Missouri, United States
| | - Brajesh Narayan
- School of Physics, University College Dublin, Belfield, Dublin, Ireland
| | - Ron Elber
- Department of Chemistry, Oden Institute for Computational Engineering and Science, The University of Texas at Austin, Austin, Texas, United States
| | - Chung F. Wong
- Department of Chemistry and Biochemistry, University of Missouri-St. Louis, St. Louis, Missouri, United States
| |
Collapse
|
34
|
Smart K, Zheng MQ, Holden D, Felchner Z, Zhang L, Han Y, Ropchan J, Carson RE, Vasdev N, Huang Y. In Vivo Imaging and Kinetic Modeling of Novel Glycogen Synthase Kinase-3 Radiotracers [ 11C]OCM-44 and [ 18F]OCM-50 in Non-Human Primates. Pharmaceuticals (Basel) 2023; 16:194. [PMID: 37259346 PMCID: PMC9959234 DOI: 10.3390/ph16020194] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 11/19/2023] Open
Abstract
Glycogen synthase kinase 3 (GSK-3) is a potential therapeutic target for a range of neurodegenerative and psychiatric disorders. The goal of this work was to evaluate two leading GSK-3 positron emission tomography (PET) radioligands, [11C]OCM-44 and [18F]OCM-50, in non-human primates to assess their potential for clinical translation. A total of nine PET scans were performed with the two radiotracers using arterial blood sampling in adult rhesus macaques. Brain regional time-activity curves were extracted and fitted with one- and two-tissue compartment models using metabolite-corrected arterial input functions. Target selectivity was assessed after pre-administration of the GSK-3 inhibitor PF-04802367 (PF-367, 0.03-0.25 mg/kg). Both radiotracers showed good brain uptake and distribution throughout grey matter. [11C]OCM-44 had a free fraction in the plasma of 3% at baseline and was metabolized quickly. The [11C]OCM-44 volume of distribution (VT) values in the brain increased with time; VT values from models fitted to truncated 60-min scan data were 1.4-2.9 mL/cm3 across brain regions. The plasma free fraction was 0.6% for [18F]OCM-50 and VT values (120-min) were 0.39-0.87 mL/cm3 in grey matter regions. After correcting for plasma free fraction increases during blocking scans, reductions in regional VT indicated >80% target occupancy by 0.1 mg/kg of PF-367 for both radiotracers, supporting target selectivity in vivo. [11C]OCM-44 and [18F]OCM-50 warrant further evaluation as radioligands for imaging GSK-3 in the brain, though radio-metabolite accumulation may confound image analysis.
Collapse
Affiliation(s)
- Kelly Smart
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College St., Toronto, ON M5T 1R8, Canada
- Yale PET Center, Yale School of Medicine, 801 Howard Ave., New Haven, CT 06519, USA
| | - Ming-Qiang Zheng
- Yale PET Center, Yale School of Medicine, 801 Howard Ave., New Haven, CT 06519, USA
| | - Daniel Holden
- Yale PET Center, Yale School of Medicine, 801 Howard Ave., New Haven, CT 06519, USA
| | - Zachary Felchner
- Yale PET Center, Yale School of Medicine, 801 Howard Ave., New Haven, CT 06519, USA
| | - Li Zhang
- Yale PET Center, Yale School of Medicine, 801 Howard Ave., New Haven, CT 06519, USA
| | - Yanjiang Han
- Yale PET Center, Yale School of Medicine, 801 Howard Ave., New Haven, CT 06519, USA
- Nanfang Hospital, Southern Medical University, 1838 Guangzhou Blvd North, Guangzhou 510515, China
| | - Jim Ropchan
- Yale PET Center, Yale School of Medicine, 801 Howard Ave., New Haven, CT 06519, USA
| | - Richard E. Carson
- Yale PET Center, Yale School of Medicine, 801 Howard Ave., New Haven, CT 06519, USA
| | - Neil Vasdev
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, 250 College St., Toronto, ON M5T 1R8, Canada
- Department of Psychiatry, University of Toronto, 250 College St., Toronto, ON M5T 1R8, Canada
| | - Yiyun Huang
- Yale PET Center, Yale School of Medicine, 801 Howard Ave., New Haven, CT 06519, USA
| |
Collapse
|
35
|
Yan N, Shi XL, Tang LQ, Wang DF, Li X, Liu C, Liu ZP. Synthesis and biological evaluation of thieno[3,2- c]pyrazol-3-amine derivatives as potent glycogen synthase kinase 3β inhibitors for Alzheimer's disease. J Enzyme Inhib Med Chem 2022; 37:1724-1736. [PMID: 35698879 PMCID: PMC9225722 DOI: 10.1080/14756366.2022.2086867] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Glycogen synthase kinase 3β (GSK-3β) catalyses the hyperphosphorylation of tau protein in the Alzheimer's disease (AD) pathology. A series of novel thieno[3,2-c]pyrazol-3-amine derivatives were designed and synthesised and evaluated as potential GSK-3β inhibitors by structure-guided drug rational design approach. The thieno[3,2-c]pyrazol-3-amine derivative 16b was identified as a potent GSK-3β inhibitor with an IC50 of 3.1 nM in vitro and showed accepted kinase selectivity. In cell levels, 16b showed no toxicity on the viability of SH-SY5Y cells at the concentration up to 50 μM and targeted GSK-3β with the increased phosphorylated GSK-3β at Ser9. Western blot analysis indicated that 16b decreased the phosphorylated tau at Ser396 in a dose-dependent way. Moreover, 16b effectively increased expressions of β-catenin as well as the GAP43, N-myc, and MAP-2, and promoted the differentiated neuronal neurite outgrowth. Therefore, the thieno[3,2-c]pyrazol-3-amine derivative 16b could serve as a promising GSK-3β inhibitor for the treatment of AD.
Collapse
Affiliation(s)
- Ning Yan
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Xiao-Long Shi
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Long-Qian Tang
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - De-Feng Wang
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Xun Li
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, PR China
| | - Chao Liu
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| | - Zhao-Peng Liu
- Institute of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, PR China
| |
Collapse
|
36
|
Zeng P, Liu YC, Wang XM, Ye CY, Sun YW, Su HF, Qiu SW, Li YN, Wang Y, Wang YC, Ma J, Li M, Tian Q. Targets and mechanisms of Alpinia oxyphylla Miquel fruits in treating neurodegenerative dementia. Front Aging Neurosci 2022; 14:1013891. [PMID: 36533181 PMCID: PMC9749063 DOI: 10.3389/fnagi.2022.1013891] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/10/2022] [Indexed: 11/04/2023] Open
Abstract
The dried and ripe fruits of Alpinia oxyphylla and ripe fruits of Alpinia oxyphylla Miquel (AO) have the effects of tonifying kidney-essence and nourishing intelligence and thus have been widely used in treating dementia. Alzheimer's disease (AD) is a typical form of neurodegenerative dementia with kidney-essence deficiency in Traditional Chinese Medicine (TCM). So far, there is a lack of systematic studies on the biological basis of tonifying kidney-essence and nourishing intelligence and the corresponding phytochemicals. In this study, we investigated the targets of AO in tonifying kidney-essence and nourishing intelligence based on the key pathophysiological processes of neurodegenerative dementia. According to ultra-high-performance liquid chromatography with triple quadrupole mass spectrometry data and Lipinski's rule of five, 49 bioactive phytochemicals from AO were identified, and 26 of them were found to target 168 key molecules in the treatment of neurodegenerative dementia. Nine phytochemicals of AO were shown to target acetylcholinesterase (ACHE), and 19 phytochemicals were shown to target butyrylcholinesterase (BCHE). A database of neurodegenerative dementia with kidney-essence deficiency involving 731 genes was constructed. Furthermore, yakuchinone B, 5-hydroxy-1,7-bis (4-hydroxy-3-methoxyphenyl) heptan-3-one (5-HYD), oxyhylladiketone, oxyphyllacinol, butyl-β-D-fructopyranoside, dibutyl phthalate, chrysin, yakuchinone A, rhamnetin, and rhamnocitrin were identified as the key phytochemicals from AO that regulate the pathogenesis of neurodegenerative dementia in a multitargeted manner. The approach of studying the pharmacological mechanism underlying the effects of medicinal plants and the biological basis of TCM syndrome may be helpful in studying the translation of TCM.
Collapse
Affiliation(s)
- Peng Zeng
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Histology and Embryology, School of Basic Medicine, Hengyang Medical College, University of South China, Hengyang, China
| | - Yuan-Cheng Liu
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Ming Wang
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao-Yuan Ye
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi-Wen Sun
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Fei Su
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuo-Wen Qiu
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ya-Nan Li
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Yao Wang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Yan-Chun Wang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Jun Ma
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan, China
| | - Man Li
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Tian
- Key Laboratory of Neurological Disease of National Education Ministry, School of Basic Medicine of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Hottin C, Perron M, Roger JE. GSK3 Is a Central Player in Retinal Degenerative Diseases but a Challenging Therapeutic Target. Cells 2022; 11:cells11182898. [PMID: 36139472 PMCID: PMC9496697 DOI: 10.3390/cells11182898] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022] Open
Abstract
Glycogen synthase kinase 3 (GSK3) is a key regulator of many cellular signaling processes and performs a wide range of biological functions in the nervous system. Due to its central role in numerous cellular processes involved in cell degeneration, a rising number of studies have highlighted the interest in developing therapeutics targeting GSK3 to treat neurodegenerative diseases. Although recent works strongly suggest that inhibiting GSK3 might also be a promising therapeutic approach for retinal degenerative diseases, its full potential is still under-evaluated. In this review, we summarize the literature on the role of GSK3 on the main cellular functions reported as deregulated during retinal degeneration, such as glucose homeostasis which is critical for photoreceptor survival, or oxidative stress, a major component of retinal degeneration. We also discuss the interest in targeting GSK3 for its beneficial effects on inflammation, for reducing neovascularization that occurs in some retinal dystrophies, or for cell-based therapy by enhancing Müller glia cell proliferation in diseased retina. Together, although GSK3 inhibitors hold promise as therapeutic agents, we highlight the complexity of targeting such a multitasked kinase and the need to increase our knowledge of the impact of reducing GSK3 activity on these multiple cellular pathways and biological processes.
Collapse
Affiliation(s)
- Catherine Hottin
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Muriel Perron
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Jérôme E Roger
- Paris-Saclay Institute of Neuroscience, CERTO-Retina France, CNRS, Université Paris-Saclay, 91400 Saclay, France
| |
Collapse
|
38
|
(S)-3-(3-((7-Ethynyl-9H-pyrimido[4,5-b]indol-4-yl)amino)piperidin-1-yl)propanenitrile. MOLBANK 2022. [DOI: 10.3390/m1437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The title compound (S)-3-(3-((7-ethynyl-9H-pyrimido[4,5-b]indol-4-yl)amino)piperidin-1-yl)propanenitrile (2) was synthesized in five steps, starting from 4-chloro-7-iodo-9H-pyrimido[4,5-b]indole (3), and was characterized by 1H-NMR, 13C-NMR, MS and HPLC. Moreover, its structure was confirmed by single crystal X-ray diffraction. Pyrimido[4,5-b]indole 2 demonstrated an IC50 value of 2.24 µM in a NanoBRETTM TE intracellular glycogen synthase kinase-3β assay.
Collapse
|
39
|
Hearing loss drug discovery and medicinal chemistry: Current status, challenges, and opportunities. PROGRESS IN MEDICINAL CHEMISTRY 2022; 61:1-91. [PMID: 35753714 DOI: 10.1016/bs.pmch.2022.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Hearing loss is a severe high unmet need condition affecting more than 1.5 billion people globally. There are no licensed medicines for the prevention, treatment or restoration of hearing. Prosthetic devices, such as hearing aids and cochlear implants, do not restore natural hearing and users struggle with speech in the presence of background noise. Hearing loss drug discovery is immature, and small molecule approaches include repurposing existing drugs, combination therapeutics, late-stage discovery optimisation of known chemotypes for identified molecular targets of interest, phenotypic tissue screening and high-throughput cell-based screening. Hearing loss drug discovery requires the integration of specialist therapeutic area biology and otology clinical expertise. Small molecule drug discovery projects in the global clinical portfolio for hearing loss are here collated and reviewed. An overview is provided of human hearing, inner ear anatomy, inner ear delivery, types of hearing loss and hearing measurement. Small molecule experimental drugs in clinical development for hearing loss are reviewed, including their underpinning biology, discovery strategy and activities, medicinal chemistry, calculated physicochemical properties, pharmacokinetics and clinical trial status. SwissADME BOILED-Egg permeability modelling is applied to the molecules reviewed, and these results are considered. Non-small molecule hearing loss assets in clinical development are briefly noted in this review. Future opportunities in hearing loss drug discovery for human genomics and targeted protein degradation are highlighted.
Collapse
|
40
|
Discovery of GSK3β Inhibitors through In Silico Prediction-and-Experiment Cycling Strategy, and Biological Evaluation. Molecules 2022; 27:molecules27123825. [PMID: 35744952 PMCID: PMC9230645 DOI: 10.3390/molecules27123825] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/08/2022] [Accepted: 06/11/2022] [Indexed: 11/16/2022] Open
Abstract
Direct inhibitors of glycogen synthase kinase 3β (GSK3β) have been investigated and reported for the past 20 years. In the search for novel scaffold inhibitors, 3000 compounds were selected through structure-based virtual screening (SBVS), and then high-throughput enzyme screening was performed. Among the active hit compounds, pyrazolo [1,5-a]pyrimidin-7-amine derivatives showed strong inhibitory potencies on the GSK3β enzyme and markedly activated Wnt signaling. The result of the molecular dynamics (MD) simulation, enhanced by the upper-wall restraint, was used as an advanced structural query for the SBVS. In this study, strong inhibitors designed to inhibit the GSK3β enzyme were discovered through SBVS. Our study provides structural insights into the binding mode of the inhibitors for further lead optimization.
Collapse
|
41
|
Arciniegas Ruiz S, Rippin I, Eldar-Finkelman H. Prospects in GSK-3 Signaling: From Cellular Regulation to Disease Therapy. Cells 2022; 11:cells11101618. [PMID: 35626655 PMCID: PMC9139866 DOI: 10.3390/cells11101618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 02/01/2023] Open
|
42
|
Xiong Y, Song J, Huang X, Pan Z, Goldbrunner R, Stavrinou L, Lin S, Hu W, Zheng F, Stavrinou P. Exosomes Derived From Mesenchymal Stem Cells: Novel Effects in the Treatment of Ischemic Stroke. Front Neurosci 2022; 16:899887. [PMID: 35585925 PMCID: PMC9108502 DOI: 10.3389/fnins.2022.899887] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/13/2022] [Indexed: 12/12/2022] Open
Abstract
Ischemic stroke is defined as an infarction in the brain, caused by impaired cerebral blood supply, leading to local brain tissue ischemia, hypoxic necrosis, and corresponding neurological deficits. At present, revascularization strategies in patients with acute ischemic stroke include intravenous thrombolysis and mechanical endovascular treatment. However, due to the short treatment time window (<4.5 h) and method restrictions, clinical research is focused on new methods to treat ischemic stroke. Exosomes are nano-sized biovesicles produced in the endosomal compartment of most eukaryotic cells, containing DNA, complex RNA, and protein (30-150 nm). They are released into surrounding extracellular fluid upon fusion between multivesicular bodies and the plasma membrane. Exosomes have the characteristics of low immunogenicity, good innate stability, high transmission efficiency, and the ability to cross the blood-brain barrier, making them potential therapeutic modalities for the treatment of ischemic stroke. The seed sequence of miRNA secreted by exosomes is base-paired with complementary mRNA to improve the microenvironment of ischemic tissue, thereby regulating downstream signal transduction activities. With exosome research still in the theoretical and experimental stages, this review aims to shed light on the potential of exosomes derived from mesenchymal stem cells in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yu Xiong
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Jianping Song
- Department of Neurosurgery, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
- State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
- Department of Neurosurgery, National Regional Medical Center, Fudan University Huashan Hospital Fujian Campus, The First Affiliated Hospital Binhai Campus, Fujian Medical University, Fuzhou, China
| | - Xinyue Huang
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Zhigang Pan
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Roland Goldbrunner
- Department of Neurosurgery, Faculty of Medicine and University Hospital, Center for Neurosurgery, University of Cologne, Cologne, Germany
| | - Lampis Stavrinou
- 2nd Department of Neurosurgery, Athens Medical School, “Attikon” University Hospital, National and Kapodistrian University, Athens, Greece
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Weipeng Hu
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Feng Zheng
- Department of Neurosurgery, The Second Affiliated Hospital, Fujian Medical University, Quanzhou, China
| | - Pantelis Stavrinou
- Department of Neurosurgery, Faculty of Medicine and University Hospital, Center for Neurosurgery, University of Cologne, Cologne, Germany
- Department of Neurosurgery, Metropolitan Hospital, Athens, Greece
| |
Collapse
|
43
|
Moaraf S, Rippin I, Terkel J, Eldar-Finkelman H, Barnea A. GSK-3β Inhibition in Birds Affects Social Behavior and Increases Motor Activity. Front Physiol 2022; 13:881174. [PMID: 35574473 PMCID: PMC9095836 DOI: 10.3389/fphys.2022.881174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/11/2022] [Indexed: 01/25/2023] Open
Abstract
Glycogen synthase kinase-3 (GSK-3) is a highly conserved serine/threonine protein kinase that plays a central role in a wide variety of cellular processes, cognition and behaviour. In a previous study we showed that its α and β isozymes are highly conserved in vertebrates, however the α gene is missing in birds. This selective loss offers a unique opportunity to study the role of GSK-3β independently. Accordingly, in the present study we aimed to investigate the role of GSK-3β in social behaviour, motivation, and motor activity in zebra finches (Taeniopygia guttata). We did that by selective inhibition of GSK-3β and by using tests that were specifically designed in our laboratory. Our results show that GSK-3β inhibition: 1) Affected social recognition, because the treated birds tended to move closer towards a stranger, unlike the control birds that stood closer to a familiar bird. 2) Caused the treated birds to spend more time in the more middle parts of the cage compared to controls, a behaviour that might indicate anxiety. 3) As the experiment progressed, the treated birds took less time to make a decision where to stand in the cage compared to controls, suggesting an effect on decision-making. 4) Increased in the motor activity of the treated birds compared to the controls, which can be regarded as hyperactivity. 5) Caused the treated birds to pass through a barrier in order to join their flock members faster compared to controls, and regardless of the increase in the level of difficulty, possibly suggesting increased motivation. Our study calls for further investigation, because GSK-3 is well acknowledged as a central player in regulating mood behaviour, cognitive functions, and neuronal viability. Therefore, studying its impact on normal behaviour as we did in the current study, unlike most studies that were done in diseases models, can advance our understanding regarding GSK-3 various roles and can contribute to the discovery and development of effective treatments to repair cognition and behaviour.
Collapse
Affiliation(s)
- Stan Moaraf
- School of Zoology, Tel-Aviv University, Tel-Aviv, Israel
| | - Ido Rippin
- Department of Human Molecular Genetics and Biochemistry Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Joseph Terkel
- School of Zoology, Tel-Aviv University, Tel-Aviv, Israel
| | - Hagit Eldar-Finkelman
- Department of Human Molecular Genetics and Biochemistry Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anat Barnea
- Department of Natural and Life Sciences, The Open University of Israel, Ra’anana, Israel
| |
Collapse
|
44
|
Parkinson’s Disease and SARS-CoV-2 Infection: Particularities of Molecular and Cellular Mechanisms Regarding Pathogenesis and Treatment. Biomedicines 2022; 10:biomedicines10051000. [PMID: 35625737 PMCID: PMC9138688 DOI: 10.3390/biomedicines10051000] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 02/01/2023] Open
Abstract
Accumulating data suggest that chronic neuroinflammation-mediated neurodegeneration is a significant contributing factor for progressive neuronal and glial cell death in age-related neurodegenerative pathology. Furthermore, it could be encountered as long-term consequences in some viral infections, including post-COVID-19 Parkinsonism-related chronic sequelae. The current systematic review is focused on a recent question aroused during the pandemic’s successive waves: are there post-SARS-CoV-2 immune-mediated reactions responsible for promoting neurodegeneration? Does the host’s dysregulated immune counter-offensive contribute to the pathogenesis of neurodegenerative diseases, emerging as Parkinson’s disease, in a complex interrelation between genetic and epigenetic risk factors? A synthetic and systematic literature review was accomplished based on the ”Preferred Reporting Items for Systematic Principles Reviews and Meta-Analyses” (PRISMA) methodology, including registration on the specific online platform: International prospective register of systematic reviews—PROSPERO, no. 312183. Initially, 1894 articles were detected. After fulfilling the five steps of the selection methodology, 104 papers were selected for this synthetic review. Documentation was enhanced with a supplementary 47 bibliographic resources identified in the literature within a non-standardized search connected to the subject. As a final step of the PRISMA method, we have fulfilled a Population-Intervention-Comparison-Outcome-Time (PICOT)/Population-Intervention-Comparison-Outcome-Study type (PICOS)—based metanalysis of clinical trials identified as connected to our search, targeting the outcomes of rehabilitative kinesitherapeutic interventions compared to clinical approaches lacking such kind of treatment. Accordingly, we identified 10 clinical trials related to our article. The multi/interdisciplinary conventional therapy of Parkinson’s disease and non-conventional multitarget approach to an integrative treatment was briefly analyzed. This article synthesizes the current findings on the pathogenic interference between the dysregulated complex mechanisms involved in aging, neuroinflammation, and neurodegeneration, focusing on Parkinson’s disease and the acute and chronic repercussions of COVID-19. Time will tell whether COVID-19 neuroinflammatory events could trigger long-term neurodegenerative effects and contribute to the worsening and/or explosion of new cases of PD. The extent of the interrelated neuropathogenic phenomenon remains obscure, so further clinical observations and prospective longitudinal cohort studies are needed.
Collapse
|