1
|
Ansari MM, Sahu SK, Singh TG, Singh SRJ, Kaur P. Evolving significance of kinase inhibitors in the management of Alzheimer's disease. Eur J Pharmacol 2024; 979:176816. [PMID: 39038637 DOI: 10.1016/j.ejphar.2024.176816] [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: 03/23/2024] [Revised: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
Alzheimer's disease is a neurodegenerative problem with progressive loss of memory and other cognitive function disorders resulting in the imbalance of neurotransmitter activity and signaling progression, which poses the need of the potential therapeutic target to improve the intracellular signaling cascade brought by kinases. Protein kinase plays a significant and multifaceted role in the treatment of Alzheimer's disease, by targeting pathological mechanisms like tau hyperphosphorylation, neuroinflammation, amyloid-beta production and synaptic dysfunction. In this review, we thoroughly explore the essential protein kinases involved in Alzheimer's disease, detailing their physiological roles, regulatory impacts, and the newest inhibitors and compounds that are progressing into clinical trials. All the findings of studies exhibited the promising role of kinase inhibitors in the management of Alzheimer's disease. However, it still poses the need of addressing current challenges and opportunities involved with this disorder for the future perspective of kinase inhibitors in the management of Alzheimer's disease. Further study includes the development of biomarkers, combination therapy, and next-generation kinase inhibitors with increased potency and selectivity for its future prospects.
Collapse
Affiliation(s)
- Md Mustafiz Ansari
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | - Sanjeev Kumar Sahu
- School of Pharmaceutical Sciences, Lovely Professional University, Punjab, India
| | | | - Sovia R J Singh
- University Language Centre- Chitkara Business School, Chitkara University, Punjab, India
| | - Paranjeet Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
2
|
Birgbauer E. Lysophospholipid receptors in neurodegeneration and neuroprotection. EXPLORATION OF NEUROPROTECTIVE THERAPY 2024; 4:349-365. [PMID: 39247084 PMCID: PMC11379401 DOI: 10.37349/ent.2024.00088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 08/11/2024] [Indexed: 09/10/2024]
Abstract
The central nervous system (CNS) is one of the most complex physiological systems, and treatment of CNS disorders represents an area of major medical need. One critical aspect of the CNS is its lack of regeneration, such that damage is often permanent. The damage often leads to neurodegeneration, and so strategies for neuroprotection could lead to major medical advances. The G protein-coupled receptor (GPCR) family is one of the major receptor classes, and they have been successfully targeted clinically. One class of GPCRs is those activated by bioactive lysophospholipids as ligands, especially sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA). Research has been increasingly demonstrating the important roles that S1P and LPA, and their receptors, play in physiology and disease. In this review, I describe the role of S1P and LPA receptors in neurodegeneration and potential roles in neuroprotection. Much of our understanding of the role of S1P receptors has been through pharmacological tools. One such tool, fingolimod (also known as FTY720), which is a S1P receptor agonist but a functional antagonist in the immune system, is clinically efficacious in multiple sclerosis by producing a lymphopenia to reduce autoimmune attacks; however, there is evidence that fingolimod is also neuroprotective. Furthermore, fingolimod is neuroprotective in many other neuropathologies, including stroke, Parkinson's disease, Huntington's disease, Rett syndrome, Alzheimer's disease, and others that are discussed here. LPA receptors also appear to be involved, being upregulated in a variety of neuropathologies. Antagonists or mutations of LPA receptors, especially LPA1, are neuroprotective in a variety of conditions, including cortical development, traumatic brain injury, spinal cord injury, stroke and others discussed here. Finally, LPA receptors may interact with other receptors, including a functional interaction with plasticity related genes.
Collapse
Affiliation(s)
- Eric Birgbauer
- Department of Biology, Winthrop University, Rock Hill, SC 29733, USA
| |
Collapse
|
3
|
Xia L, Chen J, Huang J, Lin X, Jiang J, Liu T, Huang N, Luo Y. The role of AMPKα subunit in Alzheimer's disease: In-depth analysis and future prospects. Heliyon 2024; 10:e34254. [PMID: 39071620 PMCID: PMC11279802 DOI: 10.1016/j.heliyon.2024.e34254] [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: 04/01/2024] [Revised: 06/29/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024] Open
Abstract
The AMP-activated protein kinase α (AMPKα) subunit is the catalytic subunit in the AMPK complex, playing a crucial role in AMPK activation. It has two isoforms: AMPKα1 and AMPKα2. Emerging evidence suggests that the AMPKα subunit exhibits subtype-specific effects in Alzheimer's disease (AD). This review discusses the role of the AMPKα subunit in the pathogenesis of AD, including its impact on β-amyloid (Aβ) pathology, Tau pathology, metabolic disorders, inflammation, mitochondrial dysfunction, inflammasome and pyroptosis. Additionally, it reviews the distinct roles of its isoforms, AMPKα1 and AMPKα2, in AD, which may provide more precise targets for future drug development in AD.
Collapse
Affiliation(s)
- Lingqiong Xia
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Jianhua Chen
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Juan Huang
- Key Laboratory of Basic Pharmacology and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Guizhou, China
| | - Xianmei Lin
- Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Jingyu Jiang
- Department of Gastroenterology, Guizhou Aerospace Hospital, Zunyi, Guizhou, China
| | - Tingting Liu
- National Drug Clinical Trial Institution, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Nanqu Huang
- National Drug Clinical Trial Institution, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| | - Yong Luo
- Department of Neurology, Third Affiliated Hospital of Zunyi Medical University (The First People's Hospital of Zunyi), Zunyi, Guizhou, China
| |
Collapse
|
4
|
Sanna K, Bruno A, Balletta S, Caioli S, Nencini M, Fresegna D, Guadalupi L, Dolcetti E, Azzolini F, Buttari F, Fantozzi R, Borrelli A, Stampanoni Bassi M, Gilio L, Lauritano G, Vanni V, De Vito F, Tartacca A, Mariani F, Rovella V, Musella A, Centonze D, Mandolesi G. Re-emergence of T lymphocyte-mediated synaptopathy in progressive multiple sclerosis. Front Immunol 2024; 15:1416133. [PMID: 38911847 PMCID: PMC11190089 DOI: 10.3389/fimmu.2024.1416133] [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: 04/11/2024] [Accepted: 05/24/2024] [Indexed: 06/25/2024] Open
Abstract
Background Secondary progressive multiple sclerosis (SPMS) is defined by the irreversible accumulation of disability following a relapsing-remitting MS (RRMS) course. Despite treatments advances, a reliable tool able to capture the transition from RRMS to SPMS is lacking. A T cell chimeric MS model demonstrated that T cells derived from relapsing patients exacerbate excitatory transmission of central neurons, a synaptotoxic event absent during remitting stages. We hypothesized the re-emergence of T cell synaptotoxicity during SPMS and investigated the synaptoprotective effects of siponimod, a sphingosine 1-phosphate receptor (S1PR) modulator, known to reduce grey matter damage in SPMS patients. Methods Data from healthy controls (HC), SPMS patients, and siponimod-treated SPMS patients were collected. Chimeric experiments were performed incubating human T cells on murine cortico-striatal slices, and recording spontaneous glutamatergic activity from striatal neurons. Homologous chimeric experiments were executed incubating EAE mice T cells with siponimod and specific S1PR agonists or antagonists to identify the receptor involved in siponimod-mediated synaptic recovery. Results SPMS patient-derived T cells significantly increased the striatal excitatory synaptic transmission (n=40 synapses) compared to HC T cells (n=55 synapses), mimicking the glutamatergic alterations observed in active RRMS-T cells. Siponimod treatment rescued SPMS T cells synaptotoxicity (n=51 synapses). Homologous chimeric experiments highlighted S1P5R involvement in the siponimod's protective effects. Conclusion Transition from RRMS to SPMS involves the reappearance of T cell-mediated synaptotoxicity. Siponimod counteracts T cell-induced excitotoxicity, emphasizing the significance of inflammatory synaptopathy in progressive MS and its potential as a promising pharmacological target.
Collapse
Affiliation(s)
- Krizia Sanna
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Antonio Bruno
- Ph.D. Program in Neuroscience, Department of Systems Medicine, Tor Vergata University, Rome, Italy
- Unit of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, IS, Italy
| | - Sara Balletta
- Unit of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, IS, Italy
| | - Silvia Caioli
- Unit of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, IS, Italy
| | - Monica Nencini
- Synaptic Immunopathology Lab, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Roma, Rome, Italy
| | - Diego Fresegna
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Synaptic Immunopathology Lab, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Roma, Rome, Italy
| | - Livia Guadalupi
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Synaptic Immunopathology Lab, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Roma, Rome, Italy
| | - Ettore Dolcetti
- Ph.D. Program in Neuroscience, Department of Systems Medicine, Tor Vergata University, Rome, Italy
- Unit of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, IS, Italy
| | - Federica Azzolini
- Unit of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, IS, Italy
| | - Fabio Buttari
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Unit of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, IS, Italy
| | - Roberta Fantozzi
- Unit of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, IS, Italy
| | - Angela Borrelli
- Unit of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, IS, Italy
| | - Mario Stampanoni Bassi
- Unit of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, IS, Italy
| | - Luana Gilio
- Unit of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, IS, Italy
| | - Gianluca Lauritano
- Ph.D. Program in Neuroscience, Department of Systems Medicine, Tor Vergata University, Rome, Italy
- Unit of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, IS, Italy
| | - Valentina Vanni
- Synaptic Immunopathology Lab, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Roma, Rome, Italy
| | - Francesca De Vito
- Unit of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, IS, Italy
| | - Alice Tartacca
- Ph.D. Program in Neuroscience, Department of Systems Medicine, Tor Vergata University, Rome, Italy
| | - Fabrizio Mariani
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Valentina Rovella
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Alessandra Musella
- Synaptic Immunopathology Lab, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, Italy
| | - Diego Centonze
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Unit of Neurology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, IS, Italy
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, Rome, Italy
| |
Collapse
|
5
|
Wang W, Zhao Y, Zhu G. The role of sphingosine-1-phosphate in the development and progression of Parkinson's disease. Front Cell Neurosci 2023; 17:1288437. [PMID: 38179204 PMCID: PMC10764561 DOI: 10.3389/fncel.2023.1288437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/27/2023] [Indexed: 01/06/2024] Open
Abstract
Parkinson's disease (PD) could be viewed as a proteinopathy caused by changes in lipids, whereby modifications in lipid metabolism may lead to protein alterations, such as the accumulation of alpha-synuclein (α-syn), ultimately resulting in neurodegeneration. Although the loss of dopaminergic neurons in the substantia nigra is the major clinical manifestation of PD, the etiology of it is largely unknown. Increasing evidence has highlighted the important role of lipids in the pathophysiology of PD. Sphingosine-1-phosphate (S1P), a signaling lipid, has been suggested to have a potential association with the advancement and worsening of PD. Therefore, better understanding the mechanisms and regulatory proteins is of high interest. Most interestingly, S1P appears to be an important target to offers a new strategy for the diagnosis and treatment of PD. In this review, we first introduce the basic situation of S1P structure, function and regulation, with a special focus on the several pathways. We then briefly describe the regulation of S1P signaling pathway on cells and make a special focused on the cell growth, proliferation and apoptosis, etc. Finally, we discuss the function of S1P as potential therapeutic target to improve the clinical symptoms of PD, and even prevent the progression of the PD. In the context of PD, the functions of S1P modulators have been extensively elucidated. In conclusion, S1P modulators represent a novel and promising therapeutic principle and therapeutic method for PD. However, more research is required before these drugs can be considered as a standard treatment option for PD.
Collapse
Affiliation(s)
- Wang Wang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhao
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guoxue Zhu
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
6
|
Oizumi H, Sugimura Y, Totsune T, Kawasaki I, Ohshiro S, Baba T, Kimpara T, Sakuma H, Hasegawa T, Kawahata I, Fukunaga K, Takeda A. Plasma sphingolipid abnormalities in neurodegenerative diseases. PLoS One 2022; 17:e0279315. [PMID: 36525454 PMCID: PMC9757566 DOI: 10.1371/journal.pone.0279315] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In recent years, there has been increasing evidence that several lipid metabolism abnormalities play an important role in the pathogenesis of neurodegenerative diseases. However, it is still unclear which lipid metabolism abnormalities play the most important role in neurodegenerative diseases. Plasma lipid metabolomics (lipidomics) has been shown to be an unbiased method that can be used to explore lipid metabolism abnormalities in neurodegenerative diseases. Plasma lipidomics in neurodegenerative diseases has been performed only in idiopathic Parkinson's disease (IPD) and Alzheimer's disease (AD), and comprehensive studies are needed to clarify the pathogenesis. METHODS In this study, we investigated plasma lipids using lipidomics in individuals with neurodegenerative diseases and healthy controls (CNs). Plasma lipidomics was evaluated by liquid chromatography-tandem mass spectrometry (LC-MS/MS) in those with IPD, dementia with Lewy bodies (DLB), multiple system atrophy (MSA), AD, and progressive supranuclear palsy (PSP) and CNs. RESULTS The results showed that (1) plasma sphingosine-1-phosphate (S1P) was significantly lower in all neurodegenerative disease groups (IPD, DLB, MSA, AD, and PSP) than in the CN group. (2) Plasma monohexylceramide (MonCer) and lactosylceramide (LacCer) were significantly higher in all neurodegenerative disease groups (IPD, DLB, MSA, AD, and PSP) than in the CN group. (3) Plasma MonCer levels were significantly positively correlated with plasma LacCer levels in all enrolled groups. CONCLUSION S1P, Glucosylceramide (GlcCer), the main component of MonCer, and LacCer are sphingolipids that are biosynthesized from ceramide. Recent studies have suggested that elevated GlcCer and decreased S1P levels in neurons are related to neuronal cell death and that elevated LacCer levels induce neurodegeneration by neuroinflammation. In the present study, we found decreased plasma S1P levels and elevated plasma MonCer and LacCer levels in those with neurodegenerative diseases, which is a new finding indicating the importance of abnormal sphingolipid metabolism in neurodegeneration.
Collapse
Affiliation(s)
- Hideki Oizumi
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Yoko Sugimura
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Tomoko Totsune
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Iori Kawasaki
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Saki Ohshiro
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Toru Baba
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Teiko Kimpara
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Hiroaki Sakuma
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
| | - Takafumi Hasegawa
- Department of Neurology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ichiro Kawahata
- Department of Pharmacology, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Tohoku University Graduate School of Pharmaceutical Sciences, Sendai, Japan
| | - Atsushi Takeda
- Department of Neurology, National Hospital Organization Sendai Nishitaga Hospital, Sendai, Japan
- Department of Cognitive and Motor Aging, Tohoku University Graduate School of Medicine, Sendai, Japan
- * E-mail:
| |
Collapse
|
7
|
The Strategies for Treating "Alzheimer's Disease": Insulin Signaling May Be a Feasible Target. Curr Issues Mol Biol 2022; 44:6172-6188. [PMID: 36547082 PMCID: PMC9777526 DOI: 10.3390/cimb44120421] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/11/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by senile plaques formed by amyloid-beta (Aβ) extracellularly and neurofibrillary tangles (NFTs) formed by hyperphosphorylated tau protein intracellularly. Apart from these two features, insulin deficiency and insulin resistance have also been observed in AD brains. Thus, AD has also been referred to as type 3 diabetes by some of the scientists in this field. Insulin plays a pivotal role in learning and memory and is involved in regulating tau phosphorylation though the PI3KAkt-GSK3b signaling pathway. Interestingly, recent studies revealed that in AD brains the microglia transformed into a disease-associated microglia (DAM) status in a TREM2-dependent manner to restrain the toxicity of Aβ and propagation of tau. This also correlated with PI3K-Akt signaling through the adaptor of TREM2. Whether insulin has any effect on microglia activation in AD pathology is unclear so far. However, many studies demonstrated that diabetes increased the risk of AD. In this review, we summarize the main strategies for curing AD, including lowering the level of Aβ, suppressing the phosphorylation of tau, the ablation and/or repopulation of microglia, and especially the supply of insulin. We also propose that attention should be given to the influences of insulin on microglia in AD.
Collapse
|
8
|
Gutner UA, Shupik MA. The Role of Sphingosine-1-Phosphate in Neurodegenerative Diseases. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2021. [DOI: 10.1134/s1068162021050277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
9
|
Paradiso E, Lazzaretti C, Sperduti S, Antoniani F, Fornari G, Brigante G, Di Rocco G, Tagliavini S, Trenti T, Morini D, Falbo AI, Villani MT, Nofer JR, Simoni M, Potì F, Casarini L. Sphingosine-1 phosphate induces cAMP/PKA-independent phosphorylation of the cAMP response element-binding protein (CREB) in granulosa cells. Mol Cell Endocrinol 2021; 520:111082. [PMID: 33189864 DOI: 10.1016/j.mce.2020.111082] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/20/2020] [Accepted: 11/09/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIMS Sphingosine-1 phosphate (S1P) is a lysosphingolipid present in the ovarian follicular fluid. The role of the lysosphingolipid in gonads of the female is widely unclear. At nanomolar concentrations, S1P binds and activates five specific G protein-coupled receptors (GPCRs), known as S1P1-5, modulating different signaling pathways. S1P1 and S1P3 are highly expressed in human primary granulosa lutein cells (hGLC), as well as in the immortalized human primary granulosa cell line hGL5. In this study, we evaluated the signaling cascade activated by S1P and its synthetic analogues in hGLC and hGL5 cells, exploring the biological relevance of S1PR-stimulation in this context. METHODS AND RESULTS hGLC and hGL5 cells were treated with a fixed dose (0.1 μM) of S1P, or by S1P1- and S1P3-specific agonists SEW2871 and CYM5541. In granulosa cells, S1P and, at a lesser extent, SEW2871 and CYM5541, potently induced CREB phosphorylation. No cAMP production was detected and pCREB activation occurred even in the presence of the PKA inhibitor H-89. Moreover, S1P-dependent CREB phosphorylation was dampened by the mitogen-activate protein kinase (MEK) inhibitor U0126 and by the L-type Ca2+ channel blocker verapamil. The complete inhibition of CREB phosphorylation occurred by blocking either S1P2 or S1P3 with the specific receptor antagonists JTE-013 and TY52156, or under PLC/PI3K depletion. S1P-dependent CREB phosphorylation induced FOXO1 and the EGF-like epiregulin-encoding gene (EREG), confirming the exclusive role of gonadotropins and interleukins in this process, but did not affect steroidogenesis. However, S1P or agonists did not modulate granulosa cell viability and proliferation in our conditions. CONCLUSIONS This study demonstrates for the first time that S1P may induce a cAMP-independent activation of pCREB in granulosa cells, although this is not sufficient to induce intracellular steroidogenic signals and progesterone synthesis. S1P-induced FOXO1 and EREG gene expression suggests that the activation of S1P-S1PR axis may cooperate with gonadotropins in modulating follicle development.
Collapse
Affiliation(s)
- Elia Paradiso
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Modena, Italy
| | - Clara Lazzaretti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; International PhD School in Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Modena, Italy
| | - Samantha Sperduti
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesco Antoniani
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Fornari
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Brigante
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Department of Medical Specialties, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Giulia Di Rocco
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Simonetta Tagliavini
- Department of Laboratory Medicine and Pathological Anatomy, Azienda Ospedaliero-Universitaria di Modena, NOCSAE, Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathological Anatomy, Azienda Ospedaliero-Universitaria di Modena, NOCSAE, Modena, Italy
| | - Daria Morini
- Department of Obstetrics and Gynaecology, Fertility Center, ASMN. Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Modena, Italy
| | - Angela Immacolata Falbo
- Department of Obstetrics and Gynaecology, Fertility Center, ASMN. Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Modena, Italy
| | - Maria Teresa Villani
- Department of Obstetrics and Gynaecology, Fertility Center, ASMN. Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Modena, Italy
| | - Jerzy-Roch Nofer
- Central Laboratory Facility, University Hospital Münster, Münster, Germany
| | - Manuela Simoni
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Center for Genomic Research, University of Modena and Reggio Emilia, Modena, Italy; Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy; PR China, INRA, CNRS, IFCE, Université de Tours, 37380, Nouzilly, France
| | - Francesco Potì
- Unit of Neurosciences, Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | - Livio Casarini
- Unit of Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy; Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
10
|
Kang YJ, Diep YN, Tran M, Cho H. Therapeutic Targeting Strategies for Early- to Late-Staged Alzheimer's Disease. Int J Mol Sci 2020; 21:E9591. [PMID: 33339351 PMCID: PMC7766709 DOI: 10.3390/ijms21249591] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/30/2020] [Accepted: 12/08/2020] [Indexed: 02/08/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia, typically showing progressive neurodegeneration in aging brains. The key signatures of the AD progression are the deposition of amyloid-beta (Aβ) peptides, the formation of tau tangles, and the induction of detrimental neuroinflammation leading to neuronal loss. However, conventional pharmacotherapeutic options are merely relying on the alleviation of symptoms that are limited to mild to moderate AD patients. Moreover, some of these medicines discontinued to use due to either the insignificant effectiveness in improving the cognitive impairment or the adverse side effects worsening essential bodily functions. One of the reasons for the failure is the lack of knowledge on the underlying mechanisms that can accurately explain the major causes of the AD progression correlating to the severity of AD. Therefore, there is an urgent need for the better understanding of AD pathogenesis and the development of the disease-modifying treatments, particularly for severe and late-onset AD, which have not been covered thoroughly. Here, we review the underlying mechanisms of AD progression, which have been employed for the currently established therapeutic strategies. We believe this will further spur the discovery of a novel disease-modifying treatment for mild to severe, as well as early- to late-onset, AD.
Collapse
Affiliation(s)
- You Jung Kang
- Department of Mechanical Engineering and Engineering Science, Center for Biomedical Engineering and Science, University of North Carolina, Charlotte, NC 28223, USA;
- Department of Biological Sciences, Center for Biomedical Engineering and Science, University of North Carolina, Charlotte, NC 28223, USA
| | - Yen N. Diep
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Korea; (Y.N.D.); (M.T.)
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Korea
| | - Minh Tran
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Korea; (Y.N.D.); (M.T.)
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Korea
| | - Hansang Cho
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Korea; (Y.N.D.); (M.T.)
- Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Korea
| |
Collapse
|
11
|
Wang L, Bharti, Kumar R, Pavlov PF, Winblad B. Small molecule therapeutics for tauopathy in Alzheimer's disease: Walking on the path of most resistance. Eur J Med Chem 2020; 209:112915. [PMID: 33139110 DOI: 10.1016/j.ejmech.2020.112915] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 12/19/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia characterized by presence of extracellular amyloid plaques and intracellular neurofibrillary tangles composed of tau protein. Currently there are close to 50 million people living with dementia and this figure is expected to increase to 75 million by 2030 putting a huge burden on the economy due to the health care cost. Considering the effects on quality of life of patients and the increasing burden on the economy, there is an enormous need of new disease modifying therapies to tackle this disease. The current therapies are dominated by only symptomatic treatments including cholinesterase inhibitors and N-methyl-D-aspartate receptor blockers but no disease modifying treatments exist so far. After several failed attempts to develop drugs against amyloidopathy, tau targeting approaches have been in the main focus of drug development against AD. After an overview of the tauopathy in AD, this review summarizes recent findings on the development of small molecules as therapeutics targeting tau modification, aggregation, and degradation, and tau-oriented multi-target directed ligands. Overall, this work aims to provide a comprehensive and critical overview of small molecules which are being explored as a lead candidate for discovering drugs against tauopathy in AD.
Collapse
Affiliation(s)
- Lisha Wang
- Dept. of Neuroscience Care and Society, Div. of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden
| | - Bharti
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Rajnish Kumar
- Dept. of Neuroscience Care and Society, Div. of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden; Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, 221005, India
| | - Pavel F Pavlov
- Dept. of Neuroscience Care and Society, Div. of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden; Memory Clinic, Theme Aging, Karolinska University Hospital, 14186, Huddinge, Sweden
| | - Bengt Winblad
- Dept. of Neuroscience Care and Society, Div. of Neurogeriatrics, Karolinska Institutet, 17164, Solna, Sweden; Memory Clinic, Theme Aging, Karolinska University Hospital, 14186, Huddinge, Sweden.
| |
Collapse
|
12
|
Wei H, Zhang HL, Xie JZ, Meng DL, Wang XC, Ke D, Zeng J, Liu R. Protein Phosphatase 2A as a Drug Target in the Treatment of Cancer and Alzheimer's Disease. Curr Med Sci 2020; 40:1-8. [PMID: 32166659 DOI: 10.1007/s11596-020-2140-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 12/10/2019] [Indexed: 01/22/2023]
Abstract
Protein phosphatase 2A (PP2A) is a major serine/threonine phosphatase which participates in the regulation of multiple cellular processes. As a confirmed tumor suppressor, PP2A activity is downregulated in tumors and its re-activation can induce apoptosis of cancer cells. In the brains of Alzheimer's disease (AD) patients, decreased PP2A activity also plays a key role in promoting tau hyperphosphorylation and Aβ generation. In this review, we discussed compounds aiming at modulating PP2A activity in the treatment of cancer or AD. The upstream factors that inactivate PP2A in diseases have not been fully elucidated and further studies are needed. It will help for the refinement and development of novel and clinically tractable PP2A-targeted compounds or therapies for the treatment of tumor and AD.
Collapse
Affiliation(s)
- Hui Wei
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hui-Liang Zhang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jia-Zhao Xie
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dong-Li Meng
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Chuan Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dan Ke
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Ji Zeng
- Department of Clinic Laboratory, Wuhan Fourth Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Rong Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
13
|
Pépin É, Jalinier T, Lemieux GL, Massicotte G, Cyr M. Sphingosine-1-Phosphate Receptors Modulators Decrease Signs of Neuroinflammation and Prevent Parkinson's Disease Symptoms in the 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine Mouse Model. Front Pharmacol 2020; 11:77. [PMID: 32153401 PMCID: PMC7047735 DOI: 10.3389/fphar.2020.00077] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 01/27/2020] [Indexed: 12/20/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a potent bioactive lipid mediator that acts as a natural ligand upon binding to five different receptors that are located in astrocytes, oligodendrocytes, microglial and neuronal cells. Recently, global activation of these receptors by FTY720 (fingolimod) has been suggested to provide neuroprotection in animal model of Parkinson’s disease (PD). Among S1P receptors, the subtype 1 (S1P1R) has been linked to features of neuroprotection and, using the selective agonist SEW2871, the present investigation assessed potential benefits (and mechanisms) of this receptor subtype in an established animal model of PD. We demonstrated that oral treatments with SEW2871 are able to provide protection to the same levels as FTY720 against loss of dopaminergic neurons and motor deficits in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) (30 mg/kg, i.p., 5 days) mouse model of PD. At the molecular level, we observed that the beneficial effects of both S1PR agonists were not associated with alterations in ERK and Akt levels, two markers of molecular adaptations in the striatum neurons. However, these compounds have the capacity to prevent signs of neuroinflammation such as the activation of astrocytes and glial cells, as well as MPTP-induced reduction of BDNF levels in key regions of the brain implicated in motor functions. These findings suggest that selective S1P1R modulation has the ability to provide neuroprotection in response to MPTP neurotoxicity. Targeting S1P1R in PD therapy may represent a prominent candidate for treatment of this neurodegenerative conditions.
Collapse
Affiliation(s)
- Élise Pépin
- Groupe de recherche en signalisation cellulaire, Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Tim Jalinier
- Groupe de recherche en signalisation cellulaire, Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Guillaume L Lemieux
- Groupe de recherche en signalisation cellulaire, Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Guy Massicotte
- Groupe de recherche en signalisation cellulaire, Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| | - Michel Cyr
- Groupe de recherche en signalisation cellulaire, Département de biologie médicale, Université du Québec à Trois-Rivières, Trois-Rivières, QC, Canada
| |
Collapse
|
14
|
The Bewildering Effect of AMPK Activators in Alzheimer's Disease: Review of the Current Evidence. BIOMED RESEARCH INTERNATIONAL 2020; 2020:9895121. [PMID: 32149150 PMCID: PMC7049408 DOI: 10.1155/2020/9895121] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 01/14/2020] [Accepted: 01/29/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease is a multifactorial neurodegenerative disease characterized by progressive cognitive dysfunction. It is the most common form of dementia. The pathologic hallmarks of the disease include extracellular amyloid plaque, intracellular neurofibrillary tangles, and oxidative stress, to mention some of them. Despite remarkable progress in the understanding of the pathogenesis of the disease, drugs for cure or disease-modifying therapy remain somewhere in the distance. From recent time, the signaling molecule AMPK is gaining enormous attention in the AD drug research. AMPK is a master regulator of cellular energy metabolism, and recent pieces of evidence show that perturbation of its function is highly ascribed in the pathology of AD. Several drugs are known to activate AMPK, but their effect in AD remains to be controversial. In this review, the current shreds of evidence on the effect of AMPK activators in Aβ accumulation, tau aggregation, and oxidative stress are addressed. Positive and negative effects are reported with regard to Aβ and tauopathy but only positive in oxidative stress. We also tried to dissect the molecular interplays where the bewildering effects arise from.
Collapse
|
15
|
Crivelli SM, Giovagnoni C, Visseren L, Scheithauer AL, de Wit N, den Hoedt S, Losen M, Mulder MT, Walter J, de Vries HE, Bieberich E, Martinez-Martinez P. Sphingolipids in Alzheimer's disease, how can we target them? Adv Drug Deliv Rev 2020; 159:214-231. [PMID: 31911096 DOI: 10.1016/j.addr.2019.12.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/09/2019] [Accepted: 12/31/2019] [Indexed: 01/06/2023]
Abstract
Altered levels of sphingolipids and their metabolites in the brain, and the related downstream effects on neuronal homeostasis and the immune system, provide a framework for understanding mechanisms in neurodegenerative disorders and for developing new intervention strategies. In this review we will discuss: the metabolites of sphingolipids that function as second messengers; and functional aberrations of the pathway resulting in Alzheimer's disease (AD) pathophysiology. Focusing on the central product of the sphingolipid pathway ceramide, we describ approaches to pharmacologically decrease ceramide levels in the brain and we argue on how the sphingolipid pathway may represent a new framework for developing novel intervention strategies in AD. We also highlight the possible use of clinical and non-clinical drugs to modulate the sphingolipid pathway and sphingolipid-related biological cascades.
Collapse
|
16
|
Bazzari FH, Abdallah DM, El-Abhar HS. Pharmacological Interventions to Attenuate Alzheimer’s Disease Progression: The Story So Far. Curr Alzheimer Res 2019; 16:261-277. [DOI: 10.2174/1567205016666190301111120] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 12/15/2018] [Accepted: 01/31/2019] [Indexed: 12/23/2022]
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease and the most common cause of dementia in the elderly. Up to date, the available pharmacological options for AD are limited to cholinesterase inhibitors and memantine that may only provide modest symptomatic management with no significance in slowing down the disease progression. Over the past three decades, the increased interest in and the understanding of AD major pathological hallmarks have provided an insight into the mechanisms mediating its pathogenesis, which in turn introduced a number of hypotheses and novel targets for the treatment of AD. Initially, targeting amyloid-beta and tau protein was considered the most promising therapeutic approach. However, further investigations have identified other major players, such as neuroinflammation, impaired insulin signalling and defective autophagy, that may contribute to the disease progression. While some promising drugs are currently being investigated in human studies, the majority of the previously developed medical agents have come to an end in clinical trials, as they have failed to illustrate any beneficial outcome. This review aims to discuss the different introduced approaches to alleviate AD progression; in addition, provides a comprehensive overview of the drugs in the development phase as well as their mode of action and an update of their status in clinical trials.
Collapse
Affiliation(s)
- Firas H. Bazzari
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Dalaal M. Abdallah
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hanan S. El-Abhar
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
17
|
Silva VRR, Micheletti TO, Katashima CK, Lenhare L, Morari J, Moura‐Assis A, Lima‐Júnior JC, Camargo JA, Passos GR, Gaspar RS, Velloso LA, Saad MJ, da Silva ASR, Moura LP, Cintra DE, Pauli JR, Ropelle ER. Exercise activates the hypothalamic S1PR1–STAT3 axis through the central action of interleukin 6 in mice. J Cell Physiol 2018; 233:9426-9436. [DOI: 10.1002/jcp.26818] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/30/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Vagner R. R. Silva
- Laboratory of Molecular Biology of Exercise (LaBMEx) School of Applied Sciences, University of Campinas Limeira São Paulo Brazil
- Department of Internal Medicine Faculty of Medical Sciences, University of Campinas (UNICAMP) Campinas São Paulo Brazil
| | - Thayana O. Micheletti
- Department of Internal Medicine Faculty of Medical Sciences, University of Campinas (UNICAMP) Campinas São Paulo Brazil
| | - Carlos K. Katashima
- Department of Internal Medicine Faculty of Medical Sciences, University of Campinas (UNICAMP) Campinas São Paulo Brazil
| | - Luciene Lenhare
- Laboratory of Molecular Biology of Exercise (LaBMEx) School of Applied Sciences, University of Campinas Limeira São Paulo Brazil
- Department of Internal Medicine Faculty of Medical Sciences, University of Campinas (UNICAMP) Campinas São Paulo Brazil
| | - Joseane Morari
- Laboratory of Cell Signaling Obesity and Comorbidities Research Center (OCRC), University of Campinas Campinas São Paulo Brazil
| | - Alexandre Moura‐Assis
- Laboratory of Cell Signaling Obesity and Comorbidities Research Center (OCRC), University of Campinas Campinas São Paulo Brazil
| | - José C. Lima‐Júnior
- Laboratory of Cell Signaling Obesity and Comorbidities Research Center (OCRC), University of Campinas Campinas São Paulo Brazil
| | - Juliana A. Camargo
- Department of Internal Medicine Faculty of Medical Sciences, University of Campinas (UNICAMP) Campinas São Paulo Brazil
| | - Gabriela R. Passos
- Department of Internal Medicine Faculty of Medical Sciences, University of Campinas (UNICAMP) Campinas São Paulo Brazil
| | - Rodrigo S. Gaspar
- Laboratory of Molecular Biology of Exercise (LaBMEx) School of Applied Sciences, University of Campinas Limeira São Paulo Brazil
| | - Licio A. Velloso
- Laboratory of Cell Signaling Obesity and Comorbidities Research Center (OCRC), University of Campinas Campinas São Paulo Brazil
| | - Mario J. Saad
- Department of Internal Medicine Faculty of Medical Sciences, University of Campinas (UNICAMP) Campinas São Paulo Brazil
| | - Adelino S. R. da Silva
- School of Physical Education and Sport of Ribeirao Preto and Postgraduate Program in Rehabilitation and Functional Performance Ribeirao Preto Medical School, University of Sao Paulo Ribeirao Preto São Paulo Brazil
- Laboratory of Nutritional Genomics (LabGeN) School of Applied Sciences, University of Campinas (UNICAMP) Limeira São Paulo Brazil
| | - Leandro P. Moura
- Laboratory of Molecular Biology of Exercise (LaBMEx) School of Applied Sciences, University of Campinas Limeira São Paulo Brazil
| | - Dennys E. Cintra
- Laboratory of Nutritional Genomics (LabGeN) School of Applied Sciences, University of Campinas (UNICAMP) Limeira São Paulo Brazil
| | - José R. Pauli
- Laboratory of Molecular Biology of Exercise (LaBMEx) School of Applied Sciences, University of Campinas Limeira São Paulo Brazil
- CEPECE ‐ Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP) Limeira São Paulo Brazil
| | - Eduardo R. Ropelle
- Laboratory of Molecular Biology of Exercise (LaBMEx) School of Applied Sciences, University of Campinas Limeira São Paulo Brazil
- Department of Internal Medicine Faculty of Medical Sciences, University of Campinas (UNICAMP) Campinas São Paulo Brazil
- CEPECE ‐ Center of Research in Sport Sciences, School of Applied Sciences, University of Campinas (UNICAMP) Limeira São Paulo Brazil
- Laboratory of Cell Signaling Obesity and Comorbidities Research Center (OCRC), University of Campinas Campinas São Paulo Brazil
| |
Collapse
|
18
|
Taleski G, Sontag E. Protein phosphatase 2A and tau: an orchestrated 'Pas de Deux'. FEBS Lett 2017; 592:1079-1095. [PMID: 29121398 DOI: 10.1002/1873-3468.12907] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 10/18/2017] [Accepted: 11/02/2017] [Indexed: 12/21/2022]
Abstract
The neuronal microtubule-associated protein tau serves a critical role in regulating axonal microtubule dynamics to support neuronal and synaptic functions. Furthermore, it contributes to glutamatergic regulation and synaptic plasticity. Emerging evidence also suggests that tau serves as a signaling scaffold. Tau function and subcellular localization are tightly regulated, in part, by the orchestrated interplay between phosphorylation and dephosphorylation events. Significantly, protein phosphatase type 2A (PP2A), encompassing the regulatory PPP2R2A (or Bα) subunit, is a major brain heterotrimeric enzyme and the primary tau Ser/Thr phosphatase in vivo. Herein, we closely examine how the intimate and compartmentalized interactions between PP2A and tau regulate tau phosphorylation and function, and play an essential role in neuronal homeostasis. We also review evidence supporting a strong link between deregulation of tau-PP2A functional interactions and the molecular underpinnings of various neurodegenerative diseases collectively called tauopathies. Lastly, we discuss the opportunities and associated challenges in more specifically targeting PP2A-tau interactions for drug development for tauopathies.
Collapse
Affiliation(s)
- Goce Taleski
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia
| | - Estelle Sontag
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Hunter Medical Research Institute, University of Newcastle, Callaghan, Australia
| |
Collapse
|