1
|
Shahwan M, Yadav DK, Khan MS, Choudhury A, Shamsi A, Hassan MI, Anwar S. Mechanistic insights into the inhibition of microtubule affinity-regulating kinase 4 by Syringic acid: A computational and experimental study. Int J Biol Macromol 2025; 309:142812. [PMID: 40216145 DOI: 10.1016/j.ijbiomac.2025.142812] [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: 02/15/2025] [Revised: 03/20/2025] [Accepted: 04/02/2025] [Indexed: 04/19/2025]
Abstract
Microtubule affinity regulating kinase 4 (MARK4) is a vital protein kinase that serves as a dual target in cancer and neurodegenerative diseases. It is implicated in the development of tauopathies and also linked to the pathogenesis of several cancer types, implying its importance. Syringic acid is a naturally occurring phenolic molecule that has shown significant efficacies in cancer and neurodegenerative diseases by modulating several key pathways. Thus, the present study aims to investigate the inhibitory potential of syringic acid against the protein kinase MARK4, employing a combination of experimental and computational approaches. Molecular docking revealed the binding of syringic acid in the MARK4's binding pocket, interacting with key functional residues of the protein kinase. Molecular dynamic simulation (MD) studies demonstrated the conformational dynamics and structural stability of MARK4 upon the binding of syringic acid. In silico findings were further complemented by experimental assays. Enzyme inhibition assay showed that syringic acid effectively inhibits MARK4 with an IC50 value of 4.32 μM. Fluorescence binding assays revealed a strong binding affinity (K = 2.8 × 106 M-1). The findings of our study establish syringic acid as a potent MARK4 inhibitor, providing a perfect platform for its use in tackling MARK4-associated diseases.
Collapse
Affiliation(s)
- Moyad Shahwan
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, Ajman, United Arab Emirates; Center for Medical and Bio-Allied Health Sciences Research, Ajman University, United Arab Emirates
| | - Dharmendra Kumar Yadav
- Gachon Institute of Pharmaceutical Science and Department of Pharmacy, College of Pharmacy, Gachon University, Incheon, Republic of Korea.
| | | | - Arunabh Choudhury
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Anas Shamsi
- Center for Medical and Bio-Allied Health Sciences Research, Ajman University, United Arab Emirates
| | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Saleha Anwar
- Center for Medical and Bio-Allied Health Sciences Research, Ajman University, United Arab Emirates; Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India.
| |
Collapse
|
2
|
Seydel B, Hegner P, Lauerer AM, Schildt S, Bayram F, Tafelmeier M, Wermers D, Rupprecht L, Schmid C, Wagner S, Maier LS, Arzt M, Lebek S. Increased Myocardial MARK4 Expression in Patients with Heart Failure and Sleep-Disordered Breathing. Int J Mol Sci 2025; 26:3614. [PMID: 40332117 PMCID: PMC12027440 DOI: 10.3390/ijms26083614] [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: 02/19/2025] [Revised: 04/05/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Cardiovascular diseases are the leading cause of morbidity and mortality worldwide, underscoring the urgent need for novel therapeutic targets and strategies. The kinase MARK4 (MAP (microtubule-associated proteins)/microtubule affinity-regulating kinase 4) regulates microtubule-associated proteins pivotal for cell polarity, protein stability, and intracellular signaling. Animal models of heart failure revealed elevated MARK4 levels, which correlated with impaired cardiac contractility. However, the involvement of MARK4 and its potential as a molecular drug target has not yet been explored in the myocardium of cardiovascular patients. We investigated the MARK4 mRNA expression in human myocardial biopsies of 152 high-risk cardiovascular patients undergoing cardiac surgery. Comprehensive echocardiography as well as testing for sleep-disordered breathing (SDB), a critical comorbidity in heart failure, were assessed preoperatively. We observed a substantial upregulation of myocardial MARK4 expression in patients with impaired cardiac contractility, resulting in an inverse correlation with the left ventricular ejection fraction. Myocardial MARK4 expression also correlated with echocardiographic E/e', a central parameter of diastolic dysfunction. Mechanistically, our analyses revealed that MARK4 expression increases in SDB and under hypoxic conditions, as evidenced by significant correlations between myocardial MARK4 expression and factors like mean oxygen saturation, time with oxygen saturation below 90%, and the oxygen desaturation index. Multivariable regression analysis revealed that both left ventricular ejection fraction and mean oxygen saturation were independently associated with dysregulated MARK4 levels, even when controlling for important clinical covariables as potential confounders. Taken together, our findings demonstrate that MARK4 expression is highly increased in the myocardium of cardiovascular high-risk patients, suggesting it is a potential molecular target against cardiovascular diseases.
Collapse
Affiliation(s)
- Bettina Seydel
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Philipp Hegner
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Anna-Maria Lauerer
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Sönke Schildt
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Fatma Bayram
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Maria Tafelmeier
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Dominik Wermers
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Leopold Rupprecht
- Department of Cardiothoracic Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Christof Schmid
- Department of Cardiothoracic Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Stefan Wagner
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Lars Siegfried Maier
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Michael Arzt
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| | - Simon Lebek
- Department of Internal Medicine II, University Hospital Regensburg, 93053 Regensburg, Germany; (B.S.)
| |
Collapse
|
3
|
Wu G, Luo Y, Guo Q, Yang M, Mahaman YAR, Liu Y, Wang JZ, Liu R, Gao X, Wang X. Conformation pattern changes in R1-pS262 tau peptide induced endogenous tau aggregation, synaptic damage, and cognitive impairments. J Alzheimers Dis 2025; 103:951-965. [PMID: 39686621 DOI: 10.1177/13872877241307341] [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: 12/18/2024]
Abstract
BACKGROUND To date, the effect of tau phosphorylation at different amino acid sites on the conformation and function of tau is still unclear in Alzheimer's disease (AD). Protein fingerprinting, also known as the protein folding shape code (PFSC) method, is a protein structure prediction technique based on protein sequence, which can reveal proteins' most likely spatial conformation. OBJECTIVE To investigate the effect of phosphorylation on tau protein conformation using PFSC technology and further analyze the differences in the effect of phosphorylation on tau aggregation at specific sites. METHODS We performed a conformational analysis of wild-type and simulated mutant hTau441 using the PFSC method and synthesized the phosphorylated and non-phosphorylated tau fragments by the chemical solid phase method. RESULTS We found that the number of Ser262 protein fingerprints increased from six in tau S262A to nine in tau S262E, together with increased conformational changes and enhanced flexibility. The in vitro Thioflavin S assay showed that phosphorylated tau fragments R1-pS262 possessed a stronger activity of inducing tau aggregation. In contrast to the non-phosphorylated tau fragment R1-nS262, R1-pS262 promoted endogenous tau aggregation and decreased synaptic proteins. In rats, R1-pS262 caused cognitive impairments and neuronal loss in addition to endogenous tau aggregation and synaptic damage. CONCLUSIONS Our study firstly reports that tau phosphorylation at Ser262 induces tau aggregation, and phosphorylated tau fragments R1-pS262 directly result in neuropathological changes. These provide new clues to the pathogenesis of tauopathy, such as AD, and a new molecular target for possible intervention.
Collapse
Affiliation(s)
- Gang Wu
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yong Luo
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Qian Guo
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingming Yang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yacoubou Abdoul Razak Mahaman
- Hubei Provincial Key Laboratory of Occurrence and Intervention of Kidney Diseases, Medical College, Hubei Polytechnic University, Huangshi, China
| | - Yi Liu
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, School of Medicine, Jianghan University, Wuhan, China
| | - Rong Liu
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiang Gao
- Department of Pharmacy, Renmin Hospital, Wuhan University, Wuhan, China
| | - Xiaochuan Wang
- School of Basic Medicine, Key Laboratory of Education Ministry/Hubei province of China for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Hubei Key Laboratory of Cognitive and Affective Disorders, School of Medicine, Jianghan University, Wuhan, China
| |
Collapse
|
4
|
Hussain A, Jairajpuri DS, Anwar S, Choudhury A, Hawwal MF, Firdous A, Alajmi MF, Hassan MI. Apigenin-mediated MARK4 inhibition: a novel approach in advancing Alzheimer's disease therapeutics. Mol Divers 2025:10.1007/s11030-025-11104-x. [PMID: 39841316 DOI: 10.1007/s11030-025-11104-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/01/2025] [Indexed: 01/23/2025]
Abstract
Apigenin, a dietary flavonoid with notable anti-cancer properties, has emerged as a promising candidate for the treatment of neurodegenerative disorders, particularly Alzheimer's disease (AD). While extensively studied for its ability to modulate key molecular pathways in cancers, apigenin also exerts neuroprotective effects by reducing neuroinflammation, protecting neurons from oxidative stress, and enhancing neuronal survival and synaptic plasticity. This dual functionality makes apigenin an intriguing therapeutic option for diseases like AD, where kinase dysregulation plays a central role. In this study, we focus on Microtubule Affinity-Regulating Kinase 4 (MARK4), a key enzyme implicated in tauopathies associated with AD, as well as in cancer progression. Through in silico analysis, we explore the interaction between apigenin and MARK4, revealing significant structural changes within the kinase domain upon ligand binding. These computational findings were confirmed via experimental assays using purified recombinant MARK4, where apigenin demonstrated potent inhibition with an IC50 value of 2.39 µM. Fluorescence binding assays further confirmed a strong binding affinity (Ka = 108 M-1), indicating that apigenin efficiently occupies the MARK4 active site, thereby suppressing its enzymatic activity. These results position apigenin as a potent inhibitor of MARK4, offering a dual therapeutic advantage-both as an anti-cancer agent and as a neuroprotective compound for the potential treatment of AD. This study opens new avenues for the development of apigenin-based therapeutics targeting kinase dysregulation in cancer and neurodegeneration.
Collapse
Affiliation(s)
- Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Deeba Shamim Jairajpuri
- Department of Medical Biochemistry, College of Medicine and Health Sciences, Arabian Gulf University, Manama, Kingdom of Bahrain
| | - Saleha Anwar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Arunabh Choudhury
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Mohammed F Hawwal
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Anam Firdous
- Department of Biotechnology, Jamia Hamdard, New Delhi, 110062, India
| | - Mohamed F Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| |
Collapse
|
5
|
Kolling LJ, Chimenti MS, Marcinkiewcz CA. Spatial differences in gene expression across the dorsal raphe nucleus in a model of early Alzheimer's disease. J Alzheimers Dis 2025; 103:133-148. [PMID: 39584353 PMCID: PMC12047055 DOI: 10.1177/13872877241299119] [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] [Indexed: 11/26/2024]
Abstract
BACKGROUND Persons with Alzheimer's disease (AD) present with changes in mood, sleep, and arousal that may precede the clinical manifestation of cognitive decline. These early symptoms can be driven by changes in the serotonergic (5-HT) nuclei of the brainstem, particularly the dorsal raphe nucleus (DRN). It is unclear why all 5-HT neurons do not simultaneously develop AD pathology that progresses at the same rate. OBJECTIVE We sought to identify any underlying genetic components associated with susceptibility or resistance of 5-HT neurons to AD pathology. METHODS The Visium Spatial Gene Expression platform was used to identify transcriptomic changes across the DRN in a preclinical model of early AD, human tau-overexpressing mice (htau mice). We further used RNAscope and immunohistochemical assessment to validate findings of primary interest. RESULTS We find that the DRN of htau mice differentially expresses AD-related genes, including those related to kinase binding, ion channel activity, ligand-receptor interactions, and regulation of serine/threonine kinases. We further find that computational sub-clustering of the DRN is consistent with previous circuitry-driven characterizations, allowing for spatial bounding of distinct subregions within the DRN. Of these, we find the dorsolateral DRN is preferentially impacted by 5-HT neuron loss and development of tau pathology, which coincides with increased expression of the long noncoding RNA Map2k3os. CONCLUSIONS Map2k3os may serve regulatory roles relevant for tau phosphorylation and warrants further investigation to characterize its interactions. Overall, this report demonstrates the power of large-scale spatial transcriptomics technologies, while underscoring the need for convergent-data validation to overcome their limitations.
Collapse
Affiliation(s)
- Louis John Kolling
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa, USA
| | - Michael Sandro Chimenti
- Iowa Institute of Human Genetics Bioinformatics Division, University of Iowa, Iowa City, Iowa, USA
| | | |
Collapse
|
6
|
Qin B, Chen X, Wang F, Wang Y. DUBs in Alzheimer's disease: mechanisms and therapeutic implications. Cell Death Discov 2024; 10:475. [PMID: 39562545 DOI: 10.1038/s41420-024-02237-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 11/21/2024] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder characterized by the accumulation of amyloid β protein (Aβ) and the hyper-phosphorylation of the microtubule-associated protein Tau. The ubiquitin-proteasome system (UPS) plays a pivotal role in determining the fate of proteins, and its dysregulation can contribute to the buildup of Aβ and Tau. Deubiquitinating enzymes (DUBs), working in conjunction with activating enzymes (E1), ubiquitin-conjugating enzymes (E2), and ubiquitin ligases (E3), actively maintain the delicate balance of protein homeostasis. DUBs specifically remove ubiquitin tags from proteins marked for degradation, thereby averting their proteasomal breakdown. Several DUBs have demonstrated their capacity to regulate the levels of Aβ and Tau by modulating their degree of ubiquitination, underscoring their potential as therapeutic targets for AD. In this context, we present a comprehensive review of AD-associated DUBs and elucidate their physiological roles. Moreover, we delve into the current advancements in developing inhibitors targeting these DUBs, including the determination of cocrystal structures with their respective targets. Additionally, we assess the therapeutic efficacy of these inhibitors in AD, aiming to establish a theoretical foundation for future AD treatments.
Collapse
Affiliation(s)
- Biying Qin
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Xiaodong Chen
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Feng Wang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Yanfeng Wang
- Key Laboratory of Molecular Medicine and Biotherapy, School of Life Science, Beijing Institute of Technology, Beijing, China.
- Tangshan Research Institute, Beijing Institute of Technology, Tangshan, Hebei, China.
- Advanced Technology Research Institute, Beijing Institute of Technology, Jinan, Shandong, China.
| |
Collapse
|
7
|
Liu Y, Chen Y, Fukui K. α-Tocotrienol Protects Neurons by Preventing Tau Hyperphosphorylation via Inhibiting Microtubule Affinity-Regulating Kinase Activation. Int J Mol Sci 2024; 25:8428. [PMID: 39125998 PMCID: PMC11313320 DOI: 10.3390/ijms25158428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
In the pathological process of Alzheimer's disease, neuronal cell death is closely related to the accumulation of reactive oxygen species. Our previous studies have found that oxidative stress can activate microtubule affinity-regulating kinases, resulting in elevated phosphorylation levels of tau protein specifically at the Ser262 residue in N1E-115 cells that have been subjected to exposure to hydrogen peroxide. This process may be one of the pathogenic mechanisms of Alzheimer's disease. Vitamin E is a fat-soluble, naturally occurring antioxidant that plays a crucial role in biological systems. This study aimed to examine the probable processes that contribute to the inhibiting effect on the abnormal phosphorylation of tau protein and the neuroprotective activity of a particular type of vitamin E, α-tocotrienol. The experimental analysis revealed that α-tocotrienol showed significant neuroprotective effects in the N1E-115 cell line. Our data further suggest that one of the mechanisms underlying the neuroprotective effects of α-tocotrienol may be through the inhibition of microtubule affinity-regulated kinase activation, which significantly reduces the oxidative stress-induced aberrant elevation of p-Tau (Ser262) levels. These results indicate that α-tocotrienol may represent an intriguing strategy for treating or preventing Alzheimer's disease.
Collapse
Affiliation(s)
- Yuhong Liu
- Molecular Cell Biology Laboratory, Department of Functional Control Systems, Graduate School of Engineering and Science, Shibaura Institute of Technology, Saitama 337-8570, Japan;
| | - Yunxi Chen
- Molecular Cell Biology Laboratory, Department of Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Saitama 337-8570, Japan;
| | - Koji Fukui
- Molecular Cell Biology Laboratory, Department of Functional Control Systems, Graduate School of Engineering and Science, Shibaura Institute of Technology, Saitama 337-8570, Japan;
- Molecular Cell Biology Laboratory, Department of Systems Engineering and Science, Graduate School of Engineering and Science, Shibaura Institute of Technology, Saitama 337-8570, Japan;
| |
Collapse
|
8
|
Zhu Y, Li M, Wang H, Yang F, Du R, Pang X, Bai J, Huang X. Mendelian Randomization Identifies Genetically Supported Drug Targets for Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Mol Neurobiol 2024; 61:3809-3818. [PMID: 38019415 DOI: 10.1007/s12035-023-03817-7] [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: 08/26/2023] [Accepted: 11/18/2023] [Indexed: 11/30/2023]
Abstract
Currently, amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) have no effective treatments. Drug repurposing offers a rapid method to meet therapeutic need for ALS and FTD. To identify therapeutic targets associated with ALS and FTD, Mendelian randomization (MR) analysis and colocalization were performed. Genetic instruments were based on transcriptomic and proteomic data for 422 actionable proteins targeted by approved drugs or clinical drug candidates. The publicly available ALS GWAS summary data (including a total of 20,806 ALS cases and 59,804 controls) and FTD GWAS summary data (including a total of 2154 patients with FTD and 4308 controls) were used. Using cis-expression quantitative trait loci and cis-protein quantitative trait loci genetic instruments, we identified several drug targets for repurposing (ALS: MARK3, false-discovery rate (FDR) = 0.043; LTBR, FDR = 0.068) (FTD: HLA-DRB1, FDR = 0.083; ADH5, FDR = 0.056). Our MR study analyzed the actionable druggable proteins and provided potential therapeutic targets for ALS and FTD. Future studies should further elucidate the underlying mechanism of corresponding drug targets in the pathogenesis of ALS and FTD.
Collapse
Affiliation(s)
- Yahui Zhu
- Medical School of Chinese PLA, Beijing, China
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Mao Li
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hongfen Wang
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Fei Yang
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - RongRong Du
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
- College of Medicine, Nankai University, Tianjin, China
| | - Xinyuan Pang
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
- College of Medicine, Nankai University, Tianjin, China
| | - Jiongming Bai
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
- College of Medicine, Nankai University, Tianjin, China
| | - Xusheng Huang
- Medical School of Chinese PLA, Beijing, China.
- Department of Neurology, the First Medical Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
9
|
Hole KL, Zhu B, Huggon L, Brown JT, Mason JM, Williams RJ. Tau P301L disengages from the proteosome core complex and neurogranin coincident with enhanced neuronal network excitability. Cell Death Dis 2024; 15:429. [PMID: 38890273 PMCID: PMC11189525 DOI: 10.1038/s41419-024-06815-2] [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: 10/30/2023] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024]
Abstract
Tauopathies are characterised by the pathological accumulation of misfolded tau. The emerging view is that toxic tau species drive synaptic dysfunction and potentially tau propagation before measurable neurodegeneration is evident, but the underlying molecular events are not well defined. Human non-mutated 0N4R tau (tauWT) and P301L mutant 0N4R tau (tauP301L) were expressed in mouse primary cortical neurons using adeno-associated viruses to monitor early molecular changes and synaptic function before the onset of neuronal loss. In this model tauP301L was differentially phosphorylated relative to tauwt with a notable increase in phosphorylation at ser262. Affinity purification - mass spectrometry combined with tandem mass tagging was used to quantitatively compare the tauWT and tauP301L interactomes. This revealed an enrichment of tauP301L with ribosomal proteins but a decreased interaction with the proteasome core complex and reduced tauP301L degradation. Differences in the interaction of tauP301L with members of a key synaptic calcium-calmodulin signalling pathway were also identified, most notably, increased association with CaMKII but reduced association with calcineurin and the candidate AD biomarker neurogranin. Decreased association of neurogranin to tauP301L corresponded with the appearance of enhanced levels of extracellular neurogranin suggestive of potential release or leakage from synapses. Finally, analysis of neuronal network activity using micro-electrode arrays showed that overexpression of tauP301L promoted basal hyperexcitability coincident with these changes in the tau interactome and implicating tau in specific early alterations in synaptic function.
Collapse
Affiliation(s)
- Katriona L Hole
- Department of Life Sciences, University of Bath, Bath, UK
- The Francis Crick Institute, London, UK
| | - Bangfu Zhu
- Department of Life Sciences, University of Bath, Bath, UK
| | - Laura Huggon
- Department of Life Sciences, University of Bath, Bath, UK
- UK Dementia Research Institute at King's College London, London, UK
| | - Jon T Brown
- Department of Clinical and Biomedical Sciences, University of Exeter, Exeter, UK
| | - Jody M Mason
- Department of Life Sciences, University of Bath, Bath, UK
| | | |
Collapse
|
10
|
Sultanakhmetov G, Limlingan SJM, Fukuchi A, Tsuda K, Suzuki H, Kato I, Saito T, Weitemier AZ, Ando K. Mark4 ablation attenuates pathological phenotypes in a mouse model of tauopathy. Brain Commun 2024; 6:fcae136. [PMID: 38712317 PMCID: PMC11073748 DOI: 10.1093/braincomms/fcae136] [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: 12/02/2023] [Revised: 02/20/2024] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
Accumulation of abnormally phosphorylated tau proteins is linked to various neurodegenerative diseases, including Alzheimer's disease and frontotemporal dementia. Microtubule affinity-regulating kinase 4 (MARK4) has been genetically and pathologically associated with Alzheimer's disease and reported to enhance tau phosphorylation and toxicity in Drosophila and mouse traumatic brain-injury models but not in mammalian tauopathy models. To investigate the role of MARK4 in tau-mediated neuropathology, we crossed P301S tauopathy model (PS19) and Mark4 knockout mice. We performed behaviour, biochemical and histology analyses to evaluate changes in PS19 pathological phenotype with and without Mark4. Here, we demonstrated that Mark4 deletion ameliorated the tau pathology in a mouse model of tauopathy. In particular, we found that PS19 with Mark4 knockout showed improved mortality and memory compared with those bearing an intact Mark4 gene. These phenotypes were accompanied by reduced neurodegeneration and astrogliosis in response to the reduction of pathological forms of tau, such as those phosphorylated at Ser356, AT8-positive tau and thioflavin S-positive tau. Our data indicate that MARK4 critically contributes to tau-mediated neuropathology, suggesting that MARK4 inhibition may serve as a therapeutic avenue for tauopathies.
Collapse
Affiliation(s)
- Grigorii Sultanakhmetov
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Sophia Jobien M Limlingan
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Aoi Fukuchi
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Keisuke Tsuda
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Hirokazu Suzuki
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Iori Kato
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Taro Saito
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
- Department of Biological Sciences, School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Adam Z Weitemier
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
- Department of Biological Sciences, School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Kanae Ando
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
- Department of Biological Sciences, School of Science, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| |
Collapse
|
11
|
Anwar S, Choudhury A, Hussain A, AlAjmi MF, Hassan MI, Islam A. Harnessing memantine in Alzheimer's disease therapy through inhibition of microtubule affinity-regulating kinase: Mechanistic insights. Int J Biol Macromol 2024; 262:130090. [PMID: 38342269 DOI: 10.1016/j.ijbiomac.2024.130090] [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: 10/10/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/13/2024]
Abstract
Alzheimer's disease (AD) is one of the neurodegenerative disorder that primarily affects memory, thinking, and behavior, eventually leading to severe cognitive impairment. Therapeutic management of AD is urgently needed to improve the quality and lifestyle of patients. Tau phosphorylating kinases are considered attractive therapeutic targets. Microtubule affinity-regulating kinase 4 (MARK4) is directly linked with pathological phosphorylations of tau, highlighting its role in the therapeutic targeting of AD. The current manuscript shows the MARK4 inhibitory effect of Memantine (MEM), a drug used in treating AD. We have performed fluorescence based binding measurements, enzyme inhibition assay, docking and molecular dynamics (MD) simulations to understand the binding of of MARK4 and MEM and subsequent inhibition in the kinase activity. A 100 ns MD simulations provided a detailed analysis of MARK4-MEM complex and the role of potential critical residues in the binding. Finally, this study provides molecular insights into the therapeutic implication of MEM in AD therapeutics. We propose MEM effectively inhibits MARK4, it may be implicated in the development of targeted and efficient treatments for AD.
Collapse
Affiliation(s)
- Saleha Anwar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Arunabh Choudhury
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed F AlAjmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
12
|
Pena E, San Martin-Salamanca R, El Alam S, Flores K, Arriaza K. Tau Protein Alterations Induced by Hypobaric Hypoxia Exposure. Int J Mol Sci 2024; 25:889. [PMID: 38255962 PMCID: PMC10815386 DOI: 10.3390/ijms25020889] [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: 11/15/2023] [Revised: 12/21/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Tauopathies are a group of neurodegenerative diseases whose central feature is dysfunction of the microtubule-associated protein tau (MAPT). Although the exact etiology of tauopathies is still unknown, it has been hypothesized that their onset may occur up to twenty years before the clear emergence of symptoms, which has led to questions about whether the prognosis of these diseases can be improved by, for instance, targeting the factors that influence tauopathy development. One such factor is hypoxia, which is strongly linked to Alzheimer's disease because of its association with obstructive sleep apnea and has been reported to affect molecular pathways related to the dysfunction and aggregation of tau proteins and other biomarkers of neurological damage. In particular, hypobaric hypoxia exposure increases the activation of several kinases related to the hyperphosphorylation of tau in neuronal cells, such as ERK, GSK3β, and CDK5. In addition, hypoxia also increases the levels of inflammatory molecules (IL-β1, IL-6, and TNF-α), which are also associated with neurodegeneration. This review discusses the many remaining questions regarding the influence of hypoxia on tauopathies and the contribution of high-altitude exposure to the development of these diseases.
Collapse
Affiliation(s)
| | | | - Samia El Alam
- High Altitude Medicine Research Center (CEIMA), Arturo Prat University, Iquique 1110939, Chile; (E.P.); (R.S.M.-S.); (K.F.); (K.A.)
| | | | | |
Collapse
|
13
|
Mumtaz, Ahmed F, Rabbani SA, El-Tanani M, Najmi AK, Ali J, Khan MA. Tauopathy in AD: Therapeutic Potential of MARK-4. Curr Alzheimer Res 2024; 21:779-790. [PMID: 39931856 DOI: 10.2174/0115672050358397250126151707] [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: 09/17/2024] [Revised: 11/26/2024] [Accepted: 12/05/2024] [Indexed: 04/30/2025]
Abstract
Alzheimer's disease (AD) is one of the leading causes of cognitive decline, which leads to dementia and poses significant challenges for its therapy. The reason is primarily the ineffective available treatments targeting the underlying pathology of AD. It is a neurodegenerative disease that is mainly characterised by the various molecular pathways contributing to its complex pathology, including extracellular amyloid beta (Aβ) plaques, intracellular neurofibrillary tangles (NFTs), oxidative stress, and neuroinflammation. One of the crucial features is the hyperphosphorylation of tau proteins, which is facilitated by microtubule affinity-regulating kinase-4 (MARK-4). The kinase plays a crucial role in the disease development by modifying microtubule integrity, leading to neuronal dysfunction and death. MARK-4 is thus a druggable target and has a pivotal role in AD. Amongst MARK-4 inhibitors, 16 compounds demonstrate significant capacity in molecular docking studies, showing high binding affinity to MARK-4 and promising potential for tau inhibition. Further, in-vitro investigations provide evidence of their neuroprotective properties. The present review mainly focuses on the role of MARK-4 and its potential inhibitors used in treating AD, which have been thoroughly investigated in silico and in vitro..
Collapse
Affiliation(s)
- Mumtaz
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Faraha Ahmed
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Syed Arman Rabbani
- RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Mohamed El-Tanani
- RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| |
Collapse
|
14
|
Oba T, Homma D, Limlingan SJM, Fukuchi A, Asada A, Saito T, Ando K. A cell-penetrating peptide derived from SARS-CoV-2 protein Orf9b allosterically inhibits MARK4 activity and mitigates tau toxicity. Neurobiol Dis 2023; 188:106334. [PMID: 37884211 DOI: 10.1016/j.nbd.2023.106334] [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: 09/04/2023] [Revised: 10/12/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023] Open
Abstract
Abnormal activation of microtubule affinity-regulating kinase 4 (MARK4) and its phosphorylation of the microtubule-associated protein tau are believed to play a role in the pathogenesis of Alzheimer's disease, and MARK4 inhibition can be a strategy to develop disease-modifying therapy. Here we report the development of a membrane-permeable peptide that inhibits MARK4 activity in an allosteric manner. The SARS-CoV-2-derived protein Orf9b inhibited MARK4-mediated tau phosphorylation in primary neurons and Drosophila. Orf9b inhibited MARK4 activity in an allosteric manner and did not inhibit the activity of MARK2, which is another MARK family member and is closely related to MARK4. Co-expression of Orf9b in the fly retina expressing human tau and MARK4 suppressed phosphorylation of tau at the microtubule-binding repeats and tau-induced neurodegeneration. We identified the minimal sequence of Orf9b required to suppress MARK4 activity and fused it to a cell-permeable sequence (TAT-Orf9b10-18_78-95). Extracellular supplementation of TAT-Orf9b10-18_78-95 inhibited MARK4 activity in primary neurons, and feeding TAT-Orf9b10-18_78-95 to a fly model of tauopathy lowered phospho-tau levels and suppressed neurodegeneration. These results suggest that TAT-Orf9b10-18_78-95 is a unique class of MARK4 inhibitor and can be used to modify tau toxicity.
Collapse
Affiliation(s)
- Toshiya Oba
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Japan
| | - Daiki Homma
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Japan
| | - Sophia Jobien M Limlingan
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Japan
| | - Aoi Fukuchi
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Japan
| | - Akiko Asada
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Japan; Department of Biological Sciences, Faculty of Science, Tokyo Metropolitan University, Japan
| | - Taro Saito
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Japan; Department of Biological Sciences, Faculty of Science, Tokyo Metropolitan University, Japan.
| | - Kanae Ando
- Laboratory of Molecular Neuroscience, Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan University, Japan; Department of Biological Sciences, Faculty of Science, Tokyo Metropolitan University, Japan.
| |
Collapse
|
15
|
Alam M, Ahmed S, Abid M, Hasan GM, Islam A, Hassan MI. Therapeutic targeting of microtubule affinity-regulating kinase 4 in cancer and neurodegenerative diseases. J Cell Biochem 2023; 124:1223-1240. [PMID: 37661636 DOI: 10.1002/jcb.30468] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/12/2023] [Accepted: 08/21/2023] [Indexed: 09/05/2023]
Abstract
Microtubule affinity-regulating kinase 4 (MARK4) is a member of the Ser/Thr protein kinase family, phosphorylates the microtubule-connected proteins and plays a vital role in causing cancers and neurodegenerative diseases. This kinase modulates multiple signaling pathways, including mammalian target of rapamycin, nuclear factor-κB, and Hippo-signaling, presumably responsible for cancer and Alzheimer's. MARK4 acts as a negative controller of the Hippo-kinase cassette for promoting YAP/TAZ action, and the loss of MARK4 detains the tumorigenic properties of cancer cells. MARK4 is involved in tau hyperphosphorylation that consequently affects neurodegeneration. MARK4 is a promising drug target for cancer, diabetes, and Alzheimer's. Developing the potent and selective inhibitors of MAKR4 are promising in the therapeutic management of associated diseases. Despite its great significance, a few reviews are available to discuss its structure, function and clinical significance. In the current review, we aimed to provide detailed information on the structural features of MARK4 targeted in drug development and its role in various signaling pathways related to cancer and neurodegenerative diseases. We further described the therapeutic potential of MARK4 inhibitors in preventing numerous diseases. Finally, the updated information on MARK4 will be helpful in the further development of effective therapeutic molecules.
Collapse
Affiliation(s)
- Manzar Alam
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Sarfraz Ahmed
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Mohammad Abid
- Department of Biosciences, Jamia Millia Islamia, New Delhi, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Asimul Islam
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Md Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
16
|
Voura M, Anwar S, Sigala I, Parasidou E, Fragoulidou S, Hassan MI, Sarli V. Synthesis, Structural Modification, and Bioactivity Evaluation of Substituted Acridones as Potent Microtubule Affinity-Regulating Kinase 4 Inhibitors. ACS Pharmacol Transl Sci 2023; 6:1052-1074. [PMID: 37470016 PMCID: PMC10353068 DOI: 10.1021/acsptsci.3c00084] [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: 04/19/2023] [Indexed: 07/21/2023]
Abstract
Acridones present numerous pharmacological activities, including inhibition of microtubule affinity-regulating kinase 4 (MARK4) kinase activity. To investigate structure-activity relationships and develop potent MARK4 inhibitors, derivatives of 2-methylacridone were synthesized and tested for their activity against MARK4 kinase. Selective substitutions at the nitrogen atom were accomplished by treating 2-methylacridone with alkyl halides in the presence of K2CO3. In addition, amidation of acridone acetic acid 11 with piperazine or tryptophan methyl ester followed by derivatization with various amines gave a series of new acridone derivatives. Among the tested compounds, six were identified as possessing high inhibitory activity against MARK4. The molecular modeling studies showed that the derivatives bearing piperazine or tryptophan bind well to the ATP-binding site of MARK4. The antiproliferative activity of six active compounds was evaluated against HeLa and U87MG cancer cells. Tryptophan derivatives 23a, 23b, and 23c showed significant cytotoxicity against both cell lines with EC50 values ranging from 2.13 to 4.22 μM, while derivatives bearing piperazine were found to be not cytotoxic. Additionally, compound 23a decreased the proliferation of human MDA-MB-435 and U251 cancer cells in the low micromolar range; however, it also affects the non-cancerous HGF cells. Due to their high binding affinity against MARK4, the synthesized compounds could be potential agents to target MARK4 against cancer and tauopathies.
Collapse
Affiliation(s)
- Maria Voura
- Laboratory
of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Saleha Anwar
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ioanna Sigala
- Laboratory
of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Eleftheria Parasidou
- Laboratory
of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Souzanna Fragoulidou
- Laboratory
of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Md. Imtaiyaz Hassan
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Vasiliki Sarli
- Laboratory
of Organic Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| |
Collapse
|
17
|
Anwar S, Mohammad T, Azhar MK, Fatima H, Alam A, Hasan GM, Islam A, Kaur P, Hassan MI. Investigating MARK4 inhibitory potential of Bacopaside II: Targeting Alzheimer's disease. Int J Biol Macromol 2023:125364. [PMID: 37315665 DOI: 10.1016/j.ijbiomac.2023.125364] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/19/2023] [Accepted: 06/11/2023] [Indexed: 06/16/2023]
Abstract
Microtubule affinity regulating kinase (MARK4) is known to hyperphosphorylate tau protein, which subsequently causes Alzheimer's disease (AD). MARK4 is a well-validated drug target for AD; thus, we employed its structural features to discover potential inhibitors. On the other hand, complementary and alternative medicines (CAMs) have been used for the treatment of numerous diseases with little side effects. In this regard, Bacopa monnieri extracts have been extensively used to treat neurological disorders because of their neuroprotective roles. The plant extract is used as a memory enhancer and a brain tonic. Bacopaside II is a major component of Bacopa monnieri; thus, we studied its inhibitory effects and binding affinity towards the MARK4. Bacopaside II show a considerable binding affinity for MARK4 (K = 107 M-1) and inhibited kinase activity with an IC50 value of 5.4 μM. To get atomistic insights into the binding mechanism, we performed Molecular dynamics (MD) simulation studies for 100 ns. Bacopaside II binds strongly to the active site pocket residues of MARK4 and a number of hydrogen bonds remain stable throughout the MD trajectory. Our findings provide the basis for the therapeutic implication of Bacopaside and its derivatives in MARK4-related neurodegenerative diseases, especially AD and neuroinflammation.
Collapse
Affiliation(s)
- Saleha Anwar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Taj Mohammad
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110026, India
| | - Md Khabeer Azhar
- Department of Biosciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Hera Fatima
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Afsar Alam
- Department of Computer Science, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110026, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
18
|
Alrouji M, DasGupta D, Ashraf GM, Bilgrami AL, Alhumaydhi FA, Al Abdulmonem W, Shahwan M, Alsayari A, Atiya A, Shamsi A. Inhibition of microtubule affinity regulating kinase 4 by an acetylcholinesterase inhibitor, Huperzine A: Computational and experimental approaches. Int J Biol Macromol 2023; 235:123831. [PMID: 36870649 DOI: 10.1016/j.ijbiomac.2023.123831] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023]
Abstract
Microtubule affinity regulating kinase 4 (MARK4), 752 amino acids long, belonging to the AMPK superfamily, plays a vital role in regulating microtubules due to its potential to phosphorylate microtubule-associated proteins (MAP's) and thus, MARK4 plays a key role in Alzheimer's disease (AD) pathology. MARK4 is a druggable target for cancer, neurodegenerative diseases, and metabolic disorders. In this study, we have evaluated the MARK4 inhibitory potential of Huperzine A (HpA), an acetylcholinesterase inhibitor (AChEI), a potential AD drug. Molecular docking revealed the key residues governing the MARK4-HpA complex formation. The structural stability and conformational dynamics of the MARK4-HpA complex was assessed by employing Molecular dynamics (MD) simulation. The results suggested that the binding of HpA with MARK4 leads to minimal structural alterations in the native conformation of MARK4, implying the stability of the MARK4-HpA complex. Isothermal titration calorimetry (ITC) studies deciphered that HpA binds to MARK4 spontaneously. Moreover, the kinase assay depicted significant inhibition of MARK by HpA (IC50 = 4.91 μM), implying it to be a potent MARK4 inhibitor that can be implicated in the treatment of MARK4-directed diseases.
Collapse
Affiliation(s)
- Mohammed Alrouji
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Debarati DasGupta
- 428 Church Street, College of Pharmacy, University of Michigan, Ann Arbor, MI 48109, United States of America
| | - Ghulam Md Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, University City, Sharjah 27272, United Arab Emirates
| | - Anwar L Bilgrami
- Deanship of Scientific Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Fahad A Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia
| | - Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, P.O. Box 6655, Buraidah 51452, Saudi Arabia
| | - Moyad Shahwan
- College of Pharmacy and Health Sciences, Ajman University, United Arab Emirates; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, United Arab Emirates
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University (KKU), Guraiger St., Abha 62529, Saudi Arabia; Complementary and Alternative Medicine Unit, King Khalid University (KKU), Guraiger St., Abha 62529, Saudi Arabia
| | - Akhtar Atiya
- Department of Pharmacognosy, College of Pharmacy, King Khalid University (KKU), Guraiger St., Abha 62529, Saudi Arabia.
| | - Anas Shamsi
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, United Arab Emirates.
| |
Collapse
|
19
|
Yang J, Niu H, Pang S, Liu M, Chen F, Li Z, He L, Mo J, Yi H, Xiao J, Huang Y. MARK3 kinase: Regulation and physiologic roles. Cell Signal 2023; 103:110578. [PMID: 36581219 DOI: 10.1016/j.cellsig.2022.110578] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
Microtubule affinity-regulating kinase 3 (MARK3), a member of the MARK family, regulates several essential pathways, including the cell cycle, ciliated cell differentiation, and osteoclast differentiation. It is important to understand the control of their activities as MARK3 contains an N-terminal serine/threonine kinase domain, ubiquitin-associated domain, and C-terminal kinase-associated domain, which perform multiple regulatory functions. These functions include post-translational modification (e.g., phosphorylation) and interaction with scaffolding and other proteins. Differences in the amino acid sequence and domain position result in different three-dimensional protein structures and affect the function of MARK3, which distinguish it from the other MARK family members. Recent data indicate a potential role of MARK3 in several pathological conditions, including congenital blepharophimosis syndrome, osteoporosis, and tumorigenesis. The present review focuses on the physiological and pathological role of MARK3, its regulation, and recent developments in the small molecule inhibitors of the MARK3 signalling cascade.
Collapse
Affiliation(s)
- Jingyu Yang
- Surgery of Mammary Gland and Thyroid Gland, the First People's Hospital of Yunnan Province, Panlong Campus, 157 Jinbi Road, Kunming 650032, Yunnan, People's Republic of China
| | - Heng Niu
- Surgery of Mammary Gland and Thyroid Gland, the First People's Hospital of Yunnan Province, Panlong Campus, 157 Jinbi Road, Kunming 650032, Yunnan, People's Republic of China
| | - ShiGui Pang
- Cancer Research Institute, The Affiliated Hospital of Guilin Medical University, Xiufeng Campus, 15 Lequn Road, Guilin 541001, Guangxi, People's Republic of China
| | - Mignlong Liu
- Cancer Research Institute, The Affiliated Hospital of Guilin Medical University, Xiufeng Campus, 15 Lequn Road, Guilin 541001, Guangxi, People's Republic of China
| | - Feng Chen
- Cancer Research Institute, The Affiliated Hospital of Guilin Medical University, Xiufeng Campus, 15 Lequn Road, Guilin 541001, Guangxi, People's Republic of China
| | - Zhaoxin Li
- Cancer Research Institute, The Affiliated Hospital of Guilin Medical University, Xiufeng Campus, 15 Lequn Road, Guilin 541001, Guangxi, People's Republic of China
| | - Lifei He
- Cancer Research Institute, The Affiliated Hospital of Guilin Medical University, Xiufeng Campus, 15 Lequn Road, Guilin 541001, Guangxi, People's Republic of China
| | - Jianmei Mo
- Cancer Research Institute, The Affiliated Hospital of Guilin Medical University, Xiufeng Campus, 15 Lequn Road, Guilin 541001, Guangxi, People's Republic of China
| | - Huijun Yi
- Cancer Research Institute, The Affiliated Hospital of Guilin Medical University, Xiufeng Campus, 15 Lequn Road, Guilin 541001, Guangxi, People's Republic of China
| | - Juanjuan Xiao
- Cancer Research Institute, The Affiliated Hospital of Guilin Medical University, Xiufeng Campus, 15 Lequn Road, Guilin 541001, Guangxi, People's Republic of China
| | - Yingze Huang
- Cancer Research Institute, The Affiliated Hospital of Guilin Medical University, Xiufeng Campus, 15 Lequn Road, Guilin 541001, Guangxi, People's Republic of China.
| |
Collapse
|
20
|
Hmila I, Marnissi B, Kamali-Moghaddam M, Ghram A. Aptamer-Assisted Proximity Ligation Assay for Sensitive Detection of Infectious Bronchitis Coronavirus. Microbiol Spectr 2023; 11:e0208122. [PMID: 36651727 PMCID: PMC9927260 DOI: 10.1128/spectrum.02081-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 12/21/2022] [Indexed: 01/19/2023] Open
Abstract
Infectious bronchitis virus (IBV) is a coronavirus responsible for major health problems in the poultry industry. New virus strains continue to appear, causing large economic losses. To develop a rapid and accurate new quantitative assay for diagnosis of the virus without DNA extraction, we selected highly specific single-stranded DNA (ssDNA) aptamers with a high affinity to IBV, using the systematic evolution of ligands by exponential enrichment (SELEX) technology for aptamer screening, followed by high-throughput sequencing technology. Two of these aptamers, AptIBV5 and AptIBV2, were used to establish homogenous and solid-phase proximity ligation assays (PLAs). The developed assays were evaluated for their sensitivity and specificity using collected field samples and then compared to the newly developed sandwich enzyme-linked aptamer assay (ELAA) and reverse transcription-quantitative PCR (qRT-PCR), as the gold-standard method. The solid-phase PLA showed a lower limit of detection and a broader dynamic range than the two other assays. The developed technique may serve as an alternative assay for the diagnosis of IBV, with the potential to be extended to the detection of other important animal or human viruses. IMPORTANCE Infectious bronchitis virus (IBV) causes high morbidity and mortality and large economic losses in the poultry industry. The virus has the ability to genetically mutate into new IBV strains, causing devastating disease and outbreaks. To better monitor the emergence of this virus, the development of a rapid and highly sensitive diagnostic method should be implemented. For this, we generated aptamers with high affinity and specificity to the IBV in an ssDNA library. Using two high-affinity aptamers, we developed a sandwich ELAA and a very sensitive aptamer-based proximity ligation assay (PLA). The new assay showed high sensitivity and specificity and was used to detect IBV in farm samples. The PLA was compared to the newly developed sandwich ELAA and qRT-PCR, as the gold-standard technique.
Collapse
Affiliation(s)
- Issam Hmila
- Laboratory of Epidemiology and Veterinary Microbiology, Institute Pasteur of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Boutheina Marnissi
- Laboratory of Epidemiology and Veterinary Microbiology, Institute Pasteur of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Masood Kamali-Moghaddam
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Abdeljelil Ghram
- Laboratory of Epidemiology and Veterinary Microbiology, Institute Pasteur of Tunis, University of Tunis El Manar, Tunis, Tunisia
| |
Collapse
|
21
|
Targeting inhibition of microtubule affinity regulating kinase 4 by Harmaline: Strategy to combat Alzheimer's disease. Int J Biol Macromol 2022; 224:188-195. [DOI: 10.1016/j.ijbiomac.2022.10.115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/03/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022]
|
22
|
Cao MC, Scotter EL. Novel and known transcriptional targets of ALS/FTD protein TDP-43: Meta-analysis and interactive graphical database. Dis Model Mech 2022; 15:276263. [PMID: 35946434 PMCID: PMC9509890 DOI: 10.1242/dmm.049418] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 07/26/2022] [Indexed: 11/20/2022] Open
Abstract
TDP-43 proteinopathy is the major pathology in amyotrophic lateral sclerosis (ALS) and tau-negative frontotemporal dementia (FTD). Mounting evidence implicates loss of normal TDP-43 RNA processing function as a key pathomechanism. However, the RNA targets of TDP-43 differ by report, and have never been formally collated or compared between models and disease, hampering understanding of TDP-43 function. Here, we conducted re-analysis and meta-analysis of publicly available RNA-sequencing datasets from six TDP-43-knockdown models, and TDP-43-immunonegative neuronal nuclei from ALS/ FTD brain, to identify differentially expressed genes (DEGs) and exon usage (DEU) events. There was little overlap in DEGs between knockdown models, but PFKP, STMN2, CFP, KIAA1324 and TRHDE were common targets and were also differentially expressed in TDP-43-immunonegative neurons. DEG enrichment analysis revealed diverse biological pathways including immune and synaptic functions. Common DEU events in human datasets included well-known targets POLDIP3 and STMN2, and novel targets EXD3, MMAB, DLG5 and GOSR2. Our interactive database https://phpstack-449938-2576646.cloudwaysapps.com/ allows further exploration of TDP-43 DEG and DEU targets. Together, these data identify TDP-43 targets that can be exploited therapeutically or to validate loss-of-function processes.
Collapse
Affiliation(s)
- Maize C Cao
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand. 3A Symonds Street, Auckland 1010, New Zealand
| | - Emma L Scotter
- School of Biological Sciences and Centre for Brain Research, University of Auckland, Auckland, New Zealand. 3A Symonds Street, Auckland 1010, New Zealand
| |
Collapse
|
23
|
Bokharaie H, Kolch W, Krstic A. Analysis of Alternative mRNA Splicing in Vemurafenib-Resistant Melanoma Cells. Biomolecules 2022; 12:993. [PMID: 35883549 PMCID: PMC9312936 DOI: 10.3390/biom12070993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/11/2022] [Accepted: 07/14/2022] [Indexed: 01/09/2023] Open
Abstract
Alternative mRNA splicing is common in cancers. In BRAF V600E-mutated malignant melanoma, a frequent mechanism of acquired resistance to BRAF inhibitors involves alternative splicing (AS) of BRAF. The resulting shortened BRAF protein constitutively dimerizes and conveys drug resistance. Here, we have analysed AS in SK-MEL-239 melanoma cells and a BRAF inhibitor (vemurafenib)-resistant derivative that expresses an AS, shortened BRAF V600E transcript. Transcriptome analysis showed differential expression of spliceosome components between the two cell lines. As there is no consensus approach to analysing AS events, we used and compared four common AS softwares based on different principles, DEXSeq, rMATS, ASpli, and LeafCutter. Two of them correctly identified the BRAF V600E AS in the vemurafenib-resistant cells. Only 12 AS events were identified by all four softwares. Testing the AS predictions experimentally showed that these overlapping predictions are highly accurate. Interestingly, they identified AS caused alterations in the expression of melanin synthesis and cell migration genes in the vemurafenib-resistant cells. This analysis shows that combining different AS analysis approaches produces reliable results and meaningful, biologically testable hypotheses.
Collapse
Affiliation(s)
- Honey Bokharaie
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland; (H.B.); (W.K.)
- Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Walter Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland; (H.B.); (W.K.)
- Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland
| | - Aleksandar Krstic
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, D04 V1W8 Dublin 4, Ireland; (H.B.); (W.K.)
| |
Collapse
|
24
|
Park H, Yamanaka T, Nukina N. Proteomic analysis of heat-stable proteins revealed an increased proportion of proteins with compositionally biased regions. Sci Rep 2022; 12:4347. [PMID: 35289333 PMCID: PMC8921518 DOI: 10.1038/s41598-022-08044-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 02/17/2022] [Indexed: 11/09/2022] Open
Abstract
Intrinsically disordered proteins (IDPs) have been in the spotlight for their unique properties, such as their lack of secondary structures and low sequence complexity. Alpha-synuclein and tau are representative disease-related IDPs with low complexity regions in their sequences, accumulating in the brains of patients with Parkinson disease and Alzheimer disease, respectively. Their heat resistance in particular was what attracted our attention. We assumed that there exist many other unidentified proteins that are resistant to heat-treatment, referred to as heat-stable proteins, which would also have low sequence complexity. In this study, we performed proteomic analysis of heat-stable proteins of mouse brains and found that proteins with compositionally biased regions are abundant in the heat-stable proteins. The proteins related to neurodegeneration are known to undergo different types of post-translational modifications (PTMs) such as phosphorylation and ubiquitination. We then investigated the heat-stability and aggregation properties of phosphorylated synuclein and tau with different phosphorylation sites. We suggest that PTMs can be important factors that determine the heat-stability and aggregation properties of a protein. IDPs identified in the heat-stable proteins of mouse brains would be candidates for the pathogenic proteins for neurodegeneration.
Collapse
Affiliation(s)
- Hongsun Park
- Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, 1-3 Miyakodanitatara, Kyotanabe-shi, Kyoto, 610-0394, Japan
| | - Tomoyuki Yamanaka
- Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, 1-3 Miyakodanitatara, Kyotanabe-shi, Kyoto, 610-0394, Japan.,Department of Neuroscience of Disease, Brain Research Institute, Niigata University, Niigata, Japan
| | - Nobuyuki Nukina
- Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, 1-3 Miyakodanitatara, Kyotanabe-shi, Kyoto, 610-0394, Japan.
| |
Collapse
|
25
|
Deubiquitinating enzymes (DUBs): decipher underlying basis of neurodegenerative diseases. Mol Psychiatry 2022; 27:259-268. [PMID: 34285347 DOI: 10.1038/s41380-021-01233-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/25/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases (NDs) are characterized by the aggregation of neurotoxic proteins in the central nervous system. Aberrant protein accumulation in NDs is largely caused by the dysfunction of the two principal protein catabolism pathways, the ubiquitin-proteasome system (UPS), and the autophagy-lysosomal pathway (ALP). The two protein quality control pathways are bridged by ubiquitination, a post-translational modification that can induce protein degradation via both the UPS and the ALP. Perturbed ubiquitination leads to the formation of toxic aggregates and inclusion bodies that are deleterious to neurons. Ubiquitination is promoted by a cascade of ubiquitinating enzymes and counter-regulated by deubiquitinating enzymes (DUBs). As fine-tuning regulators of ubiquitination and protein degradation, DUBs modulate the stability of ND-associated pathogenic proteins including amyloid β protein, Tau, and α-synuclein. Besides, DUBs also influence ND-associated mitophagy, protein secretion, and neuroinflammation. Given the various and critical functions of DUBs in NDs, DUBs may become potential therapeutic targets for NDs.
Collapse
|
26
|
Axtman AD. Characterizing the role of the dark kinome in neurodegenerative disease - A mini review. Biochim Biophys Acta Gen Subj 2021; 1865:130014. [PMID: 34547390 DOI: 10.1016/j.bbagen.2021.130014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/26/2021] [Accepted: 09/14/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Drugs that modulate previously unexplored targets could potentially slow or halt the progression of neurodegenerative diseases. Several candidate proteins lie within the dark kinome, those human kinases that have not been well characterized. Much of the kinome (~80%) remains poorly studied, and these targets likely harbor untapped biological potential. SCOPE OF REVIEW This review highlights the significance of kinases as mediators of aberrant pathways in neurodegeneration and provides examples of published high-quality small molecules that modulate some of these kinases. MAJOR CONCLUSIONS There is a need for continued efforts to develop high-quality chemical tools to illuminate the function of understudied kinases in the brain. Potent and selective small molecules enable accurate pairing of an observed phenotype with a protein target. GENERAL SIGNIFICANCE The examples discussed herein support the premise that validation of therapeutic hypotheses surrounding kinase targets can be accomplished via small molecules and they can serve as the basis for disease-focused drug development campaigns.
Collapse
Affiliation(s)
- Alison D Axtman
- UNC Eshelman School of Pharmacy, Division of Chemical Biology and Medicinal Chemistry, Structural Genomics Consortium, Chapel Hill, NC, USA.
| |
Collapse
|
27
|
Mohamed Asik R, Suganthy N, Aarifa MA, Kumar A, Szigeti K, Mathe D, Gulyás B, Archunan G, Padmanabhan P. Alzheimer's Disease: A Molecular View of β-Amyloid Induced Morbific Events. Biomedicines 2021; 9:biomedicines9091126. [PMID: 34572312 PMCID: PMC8468668 DOI: 10.3390/biomedicines9091126] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/22/2021] [Accepted: 08/27/2021] [Indexed: 12/26/2022] Open
Abstract
Amyloid-β (Aβ) is a dynamic peptide of Alzheimer’s disease (AD) which accelerates the disease progression. At the cell membrane and cell compartments, the amyloid precursor protein (APP) undergoes amyloidogenic cleavage by β- and γ-secretases and engenders the Aβ. In addition, externally produced Aβ gets inside the cells by receptors mediated internalization. An elevated amount of Aβ yields spontaneous aggregation which causes organelles impairment. Aβ stimulates the hyperphosphorylation of tau protein via acceleration by several kinases. Aβ travels to the mitochondria and interacts with its functional complexes, which impairs the mitochondrial function leading to the activation of apoptotic signaling cascade. Aβ disrupts the Ca2+ and protein homeostasis of the endoplasmic reticulum (ER) and Golgi complex (GC) that promotes the organelle stress and inhibits its stress recovery machinery such as unfolded protein response (UPR) and ER-associated degradation (ERAD). At lysosome, Aβ precedes autophagy dysfunction upon interacting with autophagy molecules. Interestingly, Aβ act as a transcription regulator as well as inhibits telomerase activity. Both Aβ and p-tau interaction with neuronal and glial receptors elevate the inflammatory molecules and persuade inflammation. Here, we have expounded the Aβ mediated events in the cells and its cosmopolitan role on neurodegeneration, and the current clinical status of anti-amyloid therapy.
Collapse
Affiliation(s)
- Rajmohamed Mohamed Asik
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; (R.M.A.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
- Department of Animal Science, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India;
| | - Natarajan Suganthy
- Department of Nanoscience and Technology, Alagappa University, Karaikudi 630003, Tamil Nadu, India;
| | - Mohamed Asik Aarifa
- Department of Animal Science, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India;
| | - Arvind Kumar
- Centre for Cellular and Molecular Biology, Hyderabad 500007, Telangana, India;
| | - Krisztián Szigeti
- Department of Biophysics and Radiation Biology, Semmelweis University, 1094 Budapest, Hungary; (K.S.); (D.M.)
- CROmed Translational Research Centers, 1094 Budapest, Hungary
| | - Domokos Mathe
- Department of Biophysics and Radiation Biology, Semmelweis University, 1094 Budapest, Hungary; (K.S.); (D.M.)
- CROmed Translational Research Centers, 1094 Budapest, Hungary
- In Vivo Imaging Advanced Core Facility, Hungarian Center of Excellence for Molecular Medicine (HCEMM), 1094 Budapest, Hungary
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; (R.M.A.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
- Department of Clinical Neuroscience, Karolinska Institute, 17176 Stockholm, Sweden
| | - Govindaraju Archunan
- Department of Animal Science, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India;
- Marudupandiyar College, Thanjavur 613403, Tamil Nadu, India
- Correspondence: (G.A.); (P.P.)
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; (R.M.A.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
- Correspondence: (G.A.); (P.P.)
| |
Collapse
|
28
|
Hole KL, Staniaszek LE, Menon Balan G, Mason JM, Brown JT, Williams RJ. Oral (-)-Epicatechin Inhibits Progressive Tau Pathology in rTg4510 Mice Independent of Direct Actions at GSK3β. Front Neurosci 2021; 15:697319. [PMID: 34220446 PMCID: PMC8244787 DOI: 10.3389/fnins.2021.697319] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/21/2021] [Indexed: 11/23/2022] Open
Abstract
Aggregation of the microtubule-associated protein tau into paired helical filaments (PHFs) and neurofibrillary tangles is a defining characteristic of Alzheimer’s Disease. Various plant polyphenols disrupt tau aggregation in vitro but display poor bioavailability and low potency, challenging their therapeutic translation. We previously reported that oral administration of the flavonoid (−)-epicatechin (EC) reduced Amyloid-β (Aβ) plaque pathology in APP/PS1 transgenic mice. Here, we investigated whether EC impacts on tau pathology, independent of actions on Aβ, using rTg4510 mice expressing P301L mutant tau. 4 and 6.5 months old rTg4510 mice received EC (∼18 mg/day) or vehicle (ethanol) via drinking water for 21 days and the levels of total and phosphorylated tau were assessed. At 4 months, tau appeared as two bands of ∼55 kDa, phosphorylated at Ser262 and Ser396 and was unaffected by exposure to EC. At 6.5 months an additional higher molecular weight form of tau was detected at ∼64 kDa which was phosphorylated at Ser262, Ser396 and additionally at the AT8 sites, indicative of the presence of PHFs. EC consumption reduced the levels of the ∼64 kDa tau species and inhibited phosphorylation at Ser262 and AT8 phosphoepitopes. Regulation of the key tau kinase glycogen synthase kinase 3β (GSK3β) by phosphorylation at Ser9 was not altered by exposure to EC in mice or primary neurons. Furthermore, EC did not significantly inhibit GSK3β activity at physiologically-relevant concentrations in a cell free assay. Therefore, a 21-day intervention with EC inhibits or reverses the development of tau pathology in rTg4510 mice independently of direct inhibition of GSK3β.
Collapse
Affiliation(s)
- Katriona L Hole
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Lydia E Staniaszek
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Gayathri Menon Balan
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Jody M Mason
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| | - Jon T Brown
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, United Kingdom
| | - Robert J Williams
- Department of Biology and Biochemistry, University of Bath, Bath, United Kingdom
| |
Collapse
|
29
|
Khan AA, Khan Z. Comparative host-pathogen protein-protein interaction analysis of recent coronavirus outbreaks and important host targets identification. Brief Bioinform 2021; 22:1206-1214. [PMID: 32914167 PMCID: PMC7546044 DOI: 10.1093/bib/bbaa207] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/13/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022] Open
Abstract
Last two decades have witnessed several global infectious outbreaks. Among these, coronavirus is identified as a prime culprit ranging from its involvement in severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS) to COVID-19. These infections involved in huge healthcare and economic cost incurred globally. Every time, coronavirus improved its infection ability and surprised the medical practitioners and researchers. Currently, COVID-19 is also causing numerous infections and stalled global activities. Global efforts are underway to identify potential viral targets for management of these outbreaks, but significant progress in prevention of these outbreaks is not yet achieved. We explored host-pathogen protein-protein interactions of MERS, SARS and COVID-19, and identified host targets common among all recent coronavirus outbreaks. Further, we tried to understand their potential for management of coronavirus. The common proteins involved in coronavirus host-pathogen interactions indicate their indispensable role in the pathogenesis and therefore targeting these proteins can give strategies to prevent current and future coronavirus outbreaks. Viral variability necessitates development of new therapeutic modalities for every outbreak, in contrast targeting necessary human proteins required by all coronaviruses can provide us a clue to prevent current and future coronavirus outbreaks. We found that targeting FURIN and TMPRSS2 can provide good results due to their common involvement in current and previous outbreaks. We also listed some known molecules against these two targets for their potential drug repurposing evaluation. Although, several recent studies undergoing with targeting these proteins for management of coronavirus, but safety evaluation and risk assessment must be given prime importance while targeting human proteins.
Collapse
Affiliation(s)
- Abdul Arif Khan
- Corresponding author: Abdul Arif Khan, Indian Council of Medical Research-National AIDS Research Institute, Pune Maharashtra 411026, India. Tel: +91 9860530514; E-mail:
| | | |
Collapse
|
30
|
Turab Naqvi AA, Hasan GM, Hassan MI. Targeting Tau Hyperphosphorylation via Kinase Inhibition: Strategy to Address Alzheimer's Disease. Curr Top Med Chem 2021; 20:1059-1073. [PMID: 31903881 DOI: 10.2174/1568026620666200106125910] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/27/2019] [Accepted: 12/16/2019] [Indexed: 01/10/2023]
Abstract
Microtubule-associated protein tau is involved in the tubulin binding leading to microtubule stabilization in neuronal cells which is essential for stabilization of neuron cytoskeleton. The regulation of tau activity is accommodated by several kinases which phosphorylate tau protein on specific sites. In pathological conditions, abnormal activity of tau kinases such as glycogen synthase kinase-3 β (GSK3β), cyclin-dependent kinase 5 (CDK5), c-Jun N-terminal kinases (JNKs), extracellular signal-regulated kinase 1 and 2 (ERK1/2) and microtubule affinity regulating kinase (MARK) lead to tau hyperphosphorylation. Hyperphosphorylation of tau protein leads to aggregation of tau into paired helical filaments like structures which are major constituents of neurofibrillary tangles, a hallmark of Alzheimer's disease. In this review, we discuss various tau protein kinases and their association with tau hyperphosphorylation. We also discuss various strategies and the advancements made in the area of Alzheimer's disease drug development by designing effective and specific inhibitors for such kinases using traditional in vitro/in vivo methods and state of the art in silico techniques.
Collapse
Affiliation(s)
- Ahmad Abu Turab Naqvi
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi - 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj - 11942, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi - 110025, India
| |
Collapse
|
31
|
Dominguez-Meijide A, Vasili E, Outeiro TF. Pharmacological Modulators of Tau Aggregation and Spreading. Brain Sci 2020; 10:E858. [PMID: 33203009 PMCID: PMC7696562 DOI: 10.3390/brainsci10110858] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/25/2022] Open
Abstract
Tauopathies are neurodegenerative disorders characterized by the deposition of aggregates composed of abnormal tau protein in the brain. Additionally, misfolded forms of tau can propagate from cell to cell and throughout the brain. This process is thought to lead to the templated misfolding of the native forms of tau, and thereby, to the formation of newer toxic aggregates, thereby propagating the disease. Therefore, modulation of the processes that lead to tau aggregation and spreading is of utmost importance in the fight against tauopathies. In recent years, several molecules have been developed for the modulation of tau aggregation and spreading. In this review, we discuss the processes of tau aggregation and spreading and highlight selected chemicals developed for the modulation of these processes, their usefulness, and putative mechanisms of action. Ultimately, a stronger understanding of the molecular mechanisms involved, and the properties of the substances developed to modulate them, will lead to the development of safer and better strategies for the treatment of tauopathies.
Collapse
Affiliation(s)
- Antonio Dominguez-Meijide
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, 37073 Goettingen, Germany; (A.D.-M.); (E.V.)
- Laboratory of Neuroanatomy and Experimental Neurology, Dept. of Morphological Sciences, CIMUS, IDIS, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Eftychia Vasili
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, 37073 Goettingen, Germany; (A.D.-M.); (E.V.)
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, 37073 Goettingen, Germany; (A.D.-M.); (E.V.)
- Max Planck Institute for Experimental Medicine, 37075 Goettingen, Germany
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Framlington Place, Newcastle Upon Tyne NE2 4HH, UK
| |
Collapse
|
32
|
Jęśko H, Cieślik M, Gromadzka G, Adamczyk A. Dysfunctional proteins in neuropsychiatric disorders: From neurodegeneration to autism spectrum disorders. Neurochem Int 2020; 141:104853. [PMID: 32980494 DOI: 10.1016/j.neuint.2020.104853] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/05/2020] [Accepted: 09/22/2020] [Indexed: 02/06/2023]
Abstract
Despite fundamental differences in disease course and outcomes, neurodevelopmental (autism spectrum disorders - ASD) and neurodegenerative disorders (Alzheimer's disease - AD and Parkinson's disease - PD) present surprising, common traits in their molecular pathomechanisms. Uncontrolled oligomerization and aggregation of amyloid β (Aβ), microtubule-associated protein (MAP) tau, or α-synuclein (α-syn) contribute to synaptic impairment and the ensuing neuronal death in both AD and PD. Likewise, the pathogenesis of ASD may be attributed, at least in part, to synaptic dysfunction; attention has also been recently paid to irregularities in the metabolism and function of the Aβ precursor protein (APP), tau, or α-syn. Commonly affected elements include signaling pathways that regulate cellular metabolism and survival such as insulin/insulin-like growth factor (IGF) - PI3 kinase - Akt - mammalian target of rapamycin (mTOR), and a number of key synaptic proteins critically involved in neuronal communication. Understanding how these shared pathomechanism elements operate in different conditions may help identify common targets and therapeutic approaches.
Collapse
Affiliation(s)
- Henryk Jęśko
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| | - Magdalena Cieślik
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| | - Grażyna Gromadzka
- Cardinal Stefan Wyszynski University, Faculty of Medicine. Collegium Medicum, Wóycickiego 1/3, 01-938, Warsaw, Poland.
| | - Agata Adamczyk
- Department of Cellular Signalling, M. Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106, Warsaw, Poland.
| |
Collapse
|
33
|
Miller JB, Ward E, Staley LA, Stevens J, Teerlink CC, Tavana JP, Cloward M, Page M, Dayton L, Cannon-Albright LA, Kauwe JSK. Identification and genomic analysis of pedigrees with exceptional longevity identifies candidate rare variants. Neurobiol Dis 2020; 143:104972. [PMID: 32574725 PMCID: PMC7461696 DOI: 10.1016/j.nbd.2020.104972] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/05/2020] [Accepted: 06/12/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Longevity as a phenotype entails living longer than average and typically includes living without chronic age-related diseases. Recently, several common genetic components to longevity have been identified. This study aims to identify additional genetic variants associated with longevity using unique and powerful analyses of pedigrees with a statistical excess of healthy elderly individuals identified in the Utah Population Database (UPDB). METHODS From an existing biorepository of Utah pedigrees, six independent cousin pairs were selected from four extended pedigrees that exhibited an excess of healthy elderly individuals; whole exome sequencing (WES) was performed on two elderly individuals from each pedigree who were either first cousins or first cousins once removed. Rare (<.01 population frequency) variants shared by at least one elderly cousin pair in a region likely to be identical by descent were identified as candidates. Ingenuity Variant Analysis was used to prioritize putative causal variants based on quality control, frequency, and gain or loss of function. The variant frequency was compared in healthy cohorts and in an Alzheimer's disease cohort. Remaining variants were filtered based on their presence in genes reported to have an effect on the aging process, aging of cells, or the longevity process. Validation of these candidate variants included tests of segregation on other elderly relatives. RESULTS Fifteen rare candidate genetic variants spanning 17 genes shared within cousins were identified as having passed prioritization criteria. Of those variants, six were present in genes that are known or predicted to affect the aging process: rs78408340 (PAM), rs112892337 (ZFAT), rs61737629 (ESPL1), rs141903485 (CEBPE), rs144369314 (UTP4), and rs61753103 (NUP88 and RABEP1). ESPL1 rs61737629 and CEBPE rs141903485 show additional evidence of segregation with longevity in expanded pedigree analyses (p-values = .001 and .0001, respectively). DISCUSSION This unique pedigree analysis efficiently identified several novel rare candidate variants that may affect the aging process and added support to seven genes that likely contribute to longevity. Further analyses showed evidence for segregation for two rare variants, ESPL1 rs61737629 and CEBPE rs141903485, in the original longevity pedigrees in which they were initially observed. These candidate genes and variants warrant further investigation.
Collapse
Affiliation(s)
- Justin B Miller
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Elizabeth Ward
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Lyndsay A Staley
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Jeffrey Stevens
- Genetic Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Craig C Teerlink
- Genetic Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Justina P Tavana
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Matthew Cloward
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Madeline Page
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Louisa Dayton
- Department of Biology, Brigham Young University, Provo, UT 84602, USA
| | - Lisa A Cannon-Albright
- Genetic Epidemiology, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - John S K Kauwe
- Department of Biology, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
34
|
Naqvi AAT, Jairajpuri DS, Noman OMA, Hussain A, Islam A, Ahmad F, Alajmi MF, Hassan MI. Evaluation of pyrazolopyrimidine derivatives as microtubule affinity regulating kinase 4 inhibitors: Towards therapeutic management of Alzheimer’s disease. J Biomol Struct Dyn 2019; 38:3892-3907. [DOI: 10.1080/07391102.2019.1666745] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Ahmad Abu Turab Naqvi
- Center for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, New Delhi, India
| | - Deeba Shamim Jairajpuri
- Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Omar Mohammed Ali Noman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Asimul Islam
- Center for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, New Delhi, India
| | - Faizan Ahmad
- Center for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, New Delhi, India
| | - Mohammed F. Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Md. Imtaiyaz Hassan
- Center for Interdisciplinary Research in Basic Science, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
35
|
Ahrari S, Mogharrab N, Navapour L. Structure and dynamics of inactive and active MARK4: conformational switching through the activation process. J Biomol Struct Dyn 2019; 38:2468-2481. [DOI: 10.1080/07391102.2019.1655479] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Sajjad Ahrari
- Biophysics and Computational Biology Laboratory (BCBL), Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran
| | - Navid Mogharrab
- Biophysics and Computational Biology Laboratory (BCBL), Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran
| | - Leila Navapour
- Biophysics and Computational Biology Laboratory (BCBL), Department of Biology, College of Sciences, Shiraz University, Shiraz, Iran
| |
Collapse
|
36
|
Hayden EY, Putman J, Nunez S, Shin WS, Oberoi M, Charreton M, Dutta S, Li Z, Komuro Y, Joy MT, Bitan G, MacKenzie-Graham A, Jiang L, Hinman JD. Ischemic axonal injury up-regulates MARK4 in cortical neurons and primes tau phosphorylation and aggregation. Acta Neuropathol Commun 2019; 7:135. [PMID: 31429800 PMCID: PMC6700776 DOI: 10.1186/s40478-019-0783-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 08/01/2019] [Indexed: 12/11/2022] Open
Abstract
Ischemic injury to white matter tracts is increasingly recognized to play a key role in age-related cognitive decline, vascular dementia, and Alzheimer’s disease. Knowledge of the effects of ischemic axonal injury on cortical neurons is limited yet critical to identifying molecular pathways that link neurodegeneration and ischemia. Using a mouse model of subcortical white matter ischemic injury coupled with retrograde neuronal tracing, we employed magnetic affinity cell sorting with fluorescence-activated cell sorting to capture layer-specific cortical neurons and performed RNA-sequencing. With this approach, we identified a role for microtubule reorganization within stroke-injured neurons acting through the regulation of tau. We find that subcortical stroke-injured Layer 5 cortical neurons up-regulate the microtubule affinity-regulating kinase, Mark4, in response to axonal injury. Stroke-induced up-regulation of Mark4 is associated with selective remodeling of the apical dendrite after stroke and the phosphorylation of tau in vivo. In a cell-based tau biosensor assay, Mark4 promotes the aggregation of human tau in vitro. Increased expression of Mark4 after ischemic axonal injury in deep layer cortical neurons provides new evidence for synergism between axonal and neurodegenerative pathologies by priming of tau phosphorylation and aggregation.
Collapse
|
37
|
Alqaeisoom N, Qian C, Arachchige D, Colvin RA, Holub JM. Inhibiting Phosphorylation of Tau (τ) Proteins at Ser262 Using Peptide-Based R1 Domain Mimetics. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-018-9689-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
38
|
Mulcahy MJ, Paulo JA, Hawrot E. Proteomic Investigation of Murine Neuronal α7-Nicotinic Acetylcholine Receptor Interacting Proteins. J Proteome Res 2018; 17:3959-3975. [PMID: 30285449 DOI: 10.1021/acs.jproteome.8b00618] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The α7-nicotinic acetylcholine receptor (α7-nAChR) is a ligand-gated ion channel that is expressed widely in vertebrates and is the principal high-affinity α-bungarotoxin (α-bgtx) binding protein in the mammalian CNS. α7-nAChRs associate with proteins that can modulate its properties. The α7-nAChR interactome is the summation of proteins interacting or associating with α7-nAChRs in a protein complex. To identify an α7-nAChR interactome in neural tissue, we isolated α-bgtx-affinity protein complexes from wild-type and α7-nAChR knockout (α7 KO) mouse whole brain tissue homogenates using α-bgtx-affinity beads. Affinity precipitated proteins were trypsinized and analyzed with an Orbitrap Fusion mass spectrometer. Proteins isolated with the α7-nAChR specific ligand, α-bgtx, were determined to be α7-nAChR associated proteins. The α7-nAChR subunit and 120 additional proteins were identified. Additionally, 369 proteins were identified as binding to α-bgtx in the absence of α7-nAChR expression, thereby identifying nonspecific proteins for α7-nAChR investigations using α-bgtx enrichment. These results expand on our previous investigations of α7-nAChR interacting proteins using α-bgtx-affinity bead isolation by controlling for differences between α7-nAChR and α-bgtx-specific proteins, developing an improved protein isolation methodology, and incorporating the latest technology in mass spectrometry. The α7-nAChR interactome identified in this study includes proteins associated with the expression, localization, function, or modulation of α7-nAChRs, and it provides a foundation for future studies to elucidate how these interactions contribute to human disease.
Collapse
Affiliation(s)
- Matthew J Mulcahy
- Division of Biology and Biological Engineering , California Institute of Technology , 1200 East California Boulevard , Pasadena , California 91125-2900 , United States.,Department of Molecular Pharmacology, Physiology and Biotechnology , Brown University , Providence , Rhode Island 02912 , United States
| | - Joao A Paulo
- Department of Cell Biology , Harvard Medical School , 240 Longwood Avenue , Boston , Massachusetts 02115 , United States
| | - Edward Hawrot
- Department of Molecular Pharmacology, Physiology and Biotechnology , Brown University , Providence , Rhode Island 02912 , United States
| |
Collapse
|
39
|
Correani V, Di Francesco L, Mignogna G, Fabrizi C, Leone S, Giorgi A, Passeri A, Casata R, Fumagalli L, Maras B, Schininà ME. Plasma Membrane Protein Profiling in Beta-Amyloid-Treated Microglia Cell Line. Proteomics 2017; 17. [PMID: 28815942 DOI: 10.1002/pmic.201600439] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 08/01/2017] [Indexed: 01/13/2023]
Abstract
In the responsiveness of microglia to toxic stimuli, plasma membrane proteins play a key role. In this study we treated with a synthetic beta amyloid peptide murine microglial cells metabolically differently labelled with stable isotope amino acids (SILAC). The plasma membrane was selectively enriched by a multi-stage aqueous two-phase partition system. We were able to identify by 1D-LC-MS/MS analyses 1577 proteins, most of them are plasma membrane proteins according to the Gene Ontology annotation. An unchanged level of amyloid receptors in this data set suggests that microglia preserve their responsiveness capability to the environment even after 24-h challenge with amyloid peptides. On the other hand, 14 proteins were observed to change their plasma membrane abundance to a statistically significant extent. Among these, we proposed as reliable biomarkers of the inflammatory microglia phenotype in AD damaged tissues MAP/microtubule affinity-regulating kinase 3 (MARK3), Interferon-induced transmembrane protein 3 (IFITM3), Annexins A5 and A7 (ANXA5, ANXA7) and Neuropilin-1 (NRP1), all proteins known to be involved in the inflammation processes and in microtubule network assembly rate.
Collapse
Affiliation(s)
- Virginia Correani
- Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Rome, Italy
| | - Laura Di Francesco
- Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Rome, Italy
| | - Giuseppina Mignogna
- Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Rome, Italy
| | - Cinzia Fabrizi
- Dipartimento di Scienze Anatomiche Istologiche, Medico-Legali e dell'Apparato Locomotore, Sapienza Università di Roma, Rome, Italy
| | - Stefano Leone
- Dipartimento di Scienze, Università Roma Tre, Rome, Italy
| | - Alessandra Giorgi
- Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Rome, Italy
| | - Alessia Passeri
- Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Rome, Italy
| | - Roberto Casata
- Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Rome, Italy
| | - Lorenzo Fumagalli
- Dipartimento di Scienze Anatomiche Istologiche, Medico-Legali e dell'Apparato Locomotore, Sapienza Università di Roma, Rome, Italy
| | - Bruno Maras
- Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Rome, Italy
| | - M Eugenia Schininà
- Dipartimento di Scienze Biochimiche, Sapienza Università di Roma, Rome, Italy
| |
Collapse
|
40
|
Annadurai N, Agrawal K, Džubák P, Hajdúch M, Das V. Microtubule affinity-regulating kinases are potential druggable targets for Alzheimer's disease. Cell Mol Life Sci 2017; 74:4159-4169. [PMID: 28634681 PMCID: PMC11107647 DOI: 10.1007/s00018-017-2574-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 06/13/2017] [Accepted: 06/15/2017] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that affects normal functions of the brain. Currently, AD is one of the leading causes of death in developed countries and the only one of the top ten diseases without a means to prevent, cure, or significantly slow down its progression. Therefore, newer therapeutic concepts are urgently needed to improve survival and the quality of life of AD patients. Microtubule affinity-regulating kinases (MARKs) regulate tau-microtubule binding and play a crucial role in neurons. However, their role in hyperphosphorylation of tau makes them potential druggable target for AD therapy. Despite the relevance of MARKs in AD pathogenesis, only a few small molecules are known to have anti-MARK activity and not much has been done to progress these compounds into therapeutic candidates. But given the diverse role of MARKs, the specificity of novel inhibitors is imperative for their successful translation from bench to bedside. In this regard, a recent co-crystal structure of MARK4 in association with a pyrazolopyrimidine-based inhibitor offers a potential scaffold for the development of more specific MARK inhibitors. In this manuscript, we review the biological role of MARKs in health and disease, and draw attention to the largely unexplored area of MARK inhibitors for AD.
Collapse
Affiliation(s)
- Narendran Annadurai
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900, Olomouc, Czech Republic
| | - Khushboo Agrawal
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900, Olomouc, Czech Republic
| | - Petr Džubák
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900, Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900, Olomouc, Czech Republic
| | - Viswanath Das
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900, Olomouc, Czech Republic.
| |
Collapse
|
41
|
Sandí MJ, Marshall CB, Balan M, Coyaud É, Zhou M, Monson DM, Ishiyama N, Chandrakumar AA, La Rose J, Couzens AL, Gingras AC, Raught B, Xu W, Ikura M, Morrison DK, Rottapel R. MARK3-mediated phosphorylation of ARHGEF2 couples microtubules to the actin cytoskeleton to establish cell polarity. Sci Signal 2017; 10:10/503/eaan3286. [PMID: 29089450 DOI: 10.1126/scisignal.aan3286] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The PAR-1-MARK pathway controls cell polarity through the phosphorylation of microtubule-associated proteins. Rho-Rac guanine nucleotide exchange factor 2 (ARHGEF2), which activates Ras homolog family member A (RHOA), is anchored to the microtubule network and sequestered in an inhibited state through binding to dynein light chain Tctex-1 type 1 (DYNLT1). We showed in mammalian cells that liver kinase B1 (LKB1) activated the microtubule affinity-regulating kinase 3 (MARK3), which in turn phosphorylated ARHGEF2 at Ser151 This modification disrupted the interaction between ARHGEF2 and DYNLT1 by generating a 14-3-3 binding site in ARHGEF2, thus causing ARHGEF2 to dissociate from microtubules. Phosphorylation of ARHGEF2 by MARK3 stimulated RHOA activation and the formation of stress fibers and focal adhesions, and was required for organized cellular architecture in three-dimensional culture. Protein phosphatase 2A (PP2A) dephosphorylated Ser151 in ARHGEF2 to restore the inhibited state. Thus, we have identified a regulatory switch controlled by MARK3 that couples microtubules to the actin cytoskeleton to establish epithelial cell polarity through ARHGEF2.
Collapse
Affiliation(s)
- María-José Sandí
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Princess Margaret Cancer Research Tower, Toronto, Ontario M5G 1L7, Canada
| | - Christopher B Marshall
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Princess Margaret Cancer Research Tower, Toronto, Ontario M5G 1L7, Canada
| | - Marc Balan
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Princess Margaret Cancer Research Tower, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Étienne Coyaud
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Princess Margaret Cancer Research Tower, Toronto, Ontario M5G 1L7, Canada
| | - Ming Zhou
- Center for Cancer Research, National Cancer Institute at Frederick, P.O. Box B, Frederick, MD 21702, USA
| | - Daniel M Monson
- Center for Cancer Research, National Cancer Institute at Frederick, P.O. Box B, Frederick, MD 21702, USA
| | - Noboru Ishiyama
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Princess Margaret Cancer Research Tower, Toronto, Ontario M5G 1L7, Canada
| | - Arun A Chandrakumar
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Princess Margaret Cancer Research Tower, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - José La Rose
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Princess Margaret Cancer Research Tower, Toronto, Ontario M5G 1L7, Canada
| | - Amber L Couzens
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, Ontario M5G 1X5, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Brian Raught
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Princess Margaret Cancer Research Tower, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Wei Xu
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada.,Department of Biostatistics, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Mitsuhiko Ikura
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Princess Margaret Cancer Research Tower, Toronto, Ontario M5G 1L7, Canada.,Department of Medical Biophysics, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada
| | - Deborah K Morrison
- Center for Cancer Research, National Cancer Institute at Frederick, P.O. Box B, Frederick, MD 21702, USA
| | - Robert Rottapel
- Princess Margaret Cancer Centre, University Health Network, 101 College Street, Princess Margaret Cancer Research Tower, Toronto, Ontario M5G 1L7, Canada. .,Department of Medical Biophysics, University of Toronto, 1 King's College Circle, Toronto, Ontario M5S 1A8, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada.,Division of Rheumatology, St. Michael's Hospital, 30 Bond Street, Toronto, Ontario M5B 1W8, Canada
| |
Collapse
|
42
|
Ebai T, Souza de Oliveira FM, Löf L, Wik L, Schweiger C, Larsson A, Keilholtz U, Haybaeck J, Landegren U, Kamali-Moghaddam M. Analytically Sensitive Protein Detection in Microtiter Plates by Proximity Ligation with Rolling Circle Amplification. Clin Chem 2017; 63:1497-1505. [DOI: 10.1373/clinchem.2017.271833] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Accepted: 05/04/2017] [Indexed: 12/31/2022]
Abstract
Abstract
BACKGROUND
Detecting proteins at low concentrations in plasma is crucial for early diagnosis. Current techniques in clinical routine, such as sandwich ELISA, provide sensitive protein detection because of a dependence on target recognition by pairs of antibodies, but detection of still lower protein concentrations is often called for. Proximity ligation assay with rolling circle amplification (PLARCA) is a modified proximity ligation assay (PLA) for analytically specific and sensitive protein detection via binding of target proteins by 3 antibodies, and signal amplification via rolling circle amplification (RCA) in microtiter wells, easily adapted to instrumentation in use in hospitals.
METHODS
Proteins captured by immobilized antibodies were detected using a pair of oligonucleotide-conjugated antibodies. Upon target recognition these PLA probes guided oligonucleotide ligation, followed by amplification via RCA of circular DNA strands that formed in the reaction. The RCA products were detected by horseradish peroxidase-labeled oligonucleotides to generate colorimetric reaction products with readout in an absorbance microplate reader.
RESULTS
We compared detection of interleukin (IL)-4, IL-6, IL-8, p53, and growth differentiation factor 15 (GDF-15) by PLARCA and conventional sandwich ELISA or immuno-RCA. PLARCA detected lower concentrations of proteins and exhibited a broader dynamic range compared to ELISA and iRCA using the same antibodies. IL-4 and IL-6 were detected in clinical samples at femtomolar concentrations, considerably lower than for ELISA.
CONCLUSIONS
PLARCA offers detection of lower protein levels and increased dynamic ranges compared to ELISA. The PLARCA procedure may be adapted to routine instrumentation available in hospitals and research laboratories.
Collapse
Affiliation(s)
- Tonge Ebai
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Liza Löf
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Lotta Wik
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Caroline Schweiger
- Charité Comprehensive Cancer Center, University of Berlin, Berlin, Germany
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Anders Larsson
- Department of Medical Sciences, Biochemical Structure and Function, Uppsala University, Uppsala, Sweden
| | - Ulrich Keilholtz
- Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Johannes Haybaeck
- Charité Comprehensive Cancer Center, University of Berlin, Berlin, Germany
- Department of Pathology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Ulf Landegren
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Masood Kamali-Moghaddam
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
43
|
MARK4 regulates NLRP3 positioning and inflammasome activation through a microtubule-dependent mechanism. Nat Commun 2017; 8:15986. [PMID: 28656979 PMCID: PMC5493753 DOI: 10.1038/ncomms15986] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/17/2017] [Indexed: 12/17/2022] Open
Abstract
Excessive activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome is involved in many chronic inflammatory diseases, including cardiovascular and Alzheimer's disease. Here we show that microtubule-affinity regulating kinase 4 (MARK4) binds to NLRP3 and drives it to the microtubule-organizing centre, enabling the formation of one large inflammasome speck complex within a single cell. MARK4 knockdown or knockout, or disruption of MARK4-NLRP3 interaction, impairs NLRP3 spatial arrangement and limits inflammasome activation. Our results demonstrate how an evolutionarily conserved protein involved in the regulation of microtubule dynamics orchestrates NLRP3 inflammasome activation by controlling its transport to optimal activation sites, and identify a targetable function for MARK4 in the control of innate immunity.
Collapse
|
44
|
Canu N, Pagano I, La Rosa LR, Pellegrino M, Ciotti MT, Mercanti D, Moretti F, Sposato V, Triaca V, Petrella C, Maruyama IN, Levi A, Calissano P. Association of TrkA and APP Is Promoted by NGF and Reduced by Cell Death-Promoting Agents. Front Mol Neurosci 2017; 10:15. [PMID: 28197073 PMCID: PMC5281621 DOI: 10.3389/fnmol.2017.00015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 01/11/2017] [Indexed: 12/31/2022] Open
Abstract
The amyloid precursor protein (APP) interacts with the tropomyosin receptor kinase A (TrkA) in normal rat, mouse, and human brain tissue but not in Alzheimer's disease (AD) brain tissue. However, it has not been reported whether the two proteins interact directly, and if so, which domains are involved. Clarifying these points will increase our understanding of the role and regulation of the TrkA/APP interaction in normal brain functioning as well as in AD. Here we addressed these questions using bimolecular fluorescence complementation (BiFC) and the proximity ligation assay (PLA). We demonstrated that exogenously expressed APP and TrkA associate through their juxtamembrane/transmembrane domains, to form a complex that localizes mainly to the plasma membrane, endoplasmic reticulum (ER) and Golgi. Formation of the complex was inhibited by p75NTR, ShcC and Mint-2. Importantly, we demonstrated that the association between endogenous APP and TrkA in primary septal neurons were modified by NGF, or by drugs that either inhibit ER-to-Golgi transport or perturb microtubules and microfilaments. Interestingly, several agents that induce cell death [amyloid β (Aβ)-peptide, staurosporine and rapamycin], albeit via different mechanisms, all caused dissociation of APP/TrkA complexes and increased production of C-terminal fragment (β-CTF) APP fragment. These findings open new perspectives for investigating the interplay between these proteins during neurodegeneration and AD.
Collapse
Affiliation(s)
- Nadia Canu
- Department of System Medicine, University of Rome "Tor Vergata"Rome, Italy; Institute of Cellular Biology and Neurobiology, National Council of Research of RomeRome, Italy
| | - Ilaria Pagano
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Luca Rosario La Rosa
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Marsha Pellegrino
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Maria Teresa Ciotti
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Delio Mercanti
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Fabiola Moretti
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Valentina Sposato
- Institute of Cellular Biology and Neurobiology, National Council of Research of RomeRome, Italy; European Brain Research InstituteRome, Italy
| | - Viviana Triaca
- Institute of Cellular Biology and Neurobiology, National Council of Research of RomeRome, Italy; European Brain Research InstituteRome, Italy
| | - Carla Petrella
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Ichiro N Maruyama
- Information Processing Biology Unit, Okinawa Institute of Science and Technology Graduate University Okinawa, Japan
| | - Andrea Levi
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Pietro Calissano
- European Brain Research InstituteRome, Italy; Institute of Cellular Biology and Neurobiology, National Council of Research of RomeRome, Italy
| |
Collapse
|
45
|
Gender-Specific Expression of Ubiquitin-Specific Peptidase 9 Modulates Tau Expression and Phosphorylation: Possible Implications for Tauopathies. Mol Neurobiol 2016; 54:7979-7993. [PMID: 27878758 PMCID: PMC5684262 DOI: 10.1007/s12035-016-0299-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 11/14/2016] [Indexed: 01/05/2023]
Abstract
Public transcriptomic studies have shown that several genes display pronounced gender differences in their expression in the human brain, which may influence the manifestations and risk for neuronal disorders. Here, we apply a transcriptome-wide analysis to discover genes with gender-specific expression and significant alterations in public postmortem brain tissue from Alzheimer’s disease (AD) patients compared to controls. We identify the sex-linked ubiquitin-specific peptidase 9 (USP9) as an outstanding candidate gene with highly significant expression differences between the genders and male-specific underexpression in AD. Since previous studies have shown that USP9 can modulate the phosphorylation of the AD-associated protein MAPT, we investigate functional associations between USP9 and MAPT in further detail. After observing a high positive correlation between the expression of USP9 and MAPT in the public transcriptomics data, we show that USP9 knockdown results in significantly decreased MAPT expression in a DU145 cell culture model and a concentration-dependent decrease for the MAPT orthologs mapta and maptb in a zebrafish model. From the analysis of microarray and qRT-PCR experiments for the knockdown in DU145 cells and prior knowledge from the literature, we derive a data-congruent model for a USP9-dependent regulatory mechanism modulating MAPT expression via BACH1 and SMAD4. Overall, the analyses suggest USP9 may contribute to molecular gender differences observed in tauopathies and provide a new target for intervention strategies to modulate MAPT expression.
Collapse
|
46
|
Méphon-Gaspard A, Boca M, Pioche-Durieu C, Desforges B, Burgo A, Hamon L, Piétrement O, Pastré D. Role of tau in the spatial organization of axonal microtubules: keeping parallel microtubules evenly distributed despite macromolecular crowding. Cell Mol Life Sci 2016; 73:3745-60. [PMID: 27076215 PMCID: PMC5002045 DOI: 10.1007/s00018-016-2216-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 03/24/2016] [Accepted: 04/01/2016] [Indexed: 02/07/2023]
Abstract
Opposing views have been proposed regarding the role of tau, the principal microtubule-associated protein in axons. On the one hand, tau forms cross-bridges at the interface between microtubules and induces microtubule bundling in neurons. On the other hand, tau is also considered a polymer brush which efficiently separates microtubules. In mature axons, microtubules are indeed arranged in parallel arrays and are well separated from each other. To reconcile these views, we developed a mechanistic model based on in vitro and cellular approaches combined to analytical and numerical analyses. The results indicate that tau forms long-range cross-bridges between microtubules under macromolecular crowding conditions. Tau cross-bridges prevent the redistribution of tau away from the interface between microtubules, which would have occurred in the polymer brush model. Consequently, the short-range attractive force between microtubules induced by macromolecular crowding is avoided and thus microtubules remain well separated from each other. Interestingly, in this unified model, tau diffusion on microtubules enables to keep microtubules evenly distributed in axonal sections at low tau levels.
Collapse
Affiliation(s)
- Alix Méphon-Gaspard
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1204, Université Evry-Val d'Essonne, Evry, 91025, France
| | - Mirela Boca
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1204, Université Evry-Val d'Essonne, Evry, 91025, France
| | - Catherine Pioche-Durieu
- UMR 8126, CNRS, Gustave Roussy Université Paris Sud, Université Paris-Saclay, Villejuif, 94805, France
| | - Bénédicte Desforges
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1204, Université Evry-Val d'Essonne, Evry, 91025, France
| | - Andrea Burgo
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1204, Université Evry-Val d'Essonne, Evry, 91025, France
| | - Loic Hamon
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1204, Université Evry-Val d'Essonne, Evry, 91025, France
| | - Olivier Piétrement
- UMR 8126, CNRS, Gustave Roussy Université Paris Sud, Université Paris-Saclay, Villejuif, 94805, France
| | - David Pastré
- Institut National de la Santé et de la Recherche Médicale (INSERM), UMR1204, Université Evry-Val d'Essonne, Evry, 91025, France.
| |
Collapse
|
47
|
Tau phosphorylation at Alzheimer's disease-related Ser356 contributes to tau stabilization when PAR-1/MARK activity is elevated. Biochem Biophys Res Commun 2016; 478:929-34. [PMID: 27520376 DOI: 10.1016/j.bbrc.2016.08.053] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 08/08/2016] [Indexed: 02/08/2023]
Abstract
Abnormal phosphorylation of the microtubule-associated protein tau is observed in many neurodegenerative diseases, including Alzheimer's disease (AD). AD-related phosphorylation of two tau residues, Ser262 and Ser356, by PAR-1/MARK stabilizes tau in the initial phase of mismetabolism, leading to subsequent phosphorylation events, accumulation, and toxicity. However, the relative contribution of phosphorylation at each of these sites to tau stabilization has not yet been elucidated. In a Drosophila model of human tau toxicity, we found that tau was phosphorylated at Ser262, but not at Ser356, and that blocking Ser262 phosphorylation decreased total tau levels. By contrast, when PAR-1 was co-overexpressed with tau, tau was hyperphosphorylated at both Ser262 and Ser356. Under these conditions, the protein levels of tau were significantly elevated, and prevention of tau phosphorylation at both residues was necessary to completely suppress this elevation. These results suggest that tau phosphorylation at Ser262 plays the predominant role in tau stabilization when PAR-1/MARK activity is normal, whereas Ser356 phosphorylation begins to contribute to this process when PAR-1/MARK activity is abnormally elevated, as in diseased brains.
Collapse
|
48
|
Cheng Q, Wållberg H, Grafström J, Lu L, Thorell JO, Hägg Olofsson M, Linder S, Johansson K, Tegnebratt T, Arnér ESJ, Stone-Elander S, Ahlzén HSM, Ståhl S. Preclinical PET imaging of EGFR levels: pairing a targeting with a non-targeting Sel-tagged Affibody-based tracer to estimate the specific uptake. EJNMMI Res 2016; 6:58. [PMID: 27388754 PMCID: PMC4936982 DOI: 10.1186/s13550-016-0213-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 06/28/2016] [Indexed: 01/09/2023] Open
Abstract
Background Though overexpression of epidermal growth factor receptor (EGFR) in several forms of cancer is considered to be an important prognostic biomarker related to poor prognosis, clear correlations between biomarker assays and patient management have been difficult to establish. Here, we utilize a targeting directly followed by a non-targeting tracer-based positron emission tomography (PET) method to examine some of the aspects of determining specific EGFR binding in tumors. Methods The EGFR-binding Affibody molecule ZEGFR:2377 and its size-matched non-binding control ZTaq:3638 were recombinantly fused with a C-terminal selenocysteine-containing Sel-tag (ZEGFR:2377-ST and ZTaq:3638-ST). The proteins were site-specifically labeled with DyLight488 for flow cytometry and ex vivo tissue analyses or with 11C for in vivo PET studies. Kinetic scans with the 11C-labeled proteins were performed in healthy mice and in mice bearing xenografts from human FaDu (squamous cell carcinoma) and A431 (epidermoid carcinoma) cell lines. Changes in tracer uptake in A431 xenografts over time were also monitored, followed by ex vivo proximity ligation assays (PLA) of EGFR expressions. Results Flow cytometry and ex vivo tissue analyses confirmed EGFR targeting by ZEGFR:2377-ST-DyLight488. [Methyl-11C]-labeled ZEGFR:2377-ST-CH3 and ZTaq:3638-ST-CH3 showed similar distributions in vivo, except for notably higher concentrations of the former in particularly the liver and the blood. [Methyl-11C]-ZEGFR:2377-ST-CH3 successfully visualized FaDu and A431 xenografts with moderate and high EGFR expression levels, respectively. However, in FaDu tumors, the non-specific uptake was large and sometimes equally large, illustrating the importance of proper controls. In the A431 group observed longitudinally, non-specific uptake remained at same level over the observation period. Specific uptake increased with tumor size, but changes varied widely over time in individual tumors. Total (membranous and cytoplasmic) EGFR in excised sections increased with tumor growth. There was no positive correlation between total EGFR and specific tracer uptake, which, since ZEGFR:2377 binds extracellularly and is slowly internalized, indicates a discordance between available membranous and total EGFR expression levels. Conclusions Same-day in vivo dual tracer imaging enabled by the Sel-tag technology and 11C-labeling provides a method to non-invasively monitor membrane-localized EGFR as well as factors affecting non-specific uptake of the PET ligand. Electronic supplementary material The online version of this article (doi:10.1186/s13550-016-0213-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qing Cheng
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Helena Wållberg
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Grafström
- Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden
| | - Li Lu
- Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden.,Karolinska Experimental Research and Imaging Center, Department of Comparative Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Jan-Olov Thorell
- Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden.,Neuroradiology Department, R3:00, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Maria Hägg Olofsson
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Stig Linder
- Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Katarina Johansson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Tetyana Tegnebratt
- Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden.,Neuroradiology Department, R3:00, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - Elias S J Arnér
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Sharon Stone-Elander
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden. .,Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden. .,Neuroradiology Department, R3:00, Karolinska University Hospital, SE-17176, Stockholm, Sweden.
| | | | - Stefan Ståhl
- Division of Protein Technology, School of Biotechnology, Royal Institute of Technology, Stockholm, Sweden
| | | |
Collapse
|
49
|
Li M, Quan C, Toth R, Campbell DG, MacKintosh C, Wang HY, Chen S. Fasting and Systemic Insulin Signaling Regulate Phosphorylation of Brain Proteins That Modulate Cell Morphology and Link to Neurological Disorders. J Biol Chem 2015; 290:30030-41. [PMID: 26499801 PMCID: PMC4705965 DOI: 10.1074/jbc.m115.668103] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Indexed: 12/20/2022] Open
Abstract
Diabetes is strongly associated with cognitive decline, but the molecular reasons are unknown. We found that fasting and peripheral insulin promote phosphorylation and dephosphorylation, respectively, of specific residues on brain proteins including cytoskeletal regulators such as slit-robo GTPase-activating protein 3 (srGAP3) and microtubule affinity-regulating protein kinases (MARKs), in which deficiency or dysregulation is linked to neurological disorders. Fasting activates protein kinase A (PKA) but not PKB/Akt signaling in the brain, and PKA can phosphorylate the purified srGAP3. The phosphorylation of srGAP3 and MARKs were increased when PKA signaling was activated in primary neurons. Knockdown of PKA decreased the phosphorylation of srGAP3. Furthermore, WAVE1, a protein kinase A-anchoring protein, formed a complex with srGAP3 and PKA in the brain of fasted mice to facilitate the phosphorylation of srGAP3 by PKA. Although brain cells have insulin receptors, our findings are inconsistent with the down-regulation of phosphorylation of target proteins being mediated by insulin signaling within the brain. Rather, our findings infer that systemic insulin, through a yet unknown mechanism, inhibits PKA or protein kinase(s) with similar specificity and/or activates an unknown phosphatase in the brain. Ser858 of srGAP3 was identified as a key regulatory residue in which phosphorylation by PKA enhanced the GAP activity of srGAP3 toward its substrate, Rac1, in cells, thereby inhibiting the action of this GTPase in cytoskeletal regulation. Our findings reveal novel mechanisms linking peripheral insulin sensitivity with cytoskeletal remodeling in neurons, which may help to explain the association of diabetes with neurological disorders such as Alzheimer disease.
Collapse
Affiliation(s)
- Min Li
- From the State Key Laboratory of Pharmaceutical Biotechnology and Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Chao Quan
- From the State Key Laboratory of Pharmaceutical Biotechnology and Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China
| | - Rachel Toth
- the Medical Research Council Protein Phosphorylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom, and
| | - David G Campbell
- the Medical Research Council Protein Phosphorylation Unit, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom, and
| | - Carol MacKintosh
- the Division of Cell and Developmental Biology, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Hong Yu Wang
- From the State Key Laboratory of Pharmaceutical Biotechnology and Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China,
| | - Shuai Chen
- From the State Key Laboratory of Pharmaceutical Biotechnology and Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Pukou District, Nanjing 210061, China,
| |
Collapse
|
50
|
Rovelet-Lecrux A, Charbonnier C, Wallon D, Nicolas G, Seaman MNJ, Pottier C, Breusegem SY, Mathur PP, Jenardhanan P, Le Guennec K, Mukadam AS, Quenez O, Coutant S, Rousseau S, Richard AC, Boland A, Deleuze JF, Frebourg T, Hannequin D, Campion D. De novo deleterious genetic variations target a biological network centered on Aβ peptide in early-onset Alzheimer disease. Mol Psychiatry 2015; 20:1046-56. [PMID: 26194182 DOI: 10.1038/mp.2015.100] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/27/2015] [Accepted: 06/16/2015] [Indexed: 12/21/2022]
Abstract
We hypothesized that de novo variants (DNV) might participate in the genetic determinism of sporadic early-onset Alzheimer disease (EOAD, onset before 65 years). We investigated 14 sporadic EOAD trios first by array-comparative genomic hybridization. Two patients carried a de novo copy number variation (CNV). We then performed whole-exome sequencing in the 12 remaining trios and identified 12 non-synonymous DNVs in six patients. The two de novo CNVs (an amyloid precursor protein (APP) duplication and a BACE2 intronic deletion) and 3/12 non-synonymous DNVs (in PSEN1, VPS35 and MARK4) targeted genes from a biological network centered on the Amyloid beta (Aβ) peptide. We showed that this a priori-defined genetic network was significantly enriched in amino acid-altering DNV, compared with the rest of the exome. The causality of the APP de novo duplication (which is the first reported one) was obvious. In addition, we provided evidence of the functional impact of the following three non-synonymous DNVs targeting this network: the novel PSEN1 variant resulted in exon 9 skipping in patient's RNA, leading to a pathogenic missense at exons 8-10 junction; the VPS35 missense variant led to partial loss of retromer function, which may impact neuronal APP trafficking and Aβ secretion; and the MARK4 multiple nucleotide variant resulted into increased Tau phosphorylation, which may trigger enhanced Aβ-induced toxicity. Despite the difficulty to recruit Alzheimer disease (AD) trios owing to age structures of the pedigrees and the genetic heterogeneity of the disease, this strategy allowed us to highlight the role of de novo pathogenic events, the putative involvement of new genes in AD genetics and the key role of Aβ network alteration in AD.
Collapse
Affiliation(s)
- A Rovelet-Lecrux
- Inserm U1079, Institute for Research and Innovation in Biomedicine, University of Rouen, Rouen, France
| | - C Charbonnier
- Inserm U1079, Institute for Research and Innovation in Biomedicine, University of Rouen, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France
| | - D Wallon
- Inserm U1079, Institute for Research and Innovation in Biomedicine, University of Rouen, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France.,Department of Neurology, Rouen University Hospital, Rouen, France
| | - G Nicolas
- Inserm U1079, Institute for Research and Innovation in Biomedicine, University of Rouen, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France.,Department of Genetics, Rouen University Hospital, Rouen, France
| | - M N J Seaman
- Cambridge Institute for Medical Research/Dept of Clinical Biochemistry, University of Cambridge, Addenbrookes Hospital, Cambridge, UK
| | - C Pottier
- Inserm U1079, Institute for Research and Innovation in Biomedicine, University of Rouen, Rouen, France
| | - S Y Breusegem
- Cambridge Institute for Medical Research/Dept of Clinical Biochemistry, University of Cambridge, Addenbrookes Hospital, Cambridge, UK
| | - P P Mathur
- Centre of Bioinformatics and Department of Biochemistry & Molecular Biology, School of Life Sciences, Pondicherry University, Pondicherry, India.,KIIT University, Bhubaneshwar, Odisha, India
| | - P Jenardhanan
- Centre of Bioinformatics and Department of Biochemistry & Molecular Biology, School of Life Sciences, Pondicherry University, Pondicherry, India
| | - K Le Guennec
- Inserm U1079, Institute for Research and Innovation in Biomedicine, University of Rouen, Rouen, France
| | - A S Mukadam
- Cambridge Institute for Medical Research/Dept of Clinical Biochemistry, University of Cambridge, Addenbrookes Hospital, Cambridge, UK
| | - O Quenez
- Inserm U1079, Institute for Research and Innovation in Biomedicine, University of Rouen, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France
| | - S Coutant
- Inserm U1079, Institute for Research and Innovation in Biomedicine, University of Rouen, Rouen, France
| | - S Rousseau
- Inserm U1079, Institute for Research and Innovation in Biomedicine, University of Rouen, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France
| | - A-C Richard
- Inserm U1079, Institute for Research and Innovation in Biomedicine, University of Rouen, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France
| | - A Boland
- Centre National de Génotypage, Evry, France
| | | | - T Frebourg
- Inserm U1079, Institute for Research and Innovation in Biomedicine, University of Rouen, Rouen, France.,Department of Genetics, Rouen University Hospital, Rouen, France
| | - D Hannequin
- Inserm U1079, Institute for Research and Innovation in Biomedicine, University of Rouen, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France.,Department of Neurology, Rouen University Hospital, Rouen, France.,Department of Genetics, Rouen University Hospital, Rouen, France
| | - D Campion
- Inserm U1079, Institute for Research and Innovation in Biomedicine, University of Rouen, Rouen, France.,CNR-MAJ, Rouen University Hospital, Rouen, France.,Department of Research, Centre Hospitalier du Rouvray, Sotteville-Les-Rouen, France
| | | |
Collapse
|