1
|
Zou Z, Wu F, Chen L, Yao H, Wang Z, Chen Y, Qi M, Jiang Y, Tang L, Gan X, Kong L, Yang Z, Huang X, Shu W, Li B, Tan X, Huang L, Bai S, Wu L, Mo J, Hu H, Liu H, Zou R, Wei Y. The J bs-5YP peptide can alleviate dementia in senile mice by restoring the transcription of Slc40a1 to secrete the excessive iron from brain. J Adv Res 2025; 69:51-59. [PMID: 38527587 PMCID: PMC11954793 DOI: 10.1016/j.jare.2024.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/04/2024] [Accepted: 03/21/2024] [Indexed: 03/27/2024] Open
Abstract
INTRODUCTION With age and ATP decrease in the body, the transcription factors hypophosphorylation weakens the transcription of Slc40a1 and hinders the expression of the iron discharger ferroportin. This may lead to iron accumulation in the brain and the catalysis of free radicals that damage cerebral neurons and eventually lead to Alzheimer's disease (AD). OBJECTIVES To prevent AD caused by brain iron excretion disorders and reveal the mechanism of J bs-5YP peptide restoring ferroportin. METHODS We prepared J bs-YP peptide and administered it to the senile mice with dementia. Then, the intelligence of the mice was tested using a Morris Water Maze. The ATP content in the body was detected using the ATP hydrophysis and Phosphate precipitation method. The activation of Slc40a1 transcription was assayed with ATAC seq and the ferroportin, as well as the phosphorylation levels of Ets1 in brain were detected by Western Blot. RESULTS The phosphorylation level of Ets1in brain was enhanced, and subsequently, the transcription of Slc40a1 was activated and ferroportin was increased in the brain, the levels of iron and free radicals were reduced, with the neurons protection, and the dementia was ultimately alleviated in the senile mice. CONCLUSION J bs-5YP can recover the expression of ferroportin to excrete excessive iron in the brain of senile mice with dementia by enhancing the transcription of Slc40a1 via phosphorylating Ets1, revealing the potential of J bs-5YP as a drug to alleviate senile dementia.
Collapse
Affiliation(s)
- Zhenyou Zou
- Liuzhou Key Lab of Psychosis Treatment, Brain Hospital of Guangxi Zhuang Autonomous Region, Liuzhou 545005, China; Department of Biochemistry, Purdue University, West Lafayette, IN 47006, USA.
| | - Fengyao Wu
- Liuzhou Key Lab of Psychosis Treatment, Brain Hospital of Guangxi Zhuang Autonomous Region, Liuzhou 545005, China; Laboratory Medicine School of Dalian Medical University, Dalian 116000, China
| | - Liguan Chen
- Medical School of Taizhou University, Taizhou 318000, China
| | - Hua Yao
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541199, China
| | - Zengxian Wang
- Medical School of Taizhou University, Taizhou 318000, China
| | - Yongfeng Chen
- Medical School of Taizhou University, Taizhou 318000, China
| | - Ming Qi
- Liuzhou Key Lab of Psychosis Treatment, Brain Hospital of Guangxi Zhuang Autonomous Region, Liuzhou 545005, China
| | - Yang Jiang
- Liuzhou Key Lab of Psychosis Treatment, Brain Hospital of Guangxi Zhuang Autonomous Region, Liuzhou 545005, China
| | - Longhua Tang
- Laboratory Department of Pingnan People's Hospital, Pingnan 537399, China
| | - Xinying Gan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541199, China
| | - Lingjia Kong
- Laboratory of Xiaoshan Traditional Chinese Medicine Hospital, Hangzhou 311201, China
| | - Zhicheng Yang
- Liuzhou Key Lab of Psychosis Treatment, Brain Hospital of Guangxi Zhuang Autonomous Region, Liuzhou 545005, China
| | - Xiaolan Huang
- College of Public Health, Guangxi Medical University, Nanning 530021, China
| | - Wei Shu
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541199, China
| | - Bixue Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541199, China
| | - Xinyu Tan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541199, China
| | - Liwen Huang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541199, China
| | - Shi Bai
- Medical School of Taizhou University, Taizhou 318000, China
| | - Lijuan Wu
- Medical School of Taizhou University, Taizhou 318000, China
| | - Jinping Mo
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, 541199, China
| | - Huilin Hu
- Liuzhou Key Lab of Psychosis Treatment, Brain Hospital of Guangxi Zhuang Autonomous Region, Liuzhou 545005, China
| | - Huihua Liu
- Liuzhou Key Lab of Psychosis Treatment, Brain Hospital of Guangxi Zhuang Autonomous Region, Liuzhou 545005, China
| | - Ruyi Zou
- School Chemical and Environmental Engineering, Shangrao Normal University, Shangrao 334001, China.
| | - Yuhua Wei
- Liuzhou Key Lab of Psychosis Treatment, Brain Hospital of Guangxi Zhuang Autonomous Region, Liuzhou 545005, China.
| |
Collapse
|
2
|
Bahram Sangani N, Koetsier J, Mélius J, Kutmon M, Ehrhart F, Evelo CT, Curfs LMG, Reutelingsperger CP, Eijssen LMT. A novel insight into neurological disorders through HDAC6 protein-protein interactions. Sci Rep 2024; 14:14666. [PMID: 38918466 PMCID: PMC11199618 DOI: 10.1038/s41598-024-65094-1] [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: 02/07/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Due to its involvement in physiological and pathological processes, histone deacetylase 6 (HDAC6) is considered a promising pharmaceutical target for several neurological manifestations. However, the exact regulatory role of HDAC6 in the central nervous system (CNS) is still not fully understood. Hence, using a semi-automated literature screening technique, we systematically collected HDAC6-protein interactions that are experimentally validated and reported in the CNS. The resulting HDAC6 network encompassed 115 HDAC6-protein interactions divided over five subnetworks: (de)acetylation, phosphorylation, protein complexes, regulatory, and aggresome-autophagy subnetworks. In addition, 132 indirect interactions identified through HDAC6 inhibition were collected and categorized. Finally, to display the application of our HDAC6 network, we mapped transcriptomics data of Alzheimer's disease, Parkinson's disease, and Amyotrophic Lateral Sclerosis on the network and highlighted that in the case of Alzheimer's disease, alterations predominantly affect the HDAC6 phosphorylation subnetwork, whereas differential expression within the deacetylation subnetwork is observed across all three neurological disorders. In conclusion, the HDAC6 network created in the present study is a novel and valuable resource for the understanding of the HDAC6 regulatory mechanisms, thereby providing a framework for the integration and interpretation of omics data from neurological disorders and pharmacodynamic assessments.
Collapse
Affiliation(s)
- Nasim Bahram Sangani
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD, Maastricht, The Netherlands.
- GKC, Maastricht University Medical Centre, 6229 ER, Maastricht, The Netherlands.
| | - Jarno Koetsier
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD, Maastricht, The Netherlands
- GKC, Maastricht University Medical Centre, 6229 ER, Maastricht, The Netherlands
| | - Jonathan Mélius
- DataHub, Maastricht University & Maastricht UMC+, P. Debyelaan 15, 6229 HX, Maastricht, The Netherlands
| | - Martina Kutmon
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, 6200 MD, Maastricht, The Netherlands
| | - Friederike Ehrhart
- Department of Bioinformatics - BiGCaT, Research Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD, Maastricht, The Netherlands
- Department of Psychiatry and Neuropsychology, Research Institute for Mental Health and Neuroscience (MHeNs), Maastricht University, 6200 MD, Maastricht, The Netherlands
| | - Chris T Evelo
- Maastricht Centre for Systems Biology (MaCSBio), Maastricht University, 6200 MD, Maastricht, The Netherlands
- Department of Bioinformatics - BiGCaT, Research Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD, Maastricht, The Netherlands
| | - Leopold M G Curfs
- GKC, Maastricht University Medical Centre, 6229 ER, Maastricht, The Netherlands
| | - Chris P Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6200 MD, Maastricht, The Netherlands
- GKC, Maastricht University Medical Centre, 6229 ER, Maastricht, The Netherlands
| | - Lars M T Eijssen
- Department of Bioinformatics - BiGCaT, Research Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, 6200 MD, Maastricht, The Netherlands
- Department of Psychiatry and Neuropsychology, Research Institute for Mental Health and Neuroscience (MHeNs), Maastricht University, 6200 MD, Maastricht, The Netherlands
| |
Collapse
|
3
|
Afghah Z, Khan N, Datta G, Halcrow PW, Geiger JD, Chen X. Involvement of Endolysosomes and Aurora Kinase A in the Regulation of Amyloid β Protein Levels in Neurons. Int J Mol Sci 2024; 25:6200. [PMID: 38892390 PMCID: PMC11172969 DOI: 10.3390/ijms25116200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/28/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Aurora kinase A (AURKA) is a serine/threonine-protein kinase that regulates microtubule organization during neuron migration and neurite formation. Decreased activity of AURKA was found in Alzheimer's disease (AD) brain samples, but little is known about the role of AURKA in AD pathogenesis. Here, we demonstrate that AURKA is expressed in primary cultured rat neurons, neurons from adult mouse brains, and neurons in postmortem human AD brains. AURKA phosphorylation, which positively correlates with its activity, is reduced in human AD brains. In SH-SY5Y cells, pharmacological activation of AURKA increased AURKA phosphorylation, acidified endolysosomes, decreased the activity of amyloid beta protein (Aβ) generating enzyme β-site amyloid precursor protein cleaving enzyme (BACE-1), increased the activity of the Aβ degrading enzyme cathepsin D, and decreased the intracellular and secreted levels of Aβ. Conversely, pharmacological inhibition of AURKA decreased AURKA phosphorylation, de-acidified endolysosomes, decreased the activity of cathepsin D, and increased intracellular and secreted levels of Aβ. Thus, reduced AURKA activity in AD may contribute to the development of intraneuronal accumulations of Aβ and extracellular amyloid plaque formation.
Collapse
Affiliation(s)
| | | | | | | | | | - Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA; (Z.A.); (N.K.); (G.D.); (P.W.H.); (J.D.G.)
| |
Collapse
|
4
|
Bhattarai P, Taha A, Soni B, Thakuri DS, Ritter E, Chand GB. Predicting cognitive dysfunction and regional hubs using Braak staging amyloid-beta biomarkers and machine learning. Brain Inform 2023; 10:33. [PMID: 38043122 PMCID: PMC10694120 DOI: 10.1186/s40708-023-00213-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 11/21/2023] [Indexed: 12/05/2023] Open
Abstract
Mild cognitive impairment (MCI) is a transitional stage between normal aging and early Alzheimer's disease (AD). The presence of extracellular amyloid-beta (Aβ) in Braak regions suggests a connection with cognitive dysfunction in MCI/AD. Investigating the multivariate predictive relationships between regional Aβ biomarkers and cognitive function can aid in the early detection and prevention of AD. We introduced machine learning approaches to estimate cognitive dysfunction from regional Aβ biomarkers and identify the Aβ-related dominant brain regions involved with cognitive impairment. We employed Aβ biomarkers and cognitive measurements from the same individuals to train support vector regression (SVR) and artificial neural network (ANN) models and predict cognitive performance solely based on Aβ biomarkers on the test set. To identify Aβ-related dominant brain regions involved in cognitive prediction, we built the local interpretable model-agnostic explanations (LIME) model. We found elevated Aβ in MCI compared to controls and a stronger correlation between Aβ and cognition, particularly in Braak stages III-IV and V-VII (p < 0.05) biomarkers. Both SVR and ANN, especially ANN, showed strong predictive relationships between regional Aβ biomarkers and cognitive impairment (p < 0.05). LIME integrated with ANN showed that the parahippocampal gyrus, inferior temporal gyrus, and hippocampus were the most decisive Braak regions for predicting cognitive decline. Consistent with previous findings, this new approach suggests relationships between Aβ biomarkers and cognitive impairment. The proposed analytical framework can estimate cognitive impairment from Braak staging Aβ biomarkers and delineate the dominant brain regions collectively involved in AD pathophysiology.
Collapse
Affiliation(s)
- Puskar Bhattarai
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ahmed Taha
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Bhavin Soni
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Deepa S Thakuri
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- University of Missouri School of Medicine, Columbia, MO, USA
| | - Erin Ritter
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University McKelvey School of Engineering, St. Louis, MO, USA
| | - Ganesh B Chand
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA.
- Imaging Core, Knight Alzheimer Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
- Institute of Clinical and Translational Sciences, Washington University School of Medicine, St. Louis, MO, USA.
- NeuroGenomics and Informatics Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
5
|
Savva K, Zachariou M, Kynigopoulos D, Fella E, Vitali MI, Kosofidou X, Spyrou M, Sargiannidou I, Panayiotou E, Dietis N, Spyrou GM. Preliminary In Vitro and In Vivo Insights of In Silico Candidate Repurposed Drugs for Alzheimer's Disease. Life (Basel) 2023; 13:life13051095. [PMID: 37240740 DOI: 10.3390/life13051095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/04/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease and is the most common type of dementia. Although a considerably large amount of money has been invested in drug development for AD, no disease modifying treatment has been detected so far. In our previous work, we developed a computational method to highlight stage-specific candidate repurposed drugs against AD. In this study, we tested the effect of the top 13 candidate repurposed drugs that we proposed in our previous work in a severity stage-specific manner using an in vitro BACE1 assay and the effect of a top-ranked drug from the list of our previous work, tetrabenazine (TBZ), in the 5XFAD as an AD mouse model. From our in vitro screening, we detected 2 compounds (clomiphene citrate and Pik-90) that showed statistically significant inhibition against the activity of the BACE1 enzyme. The administration of TBZ at the selected dose and therapeutic regimen in 5XFAD in male and female mice showed no significant effect in behavioral tests using the Y-maze and the ELISA immunoassay of Aβ40. To our knowledge, this is the first time the drug tetrabenazine has been tested in the 5XFAD mouse model of AD in a sex-stratified manner. Our results highlight 2 drugs (clomiphene citrate and Pik-90) from our previous computational work for further investigation.
Collapse
Affiliation(s)
- Kyriaki Savva
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Margarita Zachariou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Demos Kynigopoulos
- Department of Neuropathology, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Eleni Fella
- Department of Neuropathology, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Maria-Ioanna Vitali
- Experimental Pharmacology Laboratory, Medical School, University of Cyprus, 2109 Nicosia, Cyprus
| | - Xeni Kosofidou
- Experimental Pharmacology Laboratory, Medical School, University of Cyprus, 2109 Nicosia, Cyprus
| | - Michail Spyrou
- Experimental Pharmacology Laboratory, Medical School, University of Cyprus, 2109 Nicosia, Cyprus
| | - Irene Sargiannidou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Elena Panayiotou
- Department of Neuropathology, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Nikolas Dietis
- Experimental Pharmacology Laboratory, Medical School, University of Cyprus, 2109 Nicosia, Cyprus
| | - George M Spyrou
- Bioinformatics Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| |
Collapse
|
6
|
Wu S, Xue Q, Yang M, Wang Y, Kim P, Zhou X, Huang L. Genetic control of RNA editing in neurodegenerative disease. Brief Bioinform 2023; 24:bbad007. [PMID: 36681936 PMCID: PMC10387301 DOI: 10.1093/bib/bbad007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/07/2022] [Accepted: 12/31/2022] [Indexed: 01/23/2023] Open
Abstract
A-to-I RNA editing diversifies human transcriptome to confer its functional effects on the downstream genes or regulations, potentially involving in neurodegenerative pathogenesis. Its variabilities are attributed to multiple regulators, including the key factor of genetic variants. To comprehensively investigate the potentials of neurodegenerative disease-susceptibility variants from the view of A-to-I RNA editing, we analyzed matched genetic and transcriptomic data of 1596 samples across nine brain tissues and whole blood from two large consortiums, Accelerating Medicines Partnership-Alzheimer's Disease and Parkinson's Progression Markers Initiative. The large-scale and genome-wide identification of 95 198 RNA editing quantitative trait loci revealed the preferred genetic effects on adjacent editing events. Furthermore, to explore the underlying mechanisms of the genetic controls of A-to-I RNA editing, several top RNA-binding proteins were pointed out, such as EIF4A3, U2AF2, NOP58, FBL, NOP56 and DHX9, since their regulations on multiple RNA-editing events were probably interfered by these genetic variants. Moreover, these variants may also contribute to the variability of other molecular phenotypes associated with RNA editing, including the functions of 3 proteins, expressions of 277 genes and splicing of 449 events. All the analyses results shown in NeuroEdQTL (https://relab.xidian.edu.cn/NeuroEdQTL/) constituted a unique resource for the understanding of neurodegenerative pathogenesis from genotypes to phenotypes related to A-to-I RNA editing.
Collapse
Affiliation(s)
- Sijia Wu
- School of Life Science and Technology, Xidian University, Xi’an 710071, China
| | - Qiuping Xue
- School of Life Science and Technology, Xidian University, Xi’an 710071, China
| | - Mengyuan Yang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yanfei Wang
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, Texas 77030, USA
| | - Pora Kim
- Corresponding authors: Liyu Huang, School of Life Science and Technology, Xidian University, Xi’an 710071, China. E-mail: ; Xiaobo Zhou, Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA. E-mail: ; Pora Kim, Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA. E-mail:
| | - Xiaobo Zhou
- Corresponding authors: Liyu Huang, School of Life Science and Technology, Xidian University, Xi’an 710071, China. E-mail: ; Xiaobo Zhou, Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA. E-mail: ; Pora Kim, Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA. E-mail:
| | - Liyu Huang
- Corresponding authors: Liyu Huang, School of Life Science and Technology, Xidian University, Xi’an 710071, China. E-mail: ; Xiaobo Zhou, Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA. E-mail: ; Pora Kim, Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA. E-mail:
| |
Collapse
|
7
|
Hagihara H, Murano T, Miyakawa T. The gene expression patterns as surrogate indices of pH in the brain. Front Psychiatry 2023; 14:1151480. [PMID: 37200901 PMCID: PMC10185791 DOI: 10.3389/fpsyt.2023.1151480] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/11/2023] [Indexed: 05/20/2023] Open
Abstract
Hydrogen ion (H+) is one of the most potent intrinsic neuromodulators in the brain in terms of concentration. Changes in H+ concentration, expressed as pH, are thought to be associated with various biological processes, such as gene expression, in the brain. Accumulating evidence suggests that decreased brain pH is a common feature of several neuropsychiatric disorders, including schizophrenia, bipolar disorder, autism spectrum disorder, and Alzheimer's disease. However, it remains unclear whether gene expression patterns can be used as surrogates for pH changes in the brain. In this study, we performed meta-analyses using publicly available gene expression datasets to profile the expression patterns of pH-associated genes, whose expression levels were correlated with brain pH, in human patients and mouse models of major central nervous system (CNS) diseases, as well as in mouse cell-type datasets. Comprehensive analysis of 281 human datasets from 11 CNS disorders revealed that gene expression associated with decreased pH was over-represented in disorders including schizophrenia, bipolar disorder, autism spectrum disorders, Alzheimer's disease, Huntington's disease, Parkinson's disease, and brain tumors. Expression patterns of pH-associated genes in mouse models of neurodegenerative disease showed a common time course trend toward lower pH over time. Furthermore, cell type analysis identified astrocytes as the cell type with the most acidity-related gene expression, consistent with previous experimental measurements showing a lower intracellular pH in astrocytes than in neurons. These results suggest that the expression pattern of pH-associated genes may be a surrogate for the state- and trait-related changes in pH in brain cells. Altered expression of pH-associated genes may serve as a novel molecular mechanism for a more complete understanding of the transdiagnostic pathophysiology of neuropsychiatric and neurodegenerative disorders.
Collapse
|
8
|
Haghighijoo Z, Zamani L, Moosavi F, Emami S. Therapeutic potential of quinazoline derivatives for Alzheimer's disease: A comprehensive review. Eur J Med Chem 2022; 227:113949. [PMID: 34742016 DOI: 10.1016/j.ejmech.2021.113949] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/02/2021] [Accepted: 10/22/2021] [Indexed: 12/24/2022]
Abstract
Quinazolines are considered as a promising class of bioactive heterocyclic compounds with broad properties. Particularly, the quinazoline scaffold has an impressive role in the design and synthesis of new CNS-active drugs. The drug-like properties and pharmacological characteristics of quinazoline could lead to different drugs with various targets. Among CNS disorders, Alzheimer's disease (AD) is a progressive neurodegenerative disorder with memory loss, cognitive decline and language dysfunction. AD is a complex and multifactorial disease therefore, the need for finding multi-target drugs against this devastative disease is urgent. A literature survey revealed that quinazoline derivatives have diverse therapeutic potential for AD as modulators/inhibitors of β-amyloid, tau protein, cholinesterases, monoamine oxidases, and phosphodiesterases as well as other protective effects. Thus, we describe here the most relevant and recent studies about anti-AD agents with quinazoline structure which can further aid the development and discovery of new anti-AD agents.
Collapse
Affiliation(s)
- Zahra Haghighijoo
- Department of Chemistry, University of Louisiana at Lafayette, Lafayette, LA, 70504, USA
| | - Leila Zamani
- Worcester Polytechnic Institute, 100 Institute Road, Worcester, MA, 01609, USA
| | - Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Emami
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
9
|
DePasquale EAK, Alganem K, Bentea E, Nawreen N, McGuire JL, Tomar T, Naji F, Hilhorst R, Meller J, McCullumsmith RE. KRSA: An R package and R Shiny web application for an end-to-end upstream kinase analysis of kinome array data. PLoS One 2021; 16:e0260440. [PMID: 34919543 PMCID: PMC8682895 DOI: 10.1371/journal.pone.0260440] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 11/09/2021] [Indexed: 12/22/2022] Open
Abstract
Phosphorylation by serine-threonine and tyrosine kinases is critical for determining protein function. Array-based platforms for measuring reporter peptide signal levels allow for differential phosphorylation analysis between conditions for distinct active kinases. Peptide array technologies like the PamStation12 from PamGene allow for generating high-throughput, multi-dimensional, and complex functional proteomics data. As the adoption rate of such technologies increases, there is an imperative need for software tools that streamline the process of analyzing such data. We present Kinome Random Sampling Analyzer (KRSA), an R package and R Shiny web-application for analyzing kinome array data to help users better understand the patterns of functional proteomics in complex biological systems. KRSA is an All-In-One tool that reads, formats, fits models, analyzes, and visualizes PamStation12 kinome data. While the underlying algorithm has been experimentally validated in previous publications, we demonstrate KRSA workflow on dorsolateral prefrontal cortex (DLPFC) in male (n = 3) and female (n = 3) subjects to identify differential phosphorylation signatures and upstream kinase activity. Kinase activity differences between males and females were compared to a previously published kinome dataset (11 female and 7 male subjects) which showed similar global phosphorylation signals patterns.
Collapse
Affiliation(s)
- Erica A. K. DePasquale
- Division of Hematology, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Boston, Massachusetts, United States of America
| | - Khaled Alganem
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, Ohio, United States of America
- * E-mail:
| | - Eduard Bentea
- Neuro-Aging & Viro-Immunotherapy, Center for Neurosciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nawshaba Nawreen
- Department of Neuroscience, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Jennifer L. McGuire
- Department of Neurosurgery, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Tushar Tomar
- PamGene International B.V., s’-Hertogenbosch, The Netherlands
| | - Faris Naji
- Tercen Data Analytics Ltd, Co Waterford, Ireland
| | - Riet Hilhorst
- PamGene International B.V., s’-Hertogenbosch, The Netherlands
| | - Jaroslaw Meller
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Department of Environmental Health, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Department of Electrical Engineering and Computing Systems, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- Department of Informatics, Nicolaus Copernicus University, Torun, Poland
| | - Robert E. McCullumsmith
- Department of Neurosciences, University of Toledo College of Medicine, Toledo, Ohio, United States of America
- Neurosciences institute, ProMedica, Toledo, Ohio, United States of America
| |
Collapse
|
10
|
Figueroa PBS, Ferreira AFF, Britto LR, Doussoulin AP, Torrão ADS. Association between thyroid function and Alzheimer's disease: A systematic review. Metab Brain Dis 2021; 36:1523-1543. [PMID: 34146214 DOI: 10.1007/s11011-021-00760-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 06/06/2021] [Indexed: 11/25/2022]
Abstract
Alterations in metabolic parameters have been associated with an increased risk of dementia, among which thyroid function has gained great importance in Alzheimer's disease (AD) pathology in recent years. However, it remains unclear whether thyroid dysfunctions could influence and contribute to the beginning and/or progression of AD or if it results from AD. This systematic review was conducted to examine the association between thyroid hormone (TH) levels and AD. Medline, ISI Web of Science, EMBASE, Cochrane library, Scopus, Scielo, and LILACS were searched, from January 2010 to March 2020. A total of 17 articles were selected. The studies reported alterations in TH and circadian rhythm in AD patients. Behavior, cognition, cerebral blood flow, and glucose consumption were correlated with TH deficits in AD patients. Whether thyroid dysfunctions and AD have a cause-effect relationship was inconclusive, however, the literature was able to provide enough data to corroborate a relationship between TH and AD. Although further studies are needed in this field, the current systematic review provides information that could help future investigations.
Collapse
Affiliation(s)
- Paulina Belén Sepulveda Figueroa
- Department of Preclinical Science, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile.
- Laboratory of Neuronal Communication, Departamento de Fisiologia e Biofisica, Universidade de Sao Paulo, Av. Professor Lineu Prestes, 1524 - Cidade Universitária, São Paulo, SP, Brasil, 05508900.
| | - Ana Flávia Fernandes Ferreira
- Laboratory of Cellular Neurobiology, Departamento de Fisiologia e Biofisica, Universidade de Sao Paulo, Av. Professor Lineu Prestes, 1524 - Cidade Universitária, São Paulo, SP, Brasil, 05508900.
| | - Luiz Roberto Britto
- Laboratory of Cellular Neurobiology, Departamento de Fisiologia e Biofisica, Universidade de Sao Paulo, Av. Professor Lineu Prestes, 1524 - Cidade Universitária, São Paulo, SP, Brasil, 05508900
| | | | - Andréa da Silva Torrão
- Laboratory of Neuronal Communication, Departamento de Fisiologia e Biofisica, Universidade de Sao Paulo, Av. Professor Lineu Prestes, 1524 - Cidade Universitária, São Paulo, SP, Brasil, 05508900
| |
Collapse
|
11
|
Yang M, Ke Y, Kim P, Zhou X. ExonSkipAD provides the functional genomic landscape of exon skipping events in Alzheimer's disease. Brief Bioinform 2021; 22:bbaa438. [PMID: 33497435 PMCID: PMC8425305 DOI: 10.1093/bib/bbaa438] [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: 09/27/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 12/18/2022] Open
Abstract
Exon skipping (ES), the most common alternative splicing event, has been reported to contribute to diverse human diseases due to the loss of functional domains/sites or frameshifting of the open reading frame (ORF) and noticed as therapeutic targets. Accumulating transcriptomic studies of aging brains show the splicing disruption is a widespread hallmark of neurodegenerative diseases such as Alzheimer's disease (AD). Here, we built ExonSkipAD, the ES annotation database aiming to provide a resource/reference for functional annotation of ES events in AD and identify therapeutic targets in exon units. We identified 16 414 genes that have ~156 K, ~ 69 K, ~ 231 K ES events from the three representative AD cohorts of ROSMAP, MSBB and Mayo, respectively. For these ES events, we performed multiple functional annotations relating to ES mechanisms or downstream. Specifically, through the functional feature retention studies followed by the open reading frames (ORFs), we identified 275 important cellular regulators that might lose their cellular regulator roles due to exon skipping in AD. ExonSkipAD provides twelve categories of annotations: gene summary, gene structures and expression levels, exon skipping events with PSIs, ORF annotation, exon skipping events in the canonical protein sequence, 3'-UTR located exon skipping events lost miRNA-binding sites, SNversus in the skipped exons with a depth of coverage, AD stage-associated exon skipping events, splicing quantitative trait loci (sQTLs) in the skipped exons, correlation with RNA-binding proteins, and related drugs & diseases. ExonSkipAD will be a unique resource of transcriptomic diversity research for understanding the mechanisms of neurodegenerative disease development and identifying potential therapeutic targets in AD. Significance AS the first comprehensive resource of the functional genomics of the alternative splicing events in AD, ExonSkipAD will be useful for many researchers in the fields of pathology, AD genomics and precision medicine, and pharmaceutical and therapeutic researches.
Collapse
|
12
|
Smith RG, Pishva E, Shireby G, Smith AR, Roubroeks JAY, Hannon E, Wheildon G, Mastroeni D, Gasparoni G, Riemenschneider M, Giese A, Sharp AJ, Schalkwyk L, Haroutunian V, Viechtbauer W, van den Hove DLA, Weedon M, Brokaw D, Francis PT, Thomas AJ, Love S, Morgan K, Walter J, Coleman PD, Bennett DA, De Jager PL, Mill J, Lunnon K. A meta-analysis of epigenome-wide association studies in Alzheimer's disease highlights novel differentially methylated loci across cortex. Nat Commun 2021; 12:3517. [PMID: 34112773 PMCID: PMC8192929 DOI: 10.1038/s41467-021-23243-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/16/2021] [Indexed: 01/01/2023] Open
Abstract
Epigenome-wide association studies of Alzheimer's disease have highlighted neuropathology-associated DNA methylation differences, although existing studies have been limited in sample size and utilized different brain regions. Here, we combine data from six DNA methylomic studies of Alzheimer's disease (N = 1453 unique individuals) to identify differential methylation associated with Braak stage in different brain regions and across cortex. We identify 236 CpGs in the prefrontal cortex, 95 CpGs in the temporal gyrus and ten CpGs in the entorhinal cortex at Bonferroni significance, with none in the cerebellum. Our cross-cortex meta-analysis (N = 1408 donors) identifies 220 CpGs associated with neuropathology, annotated to 121 genes, of which 84 genes have not been previously reported at this significance threshold. We have replicated our findings using two further DNA methylomic datasets consisting of a further >600 unique donors. The meta-analysis summary statistics are available in our online data resource ( www.epigenomicslab.com/ad-meta-analysis/ ).
Collapse
Affiliation(s)
- Rebecca G Smith
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Ehsan Pishva
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - Gemma Shireby
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Adam R Smith
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Janou A Y Roubroeks
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - Eilis Hannon
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Gregory Wheildon
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Diego Mastroeni
- Banner ASU Neurodegenerative Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Gilles Gasparoni
- Department of Genetics, University of Saarland (UdS), Saarbruecken, Germany
| | - Matthias Riemenschneider
- Department of Psychiatry and Psychotherapy, Saarland University Hospital (UKS), Homburg, Germany
| | - Armin Giese
- Center for Neuropathology and Prion Research, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Andrew J Sharp
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Vahram Haroutunian
- Department of Psychiatry, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, The Icahn School of Medicine at Mount Sinai, New York, NY, USA
- JJ Peters VA Medical Center, Bronx, NY, USA
| | - Wolfgang Viechtbauer
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
| | - Daniel L A van den Hove
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNS), Maastricht University, Maastricht, The Netherlands
- Laboratory of Translational Neuroscience, Department of Psychiatry, Psychosomatics and Psychotherapy, University of Wuerzburg, Würzburg, Germany
| | - Michael Weedon
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Danielle Brokaw
- Banner ASU Neurodegenerative Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - Paul T Francis
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Alan J Thomas
- Institute of Neuroscience, Newcastle University, Newcastle Upon Tyne, UK
| | - Seth Love
- Dementia Research Group, Institute of Clinical Neurosciences, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Kevin Morgan
- Human Genetics Group, University of Nottingham, Nottingham, UK
| | - Jörn Walter
- Department of Genetics, University of Saarland (UdS), Saarbruecken, Germany
| | - Paul D Coleman
- Banner ASU Neurodegenerative Research Center, Biodesign Institute, Arizona State University, Tempe, AZ, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Philip L De Jager
- Center for Translational and Computational Neuroimmunology, Department of Neurology and Taub Institute, Columbia University Medical Center, New York, NY, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan Mill
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK
| | - Katie Lunnon
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter, UK.
| |
Collapse
|
13
|
Khan H, Singh A, Thapa K, Garg N, Grewal AK, Singh TG. Therapeutic modulation of the phosphatidylinositol 3-kinases (PI3K) pathway in cerebral ischemic injury. Brain Res 2021; 1761:147399. [PMID: 33662337 DOI: 10.1016/j.brainres.2021.147399] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/09/2021] [Accepted: 02/23/2021] [Indexed: 12/16/2022]
Abstract
The cerebral ischemic reperfusion injury may leads to morbidity and mortality in patients. phosphatidylinositol 3-kinase (PI3K) signaling pathway has been believed to work in association with its downstream targets, other receptors, and pathways that may offer antioxidant, anti-inflammatory, anti-apoptotic effects, neuroprotective role in neuronal excitotoxicity. This review elaborates the mechanistic interventions of the PI3K pathway in cerebral ischemic injury in context to nuclear factor erythroid 2-related factor 2 (Nrf2) regulation, Hypoxia-inducible factor 1 signaling (HIF-1), growth factors, Endothelial NOS (eNOS) proinflammatory cytokines, Erythropoietin (EPO), Phosphatase and tensin homologous protein of chromosome 10 gene (PTEN) signaling, NF-κB/Notch signaling, c-Jun N-terminal kinase (JNK) and Glycogen synthase kinase-3β (GSK-3β) signaling pathway. Evidences showing the activation of PI3K inhibits apoptotic pathway, which results in its neuroprotective effect in ischemic injury. Despite discussing the therapeutic role of the PI3K pathway in treating cerebral ischemic injury, the review also enlighten the selective modulation of PI3K pathway with activators and inhibitors which may provide promising results in clinical and preclinical settings.
Collapse
Affiliation(s)
- Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Anjali Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Komal Thapa
- Chitkara College of Pharmacy, Chitkara University, Punjab, India; School of Pharmacy, Chitkara University, Himachal Pradesh, India
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | | |
Collapse
|
14
|
Sepulveda-Falla D, Chavez-Gutierrez L, Portelius E, Vélez JI, Dujardin S, Barrera-Ocampo A, Dinkel F, Hagel C, Puig B, Mastronardi C, Lopera F, Hyman BT, Blennow K, Arcos-Burgos M, de Strooper B, Glatzel M. A multifactorial model of pathology for age of onset heterogeneity in familial Alzheimer's disease. Acta Neuropathol 2021; 141:217-233. [PMID: 33319314 PMCID: PMC7847436 DOI: 10.1007/s00401-020-02249-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/22/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022]
Abstract
Presenilin-1 (PSEN1) mutations cause familial Alzheimer's disease (FAD) characterized by early age of onset (AoO). Examination of a large kindred harboring the PSEN1-E280A mutation reveals a range of AoO spanning 30 years. The pathophysiological drivers and clinical impact of AoO variation in this population are unknown. We examined brains of 23 patients focusing on generation and deposition of beta-amyloid (Aβ) and Tau pathology profile. In 14 patients distributed at the extremes of AoO, we performed whole-exome capture to identify genotype-phenotype correlations. We also studied kinome activity, proteasome activity, and protein polyubiquitination in brain tissue, associating it with Tau phosphorylation profiles. PSEN1-E280A patients showed a bimodal distribution for AoO. Besides AoO, there were no clinical differences between analyzed groups. Despite the effect of mutant PSEN1 on production of Aβ, there were no relevant differences between groups in generation and deposition of Aβ. However, differences were found in hyperphosphorylated Tau (pTau) pathology, where early onset patients showed severe pathology with diffuse aggregation pattern associated with increased activation of stress kinases. In contrast, late-onset patients showed lesser pTau pathology and a distinctive kinase activity. Furthermore, we identified new protective genetic variants affecting ubiquitin-proteasome function in early onset patients, resulting in higher ubiquitin-dependent degradation of differentially phosphorylated Tau. In PSEN1-E280A carriers, altered γ-secretase activity and resulting Aβ accumulation are prerequisites for early AoO. However, Tau hyperphosphorylation pattern, and its degradation by the proteasome, drastically influences disease onset in individuals with otherwise similar Aβ pathology, hinting toward a multifactorial model of disease for FAD. In sporadic AD (SAD), a wide range of heterogeneity, also influenced by Tau pathology, has been identified. Thus, Tau-induced heterogeneity is a common feature in both AD variants, suggesting that a multi-target therapeutic approach should be used to treat AD.
Collapse
Affiliation(s)
- Diego Sepulveda-Falla
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Neuroscience Group of Antioquia, Faculty of Medicine, University of Antioquia, Medellín, Colombia.
| | - Lucia Chavez-Gutierrez
- VIB Center for Brain and Disease Research, 3000, Leuven, Belgium
- Department of Neurology, KU Leuven, Leuven, Belgium
| | - Erik Portelius
- Institute of Neuroscience and Physiology, Dept. of Psychiatry and Neurochemistry, The Sahlgrenska Academy At the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 431 80, Mölndal, Sweden
| | - Jorge I Vélez
- Department of Genome Sciences, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- Universidad del Norte, Barranquilla, Colombia
| | - Simon Dujardin
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Charlestown, USA
| | - Alvaro Barrera-Ocampo
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Facultad de Ciencias Naturales, Departamento de Ciencias Farmaceuticas, Universidad Icesi, Grupo Natura, Calle 18 No. 122 -135, Cali, Colombia
| | - Felix Dinkel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Hagel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Berta Puig
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Claudio Mastronardi
- Department of Genome Sciences, John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
- GIPSI Group, Department of Psychiatry, Medical Research Institute, University of Antioquia, Medellín, Colombia
| | - Francisco Lopera
- Neuroscience Group of Antioquia, Faculty of Medicine, University of Antioquia, Medellín, Colombia
| | - Bradley T Hyman
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, MassGeneral Institute for Neurodegenerative Disease, Charlestown, USA
| | - Kaj Blennow
- Institute of Neuroscience and Physiology, Dept. of Psychiatry and Neurochemistry, The Sahlgrenska Academy At the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, 431 80, Mölndal, Sweden
| | - Mauricio Arcos-Burgos
- GIPSI Group, Department of Psychiatry, Medical Research Institute, University of Antioquia, Medellín, Colombia
| | - Bart de Strooper
- VIB Center for Brain and Disease Research, 3000, Leuven, Belgium
- Department of Neurology, KU Leuven, Leuven, Belgium
- UK Dementia Research Institute, University College London, Queen Square, London, WC1N 3BG, UK
| | - Markus Glatzel
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
15
|
Kumar S, Lemere CA, Walter J. Phosphorylated Aβ peptides in human Down syndrome brain and different Alzheimer's-like mouse models. Acta Neuropathol Commun 2020; 8:118. [PMID: 32727580 PMCID: PMC7388542 DOI: 10.1186/s40478-020-00959-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 05/30/2020] [Indexed: 12/12/2022] Open
Abstract
The deposition of neurotoxic amyloid-β (Aβ) peptides in extracellular plaques in the brain parenchyma is one of the most prominent neuropathological features of Alzheimer's disease (AD), and considered to be closely related to the pathogenesis of this disease. A number of recent studies demonstrate the heterogeneity in the composition of Aβ deposits in AD brains, due to the occurrence of elongated, truncated and post-translationally modified Aβ peptides that have peculiar characteristics in aggregation behavior and biostability. Importantly, the detection of modified Aβ species has been explored to characterize distinct stages of AD, with phosphorylated Aβ being present in the clinical phase of AD. People with Down syndrome (DS) develop AD pathology by 40 years of age likely due to the overproduction of Aβ caused by the additional copy of the gene encoding the amyloid precursor protein on chromosome 21. In the current study, we analysed the deposition of phosphorylated and non-phosphorylated Aβ species in human DS, AD, and control brains. In addition, deposition of these Aβ species was analysed in brains of a series of established transgenic AD mouse models using phosphorylation-state specific Aβ antibodies. Significant amounts of Aβ phosphorylated at serine residue 8 (pSer8Aβ) and unmodified Aβ were detected in the brains of DS and AD cases. The brains of different transgenic mouse models with either only human mutant amyloid precursor protein (APP), or combinations of human mutant APP, Presenilin (PS), and tau transgenes showed distinct age-dependent and spatiotemporal deposition of pSer8Aβ in extracellular plaques and within the vasculature. Together, these results demonstrate the deposition of phosphorylated Aβ species in DS brains, further supporting the similarity of Aβ deposition in AD and DS. Thus, the detection of phosphorylated and other modified Aβ species could contribute to the understanding and dissection of the complexity in the age-related and spatiotemporal deposition of Aβ variants in AD and DS as well as in distinct mouse models.
Collapse
|
16
|
Krayem M, Aftimos P, Najem A, van den Hooven T, van den Berg A, Hovestad-Bijl L, de Wijn R, Hilhorst R, Ruijtenbeek R, Sabbah M, Kerger J, Awada A, Journe F, Ghanem GE. Kinome Profiling to Predict Sensitivity to MAPK Inhibition in Melanoma and to Provide New Insights into Intrinsic and Acquired Mechanism of Resistance. Cancers (Basel) 2020; 12:E512. [PMID: 32098410 PMCID: PMC7072684 DOI: 10.3390/cancers12020512] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/13/2022] Open
Abstract
Mitogen-activated protein kinase (MAPK) inhibition with the combination of BRAF (Rapidly Accelerated Fibrosarcoma) and MEK (Mitogen-activated protein kinase kinase) inhibitors has become the standard of first-line therapy of metastatic melanoma harbouring BRAF V600 mutations. However, about half of the patients present with primary resistance while the remaining develop secondary resistance under prolonged treatment. Thus, there is a need for predictive biomarkers for sensitivity and/or resistance to further refine the patient population likely to benefit from MAPK inhibitors. In this study, we explored a top-down approach using a multiplex kinase assay, first, to discover a kinome signature predicting sensitivity, intrinsic and acquired resistance to MAPK inhibitors in melanoma, and second, to understand the mechanism of resistance using cell lines. Pre-dose tissues from patients (four responders and three non-responders to BRAFi monotherapy) were profiled for phosphotyrosine kinase (PTK) and serine-threonine kinase (STK) activities on a PamChip® peptide microarray in the presence and absence of ex vivo BRAFi. In addition, molecular studies were conducted on four sensitive parental lines, their offspring with acquired resistance to BRAFi and two lines with intrinsic resistance. PTK and STK activities in cell lysates were measured in the presence and absence of ex vivo BRAFi and/or MEKi. In tissue lysates, concentration-dependent ex vivo inhibition of STK and PTK activities with dabrafenib was stronger in responders than in non-responders. This difference was confirmed in cell lines comparing sensitive and resistant ones. Interestingly, common features of resistance were increased activity of receptor tyrosine kinases, Proto-oncogene tyrosine-protein kinase Src (Src) family kinases and protein kinase B (PKB, AKT) signalling. These latter results were confirmed by Western blots. While dabrafenib alone showed an inhibition of STK and PTK activities in both tissues and cell lines, the combination of dabrafenib and trametinib showed an antagonism on the STK activities and a synergism on PTK activities, resulting in stronger inhibitions of overall tyrosine kinase activities. Altogether; these data reveal that resistance of tumours and cell lines to MAPK inhibitors can be predicted using a multiplex kinase assay and is associated with an increase in specific tyrosine kinase activities and globally to AKT signalling in the patient's tissue. Thus, such a predictive kinome signature would help to identify patients with innate resistance to MAPK double inhibition in order to propose other therapies.
Collapse
Affiliation(s)
- Mohamad Krayem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (A.N.); (M.S.); (A.A.); (F.J.); (G.E.G.)
| | - Philippe Aftimos
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (P.A.); (J.K.)
| | - Ahmad Najem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (A.N.); (M.S.); (A.A.); (F.J.); (G.E.G.)
| | - Tim van den Hooven
- PamGene International BV, 5211HH ’s-Hertogenbosch, The Netherlands; (T.v.d.H.); (A.v.d.B.); (L.H.-B.); (R.d.W.); (R.H.); (R.R.)
| | - Adriënne van den Berg
- PamGene International BV, 5211HH ’s-Hertogenbosch, The Netherlands; (T.v.d.H.); (A.v.d.B.); (L.H.-B.); (R.d.W.); (R.H.); (R.R.)
| | - Liesbeth Hovestad-Bijl
- PamGene International BV, 5211HH ’s-Hertogenbosch, The Netherlands; (T.v.d.H.); (A.v.d.B.); (L.H.-B.); (R.d.W.); (R.H.); (R.R.)
| | - Rik de Wijn
- PamGene International BV, 5211HH ’s-Hertogenbosch, The Netherlands; (T.v.d.H.); (A.v.d.B.); (L.H.-B.); (R.d.W.); (R.H.); (R.R.)
| | - Riet Hilhorst
- PamGene International BV, 5211HH ’s-Hertogenbosch, The Netherlands; (T.v.d.H.); (A.v.d.B.); (L.H.-B.); (R.d.W.); (R.H.); (R.R.)
| | - Rob Ruijtenbeek
- PamGene International BV, 5211HH ’s-Hertogenbosch, The Netherlands; (T.v.d.H.); (A.v.d.B.); (L.H.-B.); (R.d.W.); (R.H.); (R.R.)
| | - Malak Sabbah
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (A.N.); (M.S.); (A.A.); (F.J.); (G.E.G.)
| | - Joseph Kerger
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (P.A.); (J.K.)
| | - Ahmad Awada
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (A.N.); (M.S.); (A.A.); (F.J.); (G.E.G.)
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (P.A.); (J.K.)
| | - Fabrice Journe
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (A.N.); (M.S.); (A.A.); (F.J.); (G.E.G.)
| | - Ghanem E. Ghanem
- Laboratory of Oncology and Experimental Surgery, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (A.N.); (M.S.); (A.A.); (F.J.); (G.E.G.)
| |
Collapse
|
17
|
Alzheimer’s disease (AD) therapeutics – 1: Repeated clinical failures continue to question the amyloid hypothesis of AD and the current understanding of AD causality. Biochem Pharmacol 2018; 158:359-375. [DOI: 10.1016/j.bcp.2018.09.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/24/2018] [Indexed: 12/17/2022]
|
18
|
Sawada T, Hilhorst R, Rangarajan S, Yoshida M, Tanabe Y, Tamura K, Kinoshita T, Shimoyama T, van Beuningen R, Ruijtenbeek R, Tsuda H, Koizumi F. Inactive immune pathways in triple negative breast cancers that showed resistance to neoadjuvant chemotherapy as inferred from kinase activity profiles. Oncotarget 2018; 9:34229-34239. [PMID: 30344939 PMCID: PMC6188135 DOI: 10.18632/oncotarget.26026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 08/02/2018] [Indexed: 12/15/2022] Open
Abstract
About 5% of Triple negative breast cancer patients (TNBCs) who receive neoadjuvant chemotherapy (NAC) experience progressive disease (PD). Few reports are published on TNBCs with PD during NAC, whereas TNBCs that respond to NAC have been well-studied. We investigated kinase activity profiles of TNBCs to explore the biological differences underlying the lack of response to NAC. Among 740 TNBCs, 20 non-responders were identified. Seven non-responders and 10 TNBCs that did not receive NAC (control group) were evaluated. No correlation was observed between NAC response and age, menopausal status, tumor size and axillary lymph node status. Tyrosine kinase activity profiles of TNBC primary tissues from NAC non-responders and the controls were determined with a peptide microarray system. Kinase activity measurements showed that 35 peptides had significantly (p < 0.05) lower phosphorylation in non-responders. ZAP70, LCK, SYK and JAK2 were identified as differentially active upstream kinases. Pathway analysis suggested lower activity in immune-related pathways in non-responders. The number of tumor infiltrating lymphocytes (TILs) was significantly lower (p = 0.0053) in non-responders. Kinases related to the immune system are less activated in non-responders. TILs evaluation suggested that the immune system is hardly active in non-responders and is not activated by NAC treatment.
Collapse
Affiliation(s)
- Takeshi Sawada
- Shien-Lab, National Cancer Center Hospital, Tokyo, Japan.,Division of Clinical Research Support, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Riet Hilhorst
- PamGene International BV, 's-Hertogenbosch, The Netherlands
| | | | - Masayuki Yoshida
- Department of Breast Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Yuko Tanabe
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kenji Tamura
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Takayuki Kinoshita
- Department of Breast Surgery, National Cancer Center Hospital, Tokyo, Japan
| | - Tatsu Shimoyama
- Division of Clinical Research Support, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | | | | | - Hitoshi Tsuda
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital, Tokyo, Japan.,Department of Basic Pathology, National Defense Medical College, Saitama, Japan
| | - Fumiaki Koizumi
- Shien-Lab, National Cancer Center Hospital, Tokyo, Japan.,Division of Clinical Research Support, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| |
Collapse
|
19
|
Dussaq AM, Kennell T, Eustace NJ, Anderson JC, Almeida JS, Willey CD. Kinomics toolbox-A web platform for analysis and viewing of kinomic peptide array data. PLoS One 2018; 13:e0202139. [PMID: 30130366 PMCID: PMC6103510 DOI: 10.1371/journal.pone.0202139] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 07/27/2018] [Indexed: 12/14/2022] Open
Abstract
Kinomics is an emerging field of science that involves the study of global kinase activity. As kinases are essential players in virtually all cellular activities, kinomic testing can directly examine protein function, distinguishing kinomics from more remote, upstream components of the central dogma, such as genomics and transcriptomics. While there exist several different approaches for kinomic research, peptide microarrays are the most widely used and involve kinase activity assessment through measurement of phosphorylation of peptide substrates on the array. Unfortunately, bioinformatic tools for analyzing kinomic data are quite limited necessitating the development of accessible open access software in order to facilitate standardization and dissemination of kinomic data for scientific use. Here, we examine and present tools for data analysis for the popular PamChip® (PamGene International) kinomic peptide microarray. As a result, we propose (1) a procedural optimization of kinetic curve data capture, (2) new methods for background normalization, (3) guidelines for the detection of outliers during parameterization, and (4) a standardized data model to store array data at various analytical points. In order to utilize the new data model, we developed a series of tools to implement the new methods and to visualize the various data models. In the interest of accessibility, we developed this new toolbox as a series of JavaScript procedures that can be utilized as either server side resources (easily packaged as web services) or as client side scripts (web applications running in the browser). The aggregation of these tools within a Kinomics Toolbox provides an extensible web based analytic platform that researchers can engage directly and web programmers can extend. As a proof of concept, we developed three analytical tools, a technical reproducibility visualizer, an ANOVA based detector of differentially phosphorylated peptides, and a heatmap display with hierarchical clustering.
Collapse
Affiliation(s)
- Alex M. Dussaq
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Timothy Kennell
- Informatics Institute, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Nicholas J. Eustace
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Joshua C. Anderson
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Jonas S. Almeida
- Department of Biomedical Informatics, Stony Brook University School of Medicine, Stony Brook, New York, United States of America
| | - Christopher D. Willey
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
20
|
Lanke V, Moolamalla STR, Roy D, Vinod PK. Integrative Analysis of Hippocampus Gene Expression Profiles Identifies Network Alterations in Aging and Alzheimer's Disease. Front Aging Neurosci 2018; 10:153. [PMID: 29875655 PMCID: PMC5974201 DOI: 10.3389/fnagi.2018.00153] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 05/04/2018] [Indexed: 01/22/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder contributing to rapid decline in cognitive function and ultimately dementia. Most cases of AD occur in elderly and later years. There is a growing need for understanding the relationship between aging and AD to identify shared and unique hallmarks associated with the disease in a region and cell-type specific manner. Although genomic studies on AD have been performed extensively, the molecular mechanism of disease progression is still not clear. The major objective of our study is to obtain a higher-order network-level understanding of aging and AD, and their relationship using the hippocampal gene expression profiles of young (20-50 years), aging (70-99 years), and AD (70-99 years). The hippocampus is vulnerable to damage at early stages of AD and altered neurogenesis in the hippocampus is linked to the onset of AD. We combined the weighted gene co-expression network and weighted protein-protein interaction network-level approaches to study the transition from young to aging to AD. The network analysis revealed the organization of co-expression network into functional modules that are cell-type specific in aging and AD. We found that modules associated with astrocytes, endothelial cells and microglial cells are upregulated and significantly correlate with both aging and AD. The modules associated with neurons, mitochondria and endoplasmic reticulum are downregulated and significantly correlate with AD than aging. The oligodendrocytes module does not show significant correlation with neither aging nor disease. Further, we identified aging- and AD-specific interactions/subnetworks by integrating the gene expression with a human protein-protein interaction network. We found dysregulation of genes encoding protein kinases (FYN, SYK, SRC, PKC, MAPK1, ephrin receptors) and transcription factors (FOS, STAT3, CEBPB, MYC, NFKβ, and EGR1) in AD. Further, we found genes that encode proteins with neuroprotective function (14-3-3 proteins, PIN1, ATXN1, BDNF, VEGFA) to be part of the downregulated AD subnetwork. Our study highlights that simultaneously analyzing aging and AD will help to understand the pre-clinical and clinical phase of AD and aid in developing the treatment strategies.
Collapse
Affiliation(s)
- Vinay Lanke
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Hyderabad, Hyderabad, India
| | - S T R Moolamalla
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Hyderabad, Hyderabad, India
| | - Dipanjan Roy
- Cognitive Brain Dynamics Lab, National Brain Research Centre, Gurgaon, India
| | - P K Vinod
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Hyderabad, Hyderabad, India
| |
Collapse
|
21
|
Batista AF, Forny-Germano L, Clarke JR, Lyra E Silva NM, Brito-Moreira J, Boehnke SE, Winterborn A, Coe BC, Lablans A, Vital JF, Marques SA, Martinez AM, Gralle M, Holscher C, Klein WL, Houzel JC, Ferreira ST, Munoz DP, De Felice FG. The diabetes drug liraglutide reverses cognitive impairment in mice and attenuates insulin receptor and synaptic pathology in a non-human primate model of Alzheimer's disease. J Pathol 2018; 245:85-100. [PMID: 29435980 PMCID: PMC5947670 DOI: 10.1002/path.5056] [Citation(s) in RCA: 182] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 01/13/2018] [Accepted: 02/05/2018] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurological disorder that still lacks an effective treatment, and this has stimulated an intense pursuit of disease-modifying therapeutics. Given the increasingly recognized link between AD and defective brain insulin signaling, we investigated the actions of liraglutide, a glucagon-like peptide-1 (GLP-1) analog marketed for treatment of type 2 diabetes, in experimental models of AD. Insulin receptor pathology is an important feature of AD brains that impairs the neuroprotective actions of central insulin signaling. Here, we show that liraglutide prevented the loss of brain insulin receptors and synapses, and reversed memory impairment induced by AD-linked amyloid-β oligomers (AβOs) in mice. Using hippocampal neuronal cultures, we determined that the mechanism of neuroprotection by liraglutide involves activation of the PKA signaling pathway. Infusion of AβOs into the lateral cerebral ventricle of non-human primates (NHPs) led to marked loss of insulin receptors and synapses in brain regions related to memory. Systemic treatment of NHPs with liraglutide provided partial protection, decreasing AD-related insulin receptor, synaptic, and tau pathology in specific brain regions. Synapse damage and elimination are amongst the earliest known pathological changes and the best correlates of memory impairment in AD. The results illuminate mechanisms of neuroprotection by liraglutide, and indicate that GLP-1 receptor activation may be harnessed to protect brain insulin receptors and synapses in AD. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Andre F Batista
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leticia Forny-Germano
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julia R Clarke
- School of Pharmacy, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Natalia M Lyra E Silva
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jordano Brito-Moreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Susan E Boehnke
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - Brian C Coe
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Ann Lablans
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Juliana F Vital
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Suelen A Marques
- Departament of Neurobiology, Fluminense Federal University, Niteroi, Brazil
| | - Ana Mb Martinez
- Department of Pathology, Faculty of Medicine, Hospital Universitário Clementino Fraga Filho, UFRJ, Rio de Janeiro, Brazil
| | - Matthias Gralle
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Christian Holscher
- Division of Biomed and Life Sciences, Faculty of Health and Medicine Lancaster University, Lancaster, UK
| | - William L Klein
- Department of Neurobiology, Northwestern University, Evanston, Illinois, USA
| | - Jean-Christophe Houzel
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas P Munoz
- Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Centre for Neuroscience Studies, Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
22
|
Qiao Z, Shiozawa K, Kondo T. Proteomic approach toward determining the molecular background of pazopanib resistance in synovial sarcoma. Oncotarget 2017; 8:109587-109595. [PMID: 29312631 PMCID: PMC5752544 DOI: 10.18632/oncotarget.22730] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/28/2017] [Indexed: 12/13/2022] Open
Abstract
Pazopanib, a multitarget tyrosine kinase (TK) inhibitor, has been approved for treatment of soft tissue sarcoma. Elucidation of the molecular background of pazopanib resistance should lead to improved clinical outcomes in sarcomas; accordingly, we investigated this in synovial sarcoma using a proteomic approach. Pazopanib sensitivity was examined in four synovial sarcoma cell lines: SYO-1, HS-SYII, 1273/99, and YaFuSS. The 1273/99 cell line showed significantly higher IC50 values than the others for pazopanib. Expression levels of 90 TKs in the cell lines were examined by western blotting. Among these, the levels of PDGFRB, DDR1, AXL, MET, and PYK2 were higher, and those of FGFR1 and VEGFR3 were lower in the 1273/99 cell line than the other cell lines. Gene silencing analysis of the TKs upregulated in 1273/99 cells showed differing effects on cell growth: PDGFRB, MET, and PYK2 knockdown induced cell growth inhibition, whereas DDR1 and AXL knockdown did not influence cell growth. Using the PamChip peptide microarray, we found that 18 peptide substrates were highly phosphorylated in the 1273/99 cell line compared with other cell lines. Using the PhosphoNet database, we found that kinases FGFR3, RET, VEGFR1, EPHA2, EPHA4, TRKA, and SRC phosphorylated these 18 peptide substrates. Moreover, the results for overexpressed and aberrantly activated TKs in pazopanib-resistant cells showed no overlap. Taken together, our study indicates that identification of comprehensive TK profiles represents an essential approach to determining the molecular background of pazopanib resistance in synovial sarcoma.
Collapse
Affiliation(s)
- Zhiwei Qiao
- Division of Rare Cancer Research, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan
| | - Kumiko Shiozawa
- Division of Rare Cancer Research, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan
| | - Tadashi Kondo
- Division of Rare Cancer Research, National Cancer Center Research Institute, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|
23
|
Technological advances for interrogating the human kinome. Biochem Soc Trans 2017; 45:65-77. [PMID: 28202660 DOI: 10.1042/bst20160163] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 10/20/2016] [Accepted: 10/25/2016] [Indexed: 12/12/2022]
Abstract
There is increasing appreciation among researchers and clinicians of the value of investigating biology and pathobiology at the level of cellular kinase (kinome) activity. Kinome analysis provides valuable opportunity to gain insights into complex biology (including disease pathology), identify biomarkers of critical phenotypes (including disease prognosis and evaluation of therapeutic efficacy), and identify targets for therapeutic intervention through kinase inhibitors. The growing interest in kinome analysis has fueled efforts to develop and optimize technologies that enable characterization of phosphorylation-mediated signaling events in a cost-effective, high-throughput manner. In this review, we highlight recent advances to the central technologies currently available for kinome profiling and offer our perspectives on the key challenges remaining to be addressed.
Collapse
|
24
|
Peppelenbosch MP, Frijns N, Fuhler G. Systems medicine approaches for peptide array-based protein kinase profiling: progress and prospects. Expert Rev Proteomics 2016; 13:571-8. [PMID: 27241729 DOI: 10.1080/14789450.2016.1187564] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Pharmacological manipulation of signalling pathways is becoming an increasingly important avenue for the rational clinical management of disease but is hampered by a lack of technologies that allow the generation of comprehensive descriptions of cellular signalling. AREAS COVERED Herein, the authors discuss the potential of peptide array-based kinome profiling for evaluating cellular signalling in the context of drug discovery. Expert commentary: Genomic and proteomic approaches have been of significant value to our elucidation of the molecular mechanisms that govern physiology. However, an equally, if not more important goal, is to define those proteins that participate in signalling pathways that ultimately control cell fate, especially kinases. Traditional genetic and biochemical approaches can certainly provide answers here, but for technical and practical reasons, are typically pursued one gene or pathway at a time. A more comprehensive approach is one in which peptide arrays of kinase-specific substrates are incubated with cell lysates and (33)P-ATP generating comprehensive descriptions, or where arrays are interrogated with phosphospecific antibodies. Both approaches allow analysis of cellular signalling without a priori assumptions to possibly influenced pathways.
Collapse
Affiliation(s)
| | | | - Gwenny Fuhler
- c Erasmus MC , Erasmus MC Cancer Institute , Rotterdam , Zuid-Holland, CA , Netherlands
| |
Collapse
|
25
|
Orsini M, Nascimento OJ, Matta AP, Reis CHM, de Souza OG, Bastos VH, Moreira R, Ribeiro P, Fiorelli S, Novellino P, Pessoa B, Cunha M, Pupe C, Morales PS, Filho PFM, Trajano EL, Oliveira AB. Revisiting the Term Neuroprotection in Chronic and Degenerative Diseases. Neurol Int 2016; 8:6311. [PMID: 27127599 PMCID: PMC4830365 DOI: 10.4081/ni.2016.6311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/01/2016] [Indexed: 11/25/2022] Open
Abstract
Thanks to the development of several new researches, the lifetime presented a significant increase, even so, we still have many obstacles to overcome - among them, manage and get responses regarding neurodegenerative diseases. Where we are in the understanding of neuroprotection? Do we really have protective therapies for diseases considered degeneratives such as amyotrophic lateral sclerosis and its variants, Parkinson's disease, Alzheimer's disease and many others? Neuroprotection is defined by many researches as interactions and interventions that can slow down or even inhibit the progression of neuronal degeneration process. We make some considerations on this neuroprotective effect.
Collapse
Affiliation(s)
- Marco Orsini
- Department of Neurosurgery, Universidade Federal Fluminense, 95 Tavares de Macedo street, 902 Niterói, RJ, ZIP 24220-211, Brazil.
| | - Osvaldo J.M. Nascimento
- Department of Neurology, Antonio Pedro University Hospital, Fluminense Federal University, Niterói
| | - Andre P.C. Matta
- Department of Neurology, Antonio Pedro University Hospital, Fluminense Federal University, Niterói
| | | | - Olivia Gameiro de Souza
- Department of Neurology, Antonio Pedro University Hospital, Fluminense Federal University, Niterói
| | - Victor Hugo Bastos
- Brain Mapping Laboratory and Electroencephalogram, Federal University of Rio de Janeiro
- Brain Mapping and Functionality Laboratory, Federal University of Piauí
| | - Rayele Moreira
- Brain Mapping Laboratory and Electroencephalogram, Federal University of Rio de Janeiro
- Brain Mapping and Functionality Laboratory, Federal University of Piauí
| | - Pedro Ribeiro
- Brain Mapping Laboratory and Electroencephalogram, Federal University of Rio de Janeiro
- Brain Mapping and Functionality Laboratory, Federal University of Piauí
| | - Stenio Fiorelli
- Severino Sombra University Center, School of Medicine, Vassouras
| | - Pietro Novellino
- Severino Sombra University Center, School of Medicine, Vassouras
| | - Bruno Pessoa
- Department of Neurology, Antonio Pedro University Hospital, Fluminense Federal University, Niterói
| | - Mariana Cunha
- Department of Neurology, Antonio Pedro University Hospital, Fluminense Federal University, Niterói
| | - Camila Pupe
- Department of Neurology, Antonio Pedro University Hospital, Fluminense Federal University, Niterói
| | - Pedro S. Morales
- Department of Neurology, Antonio Pedro University Hospital, Fluminense Federal University, Niterói
| | - Pedro F. Moreira Filho
- Department of Neurology, Antonio Pedro University Hospital, Fluminense Federal University, Niterói
| | | | | |
Collapse
|
26
|
Rosenberger AFN, Morrema THJ, Gerritsen WH, van Haastert ES, Snkhchyan H, Hilhorst R, Rozemuller AJM, Scheltens P, van der Vies SM, Hoozemans JJM. Increased occurrence of protein kinase CK2 in astrocytes in Alzheimer's disease pathology. J Neuroinflammation 2016; 13:4. [PMID: 26732432 PMCID: PMC4702323 DOI: 10.1186/s12974-015-0470-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 12/24/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common neurodegenerative disease. In addition to the occurrence of amyloid deposits and widespread tau pathology, AD is associated with a neuroinflammatory response characterized by the activation of microglia and astrocytes. Protein kinase 2 (CK2, former casein kinase II) is involved in a wide variety of cellular processes. Previous studies on CK2 in AD showed controversial results, and the involvement of CK2 in neuroinflammation in AD remains elusive. METHODS In this study, we used immunohistochemical and immunofluorescent staining methods to investigate the localization of CK2 in the hippocampus and temporal cortex of patients with AD and non-demented controls. We compared protein levels with Western blotting analysis, and we investigated CK2 activity in human U373 astrocytoma cells and human primary adult astrocytes stimulated with IL-1β or TNF-α. RESULTS We report increased levels of CK2 in the hippocampus and temporal cortex of AD patients compared to non-demented controls. Immunohistochemical analysis shows CK2 immunoreactivity in astrocytes in AD and control cases. In AD, the presence of CK2 immunoreactive astrocytes is increased. CK2 immunopositive astrocytes are associated with amyloid deposits, suggesting an involvement of CK2 in the neuroinflammatory response. In U373 cells and human primary astrocytes, the selective CK2 inhibitor CX-4945 shows a dose-dependent reduction of the IL-1β or TNF-α induced MCP-1 and IL-6 secretion. CONCLUSIONS This data suggests that CK2 in astrocytes is involved in the neuroinflammatory response in AD. The reduction in pro-inflammatory cytokine secretion by human astrocytes using the selective CK2 inhibitor CX-4945 indicates that CK2 could be a potential target to modulate neuroinflammation in AD.
Collapse
Affiliation(s)
- Andrea F N Rosenberger
- Alzheimer center & Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands.
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Tjado H J Morrema
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Wouter H Gerritsen
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Elise S van Haastert
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Hripsime Snkhchyan
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Riet Hilhorst
- PamGene International BV, Wolvenhoek 10, 5211 HH, 's-Hertogenbosch, The Netherlands.
| | - Annemieke J M Rozemuller
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Philip Scheltens
- Alzheimer center & Department of Neurology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands.
| | - Saskia M van der Vies
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Jeroen J M Hoozemans
- Department of Pathology, Neuroscience Campus Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| |
Collapse
|
27
|
Kandimalla R, Reddy PH. Multiple faces of dynamin-related protein 1 and its role in Alzheimer's disease pathogenesis. Biochim Biophys Acta Mol Basis Dis 2015; 1862:814-828. [PMID: 26708942 DOI: 10.1016/j.bbadis.2015.12.018] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/08/2015] [Accepted: 12/15/2015] [Indexed: 01/01/2023]
Abstract
Mitochondria play a large role in neuronal function by constantly providing energy, particularly at synapses. Recent studies suggest that amyloid beta (Aβ) and phosphorylated tau interact with the mitochondrial fission protein, dynamin-related protein 1 (Drp1), causing excessive fragmentation of mitochondria and leading to abnormal mitochondrial dynamics and synaptic degeneration in Alzheimer's disease (AD) neurons. Recent research also revealed Aβ-induced and phosphorylated tau-induced changes in mitochondria, particularly affecting mitochondrial shape, size, distribution and axonal transport in AD neurons. These changes affect mitochondrial health and, in turn, could affect synaptic function and neuronal damage and ultimately leading to memory loss and cognitive impairment in patients with AD. This article highlights recent findings in the role of Drp1 in AD pathogenesis. This article also highlights Drp1 and its relationships to glycogen synthase kinase 3, cyclin-dependent kinase 5, p53, and microRNAs in AD pathogenesis.
Collapse
Affiliation(s)
- Ramesh Kandimalla
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States
| | - P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Department of Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Department of Neurology, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, 6630 S. Quaker Ste. E, MS 7495, Lubbock, TX 79413, United States.
| |
Collapse
|