1
|
Pandey JP, Nietert PJ, Namboodiri AM, Bennett DA, Barnes LL. Epistatic effects of IGHG and FCGRIIB genes on the development of Alzheimer's disease in African Americans. Immunogenetics 2024; 77:1. [PMID: 39489839 PMCID: PMC11532319 DOI: 10.1007/s00251-024-01358-4] [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: 09/05/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Genome-wide association studies (GWAS) of Alzheimer's disease (AD) have identified a large number of susceptibility genes, but most of AD heritability remains unexplained, implying the existence of additional genes. Furthermore, the majority of the GWAS have been conducted in people of European descent, and the genes important for AD susceptibility in people of African descent have been underexplored. In this hypothesis-generating prospective cohort study, we genotyped 191 African Americans (AAs) from three longitudinal cohorts on aging for the IgG3 allotype GM6, which is expressed exclusively in people of African descent, and assessed its interaction with IGHG, FCGRIIB, and HLA-DRB1 genes. Cox proportional hazards modeling showed that GM6 by itself was not significantly associated with AD development. However, there was evidence of epistatic interaction: The risk of developing AD associated with GM6 positivity was significantly different (p = 0.0098) in non-GM17/GM17 participants compared with GM 17/GM17 participants. Specifically, in non-GM17/GM17 participants, the risk of AD was over fourfold higher in GM6-positive participants compared with GM 6-negative participants (HR = 4.63). Similarly, risk of developing AD associated with GM6 positivity was marginally different in non-FCGRIIB TT participants compared with FCGRIIB TT participants. In non-FCGRIIB TT participants, the risk of developing AD was over twofold higher in GM6-positive participants compared with GM6-negative participants (HR = 2.44). This is the first report suggesting that immunoglobulin GM allotypes might play a role in AD etiology among AAs; however, since this was largely a hypothesis-generating study, replication in larger cohorts would be required to confirm this finding.
Collapse
Affiliation(s)
- Janardan P Pandey
- Department of Pharmacology & Immunology, Medical University of South Carolina, Charleston, SC, 29425-2230, USA.
| | - Paul J Nietert
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Aryan M Namboodiri
- Department of Pharmacology & Immunology, Medical University of South Carolina, Charleston, SC, 29425-2230, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
2
|
Liu Y, Li Y, Zhang Y, Fang Y, Lei L, Yu J, Tan H, Sui L, Guo Q, Zhou L. Excitatory neurons and oligodendrocyte precursor cells are vulnerable to focal cortical dysplasia type IIIa as suggested by single-nucleus multiomics. Clin Transl Med 2024; 14:e70072. [PMID: 39440467 PMCID: PMC11497056 DOI: 10.1002/ctm2.70072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Focal cortical dysplasia (FCD) is a heterogeneous group of cortical developmental malformations that constitute a common cause of medically intractable epilepsy. FCD type IIIa (FCD IIIa) refers to temporal neocortex alterations in architectural organisation or cytoarchitectural composition in the immediate vicinity of hippocampal sclerosis. Slight alterations in the temporal neocortex of FCD IIIa patients pose a challenge for the preoperative diagnosis and definition of the resection range. METHODS We have performed multimodal integration of single-nucleus RNA sequencing and single-nucleus assay for transposase-accessible chromatin sequencing in the epileptogenic cortex of four patients with FCD IIIa, and three relatively normal temporal neocortex were chosen as controls. RESULTS Our study revealed that the most significant dysregulation occurred in excitatory neurons (ENs) and oligodendrocyte precursor cells (OPCs) in the epileptogenic cortex of FCD IIIa patients. In ENs, we constructed a transcription factor (TF)-hub gene regulatory network and found DAB1high ENs subpopulation mediates neuronal immunity characteristically in FCD IIIa. Western blotting and immunofluorescence were used to validate the changes in protein expression levels caused by some of the key genes. The OPCs were activated and exhibited aberrant phenotypes in FCD IIIa, and TFs regulating reconstructed pseudotime trajectory were identified. Finally, our results revealed aberrant intercellular communication between ENs and OPCs in FCD IIIa patients. CONCLUSIONS Our study revealed significant and intricate alterations in the transcriptomes and epigenomes in ENs and OPCs of FCD IIIa patients, shedding light on their cell type-specific regulation and potential pathogenic involvement in this disorder. This work will help evaluate the pathogenesis of cortical dysplasia and epilepsy and explore potential therapeutic targets. KEY POINTS Paired snRNA-seq and snATAC-seq data were intergrated and analysed to identify crucial subpopulations of ENs and OPCs in the epileptogenic cortex of FCD IIIa patients and explore their possible pathogenic role in the disease. A TF-hub gene regulatory network was constructed in ENs, and the DAB1high Ex-1 mediated neuronal immunity was characterstically in FCD IIIa patients. The OPCs were activated and exhibited aberrant phenotypes in FCD IIIa patients, and TFs regulating reconstructed pseudotime traectory were identified. Aberrant intercelluar communications between ENs and OPCs in FCD IIIa patients were identified.
Collapse
Affiliation(s)
- Yingying Liu
- Department of NeurologyThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
- Department of NeurologyThird Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdongChina
| | - Yinchao Li
- Department of NeurologyThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Yaqian Zhang
- Department of NeurologyThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Yubao Fang
- Department of NeurologyThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Lei Lei
- Department of NeurologyThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
| | - Jiabin Yu
- Department of Epilepsy CenterThe Second Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Hongping Tan
- Epilepsy CenterGuangdong Sanjiu Brain HospitalGuangzhouGuangdongChina
| | - Lisen Sui
- Department of Epilepsy CenterThe Second Affiliated HospitalGuangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Qiang Guo
- Epilepsy CenterGuangdong Sanjiu Brain HospitalGuangzhouGuangdongChina
| | - Liemin Zhou
- Department of NeurologyThe Seventh Affiliated HospitalSun Yat‐sen UniversityShenzhenGuangdongChina
| |
Collapse
|
3
|
Meng X, Song Q, Liu Z, Liu X, Wang Y, Liu J. Neurotoxic β-amyloid oligomers cause mitochondrial dysfunction-the trigger for PANoptosis in neurons. Front Aging Neurosci 2024; 16:1400544. [PMID: 38808033 PMCID: PMC11130508 DOI: 10.3389/fnagi.2024.1400544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024] Open
Abstract
As the global population ages, the incidence of elderly patients with dementia, represented by Alzheimer's disease (AD), will continue to increase. Previous studies have suggested that β-amyloid protein (Aβ) deposition is a key factor leading to AD. However, the clinical efficacy of treating AD with anti-Aβ protein antibodies is not satisfactory, suggesting that Aβ amyloidosis may be a pathological change rather than a key factor leading to AD. Identification of the causes of AD and development of corresponding prevention and treatment strategies is an important goal of current research. Following the discovery of soluble oligomeric forms of Aβ (AβO) in 1998, scientists began to focus on the neurotoxicity of AβOs. As an endogenous neurotoxin, the active growth of AβOs can lead to neuronal death, which is believed to occur before plaque formation, suggesting that AβOs are the key factors leading to AD. PANoptosis, a newly proposed concept of cell death that includes known modes of pyroptosis, apoptosis, and necroptosis, is a form of cell death regulated by the PANoptosome complex. Neuronal survival depends on proper mitochondrial function. Under conditions of AβO interference, mitochondrial dysfunction occurs, releasing lethal contents as potential upstream effectors of the PANoptosome. Considering the critical role of neurons in cognitive function and the development of AD as well as the regulatory role of mitochondrial function in neuronal survival, investigation of the potential mechanisms leading to neuronal PANoptosis is crucial. This review describes the disruption of neuronal mitochondrial function by AβOs and elucidates how AβOs may activate neuronal PANoptosis by causing mitochondrial dysfunction during the development of AD, providing guidance for the development of targeted neuronal treatment strategies.
Collapse
Affiliation(s)
| | | | | | | | | | - Jinyu Liu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China
| |
Collapse
|
4
|
Van der Auwera S, Ameling S, Wittfeld K, Frenzel S, Bülow R, Nauck M, Völzke H, Völker U, Grabe HJ. Circulating microRNA miR-425-5p Associated with Brain White Matter Lesions and Inflammatory Processes. Int J Mol Sci 2024; 25:887. [PMID: 38255959 PMCID: PMC10815886 DOI: 10.3390/ijms25020887] [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: 12/20/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
White matter lesions (WML) emerge as a consequence of vascular injuries in the brain. While they are commonly observed in aging, associations have been established with neurodegenerative and neurological disorders such as dementia or stroke. Despite substantial research efforts, biological mechanisms are incomplete and biomarkers indicating WMLs are lacking. Utilizing data from the population-based Study of Health in Pomerania (SHIP), our objective was to identify plasma-circulating micro-RNAs (miRNAs) associated with WMLs, thus providing a foundation for a comprehensive biological model and further research. In linear regression models, direct association and moderating factors were analyzed. In 648 individuals, we identified hsa-miR-425-5p as directly associated with WMLs. In subsequent analyses, hsa-miR-425-5p was found to regulate various genes associated with WMLs with particular emphasis on the SH3PXD2A gene. Furthermore, miR-425-5p was found to be involved in immunological processes. In addition, noteworthy miRNAs associated with WMLs were identified, primarily moderated by the factors of sex or smoking status. All identified miRNAs exhibited a strong over-representation in neurodegenerative and neurological diseases. We introduced hsa-miR-425-5p as a promising candidate in WML research probably involved in immunological processes. Mir-425-5p holds the potential as a biomarker of WMLs, shedding light on potential mechanisms and pathways in vascular dementia.
Collapse
Affiliation(s)
- Sandra Van der Auwera
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17475 Greifswald, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, 17475 Greifswald, Germany
| | - Sabine Ameling
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, 17475 Greifswald, Germany; (M.N.)
| | - Katharina Wittfeld
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Stefan Frenzel
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Robin Bülow
- Institute for Diagnostic Radiology and Neuroradiology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Matthias Nauck
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, 17475 Greifswald, Germany; (M.N.)
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Henry Völzke
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, 17475 Greifswald, Germany; (M.N.)
- Institute for Community Medicine, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, 17475 Greifswald, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Greifswald, 17475 Greifswald, Germany; (M.N.)
| | - Hans J. Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, 17475 Greifswald, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Site Rostock/Greifswald, 17475 Greifswald, Germany
| |
Collapse
|
5
|
Kim SH, Cho YS, Kim Y, Park J, Yoo SM, Gwak J, Kim Y, Gwon Y, Kam TI, Jung YK. Endolysosomal impairment by binding of amyloid beta or MAPT/Tau to V-ATPase and rescue via the HYAL-CD44 axis in Alzheimer disease. Autophagy 2023; 19:2318-2337. [PMID: 36843263 PMCID: PMC10351450 DOI: 10.1080/15548627.2023.2181614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/28/2023] Open
Abstract
Impaired activities and abnormally enlarged structures of endolysosomes are frequently observed in Alzheimer disease (AD) brains. However, little is known about whether and how endolysosomal dysregulation is triggered and associated with AD. Here, we show that vacuolar ATPase (V-ATPase) is a hub that mediates proteopathy of oligomeric amyloid beta (Aβ) and hyperphosphorylated MAPT/Tau (p-MAPT/Tau). Endolysosomal integrity was largely destroyed in Aβ-overloaded or p-MAPT/Tau-positive neurons in culture and AD brains, which was a necessary step for triggering neurotoxicity, and treatments with acidic nanoparticles or endocytosis inhibitors rescued the endolysosomal impairment and neurotoxicity. Interestingly, we found that the lumenal ATP6V0C and cytosolic ATP6V1B2 subunits of the V-ATPase complex bound to the internalized Aβ and cytosolic PHF-1-reactive MAPT/Tau, respectively. Their interactions disrupted V-ATPase activity and accompanying endolysosomal activity in vitro and induced neurodegeneration. Using a genome-wide functional screen, we isolated a suppressor, HYAL (hyaluronidase), which reversed the endolysosomal dysfunction and proteopathy and alleviated the memory impairment in 3xTg-AD mice. Further, we found that its metabolite hyaluronic acid (HA) and HA receptor CD44 attenuated neurotoxicity in affected neurons via V-ATPase. We propose that endolysosomal V-ATPase is a bona fide proteotoxic receptor that binds to pathogenic proteins and deteriorates endolysosomal function in AD, leading to neurodegeneration in proteopathy.Abbreviations: AAV, adeno-associated virus; Aβ, amyloid beta; AD, Alzheimer disease; APP, amyloid beta precursor protein; ATP6V0C, ATPase H+ transporting V0 subunit c; ATP6V1A, ATPase H+ transporting V1 subunit A; ATP6V1B2, ATPase H+ transporting V1 subunit B2; CD44.Fc, CD44-mouse immunoglobulin Fc fusion construct; Co-IP, co-immunoprecipitation; CTSD, cathepsin D; HA, hyaluronic acid; HMWHA, high-molecular-weight hyaluronic acid; HYAL, hyaluronidase; i.c.v, intracerebroventricular; LMWHA, low-molecular-weight hyaluronic acid; NPs, nanoparticles; p-MAPT/Tau, hyperphosphorylated microtubule associated protein tau; PI3K, phosphoinositide 3-kinase; V-ATPase, vacuolar-type H+-translocating ATPase; WT, wild-type.
Collapse
Affiliation(s)
- Seo-Hyun Kim
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Young-Sin Cho
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Youbin Kim
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul, Korea
| | - Jisu Park
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Seung-Min Yoo
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Jimin Gwak
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Youngwon Kim
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Youngdae Gwon
- School of Medicine, Sungkyunkwan University, Suwon, Korea
| | - Tae-in Kam
- Department of Neurology and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yong-Keun Jung
- School of Biological Sciences, Seoul National University, Seoul, Korea
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul, Korea
| |
Collapse
|
6
|
Rezaei M, Faramarzpour M, Shobeiri P, Seyedmirzaei H, Sarasyabi MS, Dabiri S. A systematic review, meta-analysis, and network analysis of diagnostic microRNAs in glaucoma. Eur J Med Res 2023; 28:137. [PMID: 36973823 PMCID: PMC10041737 DOI: 10.1186/s40001-023-01093-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 03/09/2023] [Indexed: 03/29/2023] Open
Abstract
Glaucoma is a chronic neurodegenerative process of the optic nerve that is the leading cause of blindness worldwide, and early diagnosis of the disease could greatly affect patients' prognoses. The pathophysiology of glaucoma is complicated by a combination of genetic and epigenetic factors. Deciphering the early diagnostic biomarkers in glaucoma could attenuate the disease's global burden and help us understand the exact mechanisms involved in glaucoma. The microRNAs are members of a larger family of non-coding RNAs that play an essential role in the epigenetic basis of glaucoma. A systematic study and meta-analysis of diagnostic microRNAs in glaucoma, jointly with network analysis of target genes, were carried out on published papers assessing differentially expressed microRNAs in human subjects. In total, 321 articles were found, and, after screening, six studies were eligible for further analysis. 52 differentially expressed microRNAs were found, of which 28 and 24 were up-regulated and down-regulated, respectively. Only 12 microRNAs were qualified for meta-analysis, with overall sensitivity and specificity of 80% and 74%, respectively. Then, using network analysis, it became apparent that the VEGF-A, AKT1, CXCL12, and HRAS genes were the most important targets for the microRNAs. Perturbations in WNT signaling, protein transport, and extracellular matrix organization pathways were discovered to be important in the etiology of glaucoma using the community detection approach. This study tries to uncover the promising microRNAs and their target genes that govern the epigenetics of glaucoma.
Collapse
Affiliation(s)
- Masoud Rezaei
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
- Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahsa Faramarzpour
- Pathology and Stem Cell Research Center, Department of Pathology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Parnian Shobeiri
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Endocrinology and Metabolism Population Sciences Institute, Non-Communicable Diseases Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Homa Seyedmirzaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Sharifi Sarasyabi
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
- Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Department of Radiology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Shahriar Dabiri
- Pathology and Stem Cell Research Center, Department of Pathology, Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
7
|
Nahalka J. 1-L Transcription in Alzheimer's Disease. Curr Issues Mol Biol 2022; 44:3533-3551. [PMID: 36005139 PMCID: PMC9406503 DOI: 10.3390/cimb44080243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/04/2022] [Accepted: 08/06/2022] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease is a very complex disease and better explanations and models are needed to understand how neurons are affected and microglia are activated. A new model of Alzheimer's disease is presented here, the β-amyloid peptide is considered an important RNA recognition/binding peptide. 1-L transcription revealed compatible sequences with AAUAAA (PAS signal) and UUUC (class III ARE rich in U) in the Aβ peptide, supporting the peptide-RNA regulatory model. When a hypothetical model of fibril selection with the prionic character of amyloid assemblies is added to the peptide-RNA regulatory model, the downregulation of the PI3K-Akt pathway and the upregulation of the PLC-IP3 pathway are well explained. The model explains why neurons are less protected from inflammation and why microglia are activated; why mitochondria are destabilized; why the autophagic flux is destabilized; and why the post-transcriptional attenuation of the axonal signal "noise" is interrupted. For example, the model suggests that Aβ peptide may post-transcriptionally control ELAVL2 (ELAV-like RNA binding protein 2) and DCP2 (decapping mRNA protein 2), which are known to regulate RNA processing, transport, and stability.
Collapse
Affiliation(s)
- Jozef Nahalka
- Institute of Chemistry, Centre for Glycomics, Slovak Academy of Sciences, Dubravska Cesta 9, SK-84538 Bratislava, Slovakia;
- Institute of Chemistry, Centre of Excellence for White-Green Biotechnology, Slovak Academy of Sciences, Trieda Andreja Hlinku 2, SK-94976 Nitra, Slovakia
| |
Collapse
|
8
|
Augustine J, Jereesh AS. Blood-based gene-expression biomarkers identification for the non-invasive diagnosis of Parkinson's disease using two-layer hybrid feature selection. Gene X 2022; 823:146366. [PMID: 35202733 DOI: 10.1016/j.gene.2022.146366] [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: 09/04/2021] [Revised: 02/15/2022] [Accepted: 02/18/2022] [Indexed: 11/19/2022] Open
Abstract
Parkinson's disease (PD) is one of the most prevalent neurodegenerative diseases. Understanding the molecular mechanism and identifying potential biomarkers of PD promote effective treatments to the patients. Due to less invasiveness and easy accessibility, biomarkers from blood support early detection and diagnosis of PD. This study combined three independent PD microarray gene expression data from blood samples applying the early integration approach. Moderated t-statistics was employed to identify differentially expressed genes (DEGs). Relevant genes were selected using a two-layer embedded wrapper feature selection method with gradient boosting machine (GBM) in the first layer followed by an ensemble of wrappers including Recursive Feature Elimination (RFE), Genetic algorithm (GA) and Bi-directional elimination (Stepwise). All three wrappers were based on logistic regression classifier (LR). The PD-predictability of the generated signature was tested using nine supervised classification models, including eight shallow machine learning and one deep learning. On an independent dataset, GSE72267, Support Vector Machine-Radial (SVMR), and Deep Neural Network (DNN) showed the best performance with AUC 0.821 and 0.82, respectively. Comparison with existing blood-based PD signatures and the biological analysis verified the reliability of the proposed signature.
Collapse
Affiliation(s)
- Jisha Augustine
- Bioinformatics Lab, Department of Computer Science, Cochin University of Science and Technology, Kerala 682022, India.
| | - A S Jereesh
- Bioinformatics Lab, Department of Computer Science, Cochin University of Science and Technology, Kerala 682022, India.
| |
Collapse
|
9
|
Gallego Villarejo L, Bachmann L, Marks D, Brachthäuser M, Geidies A, Müller T. Role of Intracellular Amyloid β as Pathway Modulator, Biomarker, and Therapy Target. Int J Mol Sci 2022; 23:ijms23094656. [PMID: 35563046 PMCID: PMC9103247 DOI: 10.3390/ijms23094656] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/15/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023] Open
Abstract
The β- and γ-secretase-driven cleavage of the amyloid precursor protein (APP) gives rise to the amyloid β peptide, which is believed to be the main driver of neurodegeneration in Alzheimer’s disease (AD). As it is prominently detectable in extracellular plaques in post-mortem AD brain samples, research in recent decades focused on the pathological role of extracellular amyloid β aggregation, widely neglecting the potential meaning of very early generation of amyloid β inside the cell. In the last few years, the importance of intracellular amyloid β (iAβ) as a strong player in neurodegeneration has been indicated by a rising number of studies. In this review, iAβ is highlighted as a crucial APP cleavage fragment, able to manipulate intracellular pathways and foster neurodegeneration. We demonstrate its relevance as a pathological marker and shed light on initial studies aiming to modulate iAβ through pharmacological treatment, which has been shown to have beneficial effects on cognitive properties in animal models. Finally, we display the relevance of viral infections on iAβ generation and point out future directions urgently needed to manifest the potential relevance of iAβ in Alzheimer’s disease.
Collapse
Affiliation(s)
- Lucia Gallego Villarejo
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - Lisa Bachmann
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - David Marks
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - Maite Brachthäuser
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - Alexander Geidies
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
| | - Thorsten Müller
- Department of Molecular Biochemistry, Cell Signalling, Ruhr University Bochum, 44801 Bochum, Germany; (L.G.V.); (L.B.); (D.M.); (M.B.); (A.G.)
- Institute of Psychiatric Phenomics and Genomics (IPPG), LMU University Hospital, LMU Munich, 80336 Munich, Germany
- Correspondence:
| |
Collapse
|
10
|
Chum PP, Hakim MA, Behringer EJ. Cerebrovascular microRNA Expression Profile During Early Development of Alzheimer's Disease in a Mouse Model. J Alzheimers Dis 2021; 85:91-113. [PMID: 34776451 DOI: 10.3233/jad-215223] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Emerging evidence demonstrates association of Alzheimer's disease (AD) with impaired delivery of blood oxygen and nutrients to and throughout the brain. The cerebral circulation plays multiple roles underscoring optimal brain perfusion and cognition entailing moment-to-moment blood flow control, vascular permeability, and angiogenesis. With currently no effective treatment to prevent or delay the progression of AD, cerebrovascular microRNA (miRNA) markers corresponding to post-transcriptional regulation may distinguish phases of AD. OBJECTIVE We tested the hypothesis that cerebrovascular miRNA expression profiles indicate developmental stages of AD pathology. METHODS Total RNA was isolated from total brain vessel segments of male and female 3xTg-AD mice [young, 1-2 mo; cognitive impairment (CI), 4-5 mo; extracellular amyloid-β plaques (Aβ), 6-8 mo; plaques+neurofibrillary tangles (AβT), 12-15 mo]. NanoString technology nCounter miRNA Expression panel for mouse was used to screen for 599 miRNAs. RESULTS Significant (p < 0.05) downregulation of various miRNAs indicated transitions from young to CI (e.g., let-7g & miR-1944, males; miR-133a & miR-2140, females) and CI to Aβ (e.g., miR-99a, males) but not from Aβ to AβT. In addition, altered expression of select miRNAs from overall Pre-AD (young + CI) versus AD (Aβ+ AβT) were detected in both males (let-7d, let-7i, miR-23a, miR-34b-3p, miR-99a, miR-126-3p, miR-132, miR-150, miR-151-5p, miR-181a) and females (miR-150, miR-539). Altogether, at least 20 cerebrovascular miRNAs effectively delineate AD versus Pre-AD pathology. CONCLUSION Using the 3xTg-AD mouse model, these data demonstrate that cerebrovascular miRNAs pertaining to endothelial function, vascular permeability, angiogenesis, inflammation, and Aβ/tau metabolism can track early development of AD.
Collapse
Affiliation(s)
- Phoebe P Chum
- Basic Sciences, Loma Linda University, Loma Linda, CA, USA
| | - Md A Hakim
- Basic Sciences, Loma Linda University, Loma Linda, CA, USA
| | | |
Collapse
|
11
|
Pandey JP, Namboodiri AM, Nietert PJ, Barnes LL, Bennett DA. Inhibitory Fcγ Receptor and Paired Immunoglobulin Type 2 Receptor Alpha Genotypes in Alzheimer's Disease. J Alzheimers Dis 2021; 84:965-968. [PMID: 34602489 DOI: 10.3233/jad-215174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
We investigated whether FCGRIIB (rs1050501 C/T) and PILRA (rs1859788 A/G) genotypes contributed to the development of Alzheimer's disease (AD). We genotyped 209 African American (AA) and 638 European American (EA) participants for the FCGRIIB and PILRA alleles. In the AA cohort, subjects homozygous for the C allele of FCGRIIB were more than 4 times as likely to develop AD as those homozygous for the alternative T allele. This SNP also interacted with PILRA: participants who were the carriers of the FCGRIIB C allele and PILRA A allele were 3 times as likely to develop AD as those who lacked these alleles.
Collapse
Affiliation(s)
- Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Aryan M Namboodiri
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Paul J Nietert
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Lisa L Barnes
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
12
|
Ma X, Wang Y, Yin H, Hua L, Zhang X, Xiao J, Yuan Q, Wang S, Liu Y, Zhang S, Wang Y. Down-regulated long non-coding RNA RMST ameliorates dopaminergic neuron damage in Parkinson's disease rats via regulation of TLR/NF-κB signaling pathway. Brain Res Bull 2021; 174:22-30. [PMID: 33933526 DOI: 10.1016/j.brainresbull.2021.04.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 02/04/2021] [Accepted: 04/26/2021] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Current treatment and prognosis of Parkinson's disease (PD) are not ideal. This study explored the mechanism of long non-coding RNA (lncRNA) rhabdomyosarcoma 2-associated transcript (RMST) in dopaminergic (DA) neuron damage in PD rats. METHODS PD rats were modeled and injected with RMST silence or overexpression vectors to figure out its roles in oxidative stress, the apoptosis of DA neurons in brain substantia nigra (SN), and neurobehavioral activities of PD rats. Tyrosine hydroxylase (TH), synaptophysin (SYN), glial fibrillary acidic protein (GFAP) and ionized calcium-binding adaptor molecule (Iba-1) in SN were detected. RMST and Toll-like receptor (TLR)/nuclear factor kappa B (NF-κB) pathway-related factors were detected. RESULTS RMST expression in brain SN of rats, TLR2, TLR4 expression in neurons and NF-κB expression in cell nucleus were increased. Silenced RMST improved the neurobehavioral activities, depressed oxidative stress and neuronal apoptosis, increased TH and SYN expression, and reduced the activation degree of glial cells in SN and the inflammatory response via reducing GFAP and Iba-1. Moreover, reduced RMST reduced TLR2 and TLR4 expression in neurons and NF-κB expression in cell nucleus in PD rats. CONCLUSION Inhibited RMST attenuates DA neuron damage in PD rats, which may be implicated with TLR/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Xuelian Ma
- Neurology Department, The 960th Hospital of the PLA, Zibo 255300, Shandong, China
| | - Yutong Wang
- Qilu Medical University, Zibo 255300, Shandong, China
| | - Honglei Yin
- Neurology Department, The 960th Hospital of the PLA, Zibo 255300, Shandong, China
| | - Linlin Hua
- Neurology Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, Henan, China
| | - Xiaolei Zhang
- Neurology Department, The 960th Hospital of the PLA, Zibo 255300, Shandong, China
| | - Jianhao Xiao
- Neurology Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, Henan, China
| | - Qian Yuan
- Neurology Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, Henan, China
| | - Shanshan Wang
- Neurology Department, The 960th Hospital of the PLA, Zibo 255300, Shandong, China
| | - Yajun Liu
- Neurology Department, The 960th Hospital of the PLA, Zibo 255300, Shandong, China
| | - Simiao Zhang
- Neurology Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, Henan, China
| | - Yunliang Wang
- Neurology Department, The 960th Hospital of the PLA, Zibo 255300, Shandong, China; Neurology Department, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, Henan, China.
| |
Collapse
|
13
|
Roach TG, Lång HKM, Xiong W, Ryhänen SJ, Capelluto DGS. Protein Trafficking or Cell Signaling: A Dilemma for the Adaptor Protein TOM1. Front Cell Dev Biol 2021; 9:643769. [PMID: 33718385 PMCID: PMC7952518 DOI: 10.3389/fcell.2021.643769] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 02/09/2021] [Indexed: 12/29/2022] Open
Abstract
Lysosomal degradation of ubiquitinated transmembrane protein receptors (cargo) relies on the function of Endosomal Sorting Complex Required for Transport (ESCRT) protein complexes. The ESCRT machinery is comprised of five unique oligomeric complexes with distinct functions. Target of Myb1 (TOM1) is an ESCRT protein involved in the initial steps of endosomal cargo sorting. To exert its function, TOM1 associates with ubiquitin moieties on the cargo via its VHS and GAT domains. Several ESCRT proteins, including TOLLIP, Endofin, and Hrs, have been reported to form a complex with TOM1 at early endosomal membrane surfaces, which may potentiate the role of TOM1 in cargo sorting. More recently, it was found that TOM1 is involved in other physiological processes, including autophagy, immune responses, and neuroinflammation, which crosstalk with its endosomal cargo sorting function. Alteration of TOM1 function has emerged as a phosphoinositide-dependent survival mechanism for bacterial infections and cancer progression. Based on current knowledge of TOM1-dependent cellular processes, this review illustrates how TOM1 functions in coordination with an array of protein partners under physiological and pathological scenarios.
Collapse
Affiliation(s)
- Tiffany G. Roach
- Protein Signaling Domains Laboratory, Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA, United States
| | - Heljä K. M. Lång
- Division of Hematology, Oncology, and Stem Cell Transplantation, Children’s Hospital, and Pediatric Research Center, The New Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Wen Xiong
- Protein Signaling Domains Laboratory, Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA, United States
| | - Samppa J. Ryhänen
- Division of Hematology, Oncology, and Stem Cell Transplantation, Children’s Hospital, and Pediatric Research Center, The New Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Daniel G. S. Capelluto
- Protein Signaling Domains Laboratory, Department of Biological Sciences, Fralin Life Sciences Institute, and Center for Soft Matter and Biological Physics, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
14
|
Wang Q, Ge X, Zhang J, Chen L. Effect of lncRNA WT1-AS regulating WT1 on oxidative stress injury and apoptosis of neurons in Alzheimer's disease via inhibition of the miR-375/SIX4 axis. Aging (Albany NY) 2020; 12:23974-23995. [PMID: 33234729 PMCID: PMC7762490 DOI: 10.18632/aging.104079] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
Objective: To study the effect of lncRNA WT1-AS on oxidative stress injury (OSI) and apoptosis of neurons in Alzheimer's disease (AD) and its specific mechanisms related to the microRNA-375 (miR-375)/SIX4 axis and WT1 expression. Results: After bioinformatic prediction, WT1-AS was found to be downregulated in Aβ25-35treated SH-SY5Y cells, and WT1-AS overexpression inhibited WT1 expression. WT1 could target miR-375 to promote its expression. miR-375 bound to SIX4, and miR-375 overexpression inhibited SIX4 expression. WT1-AS inhibited OSI and apoptosis, while WT1 and miR-375 overexpression or SIX4 silencing reversed the WT1-AS effect on OSI and apoptosis. In vivo experiments revealed that WT1-AS improved learning/memory abilities and inhibited OSI and apoptosis in AD mice. Conclusion: Overexpression of WT1-AS can inhibit the miR-375/SIX4 axis, OSI and neuronal apoptosis in AD by inhibiting WT1 expression. Methods: Related lncRNAs were identified, and miR-375 downstream targets were predicted. WT1-AS, WT1, miR-375 and SIX4 expression was detected in a cell model induced by Aβ25-35. The binding of WT1 with miR-375 and that of miR-375 with SIX4 were further confirmed. Adenosine triphosphate (ATP), reactive oxygen species (ROS), malondialdehyde (MDA), superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and lactate dehydrogenase (LDH) activities, and apoptosis levels were tested after mitochondrial membrane potential observation. Learning/memory abilities and neuronal apoptosis were tested in a mouse model.
Collapse
Affiliation(s)
- Quanbao Wang
- Department of Neurology, The People’s Hospital of Linyi City, Linyi 276000, P.R. China
| | - Xiumin Ge
- Department of Neurology, Linyi Mental Health Center, Linyi 276000, P.R. China
| | - Jie Zhang
- Department of Emergency Internal Medicine, The People’s Hospital of Linyi City, Linyi 276000, P.R. China
| | - Licheng Chen
- Department of Neurology, The People’s Hospital of Linyi City, Linyi 276000, P.R. China
| |
Collapse
|
15
|
Shi JM, Zhu L, Lan X, Zhao DW, He YJ, Sun ZQ, Wu D, Li HY. Endocytosis Is a Key Mode of Interaction between Extracellular β-Amyloid and the Cell Membrane. Biophys J 2020; 119:1078-1090. [PMID: 32857960 PMCID: PMC7499104 DOI: 10.1016/j.bpj.2020.07.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/21/2020] [Accepted: 07/08/2020] [Indexed: 11/23/2022] Open
Abstract
Interactions between amyloid-β peptide (Aβ) and the cell membrane include interaction with membrane lipids and binding to membrane receptors, both of which are considered to be the toxicity mechanisms of Aβ. However, it is unclear whether both mechanisms lead to cytotoxicity. Thus, we aimed to analyze these two mechanisms of Aβ42 interaction with cell membranes under different Aβ aggregation states. To this end, model membrane experiments were conducted. Quantitative analysis of Aβ42 monomers or oligomers bound to the membrane of neuro-2a cells was also performed, and laser confocal microscopy was employed to assess endocytosis of FITC-Aβ42 monomers or oligomers by neuro-2a cells. We found that the binding capacity of Aβ42 to membrane lipids was weak and that the amount of Aβ42 bound to membrane lipids was low. Moreover, clathrin-mediated endocytosis of Aβ42 oligomers by neuro-2a cells was observed. Endocytosis serves as a key mode of interaction between extracellular Aβ42 and neurons. These findings provide insights into the mechanisms underlying Aβ oligomer metabolism.
Collapse
Affiliation(s)
- Jing-Ming Shi
- School of Medicine, Xizang Minzu University, Xian' yang, Shaanxi, P.R. China
| | - Li Zhu
- School of Life Sciences, Lanzhou University, Lanzhou, P.R. China
| | - Xi Lan
- MOE Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China
| | - Duan-Wei Zhao
- Gansu Provincial Institute of Drug Control, Lanzhou, P.R. China
| | - Yong-Jun He
- School of Medicine, Xizang Minzu University, Xian' yang, Shaanxi, P.R. China
| | - Zheng-Qi Sun
- School of Medicine, Xizang Minzu University, Xian' yang, Shaanxi, P.R. China
| | - Di Wu
- School of Life Sciences, Lanzhou University, Lanzhou, P.R. China
| | - Hai-Yun Li
- MOE Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, P. R. China.
| |
Collapse
|
16
|
Costa AS, Agostini S, Guerini FR, Mancuso R, Clerici M, Pandey JP. Relation between FCGRIIB rs1050501 and HSV-1 specific IgG antibodies in Alzheimer's disease. J Transl Med 2020; 18:325. [PMID: 32859213 PMCID: PMC7455989 DOI: 10.1186/s12967-020-02495-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 08/20/2020] [Indexed: 01/08/2023] Open
Abstract
Background Alzheimer’s Disease (AD) is a chronic neurodegenerative disorder characterized by extracellular plaques, intracellular neurofibrillary tangles and neuronal loss in the central nervous system (CNS). Pathogens are suspected to have a role in the development of AD; herpes simplex virus type 1 (HSV-1), in particular, is suggested to be a risk factor for the disease. The gamma receptor for the Fc portion of IgG molecules (FCGRs) plays a crucial role in regulating immune responses, and among FCGRs, FCGRIIB is endowed with an inhibitory function. Notably, the rs1050501 polymorphism of FCGRIIB gene associates with autoimmune diseases and with neuronal uptake and interneuronal accumulation of amyloid beta in animal AD models. Methods Genotype and allelic distribution of ApoE4 and FCGRIIB rs1050501 were evaluated in a case–control population of 225 AD patients, 93 MCI individuals and 201 sex and age matched healthy controls (HC). HSV-1 total IgG titers and IgG subclasses were detected and quantified in a subgroup of the main study population by ELISA. Results Genotype and allelic distribution of FCGRIIB was comparable in the study population. HSV-1-specific antibody titers were significantly higher in AD and MCI compared to HC (p < 0.01 for both); IgG3 titers, in particular, were increased in MCI compared to AD (p = 0.04). Analyses of possible correlations between the FCGRIIB rs1050501 genotype polymorphism and IgG subclasses showed that the presence of IgG3 was more frequent in MCI carrying the FCGRIIB TT (94.1%) compared to those carrying the CT genotype (63.6%) (p = 0.03). Conclusion Results herein show an association between humoral immune response against HSV-1 and FCGRIIB rs1050501 genetic variation in the first stage of the disease.
Collapse
Affiliation(s)
| | | | | | | | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milano, Milan, Italy
| | - Janardan P Pandey
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
17
|
Su X, Xiao D, Huang L, Li S, Ying J, Tong Y, Ye Q, Mu D, Qu Y. MicroRNA Alteration in Developing Rat Oligodendrocyte Precursor Cells Induced by Hypoxia-Ischemia. J Neuropathol Exp Neurol 2020; 78:900-909. [PMID: 31403686 DOI: 10.1093/jnen/nlz071] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
microRNAs (miRNAs) are involved in the pathogenesis of white matter injury (WMI). However, their roles in developing rat brains under hypoxia-ischemia (HI) insult remain unknown. Here, we examined the expression profiles of miRNAs in oligodendrocyte precursor cells using microarray analysis. We identified 162 miRNAs and only 6 were differentially regulated in HI compared with sham. Next, we used these 6 miRNAs and 525 extensively changed coding genes (fold change absolute: FC(abs) ≥2, p < 0.05) to establish the coexpression network, the result revealed that only 3 miRNAs (miR-142-3p, miR-466b-5p, and miR-146a-5p) have differentially expressed targeted mRNAs. RT-PCR analysis showed that the expression of the miRNAs was consistent with the microarray analysis. Further gene ontology and KEGG pathway analysis of the targets of these 3 miRNAs indicated that they were largely associated with neural activity. Furthermore, we found that 2 of the 3 miRNAs, miR-142-3p, and miR-466b-5p, have the same target gene, Capn6, an antiapoptotic gene that is tightly regulated in the pathogenesis of neurological diseases. Collectively, we have shown that a number of miRNAs change in oligodendrocyte precursor cells in response to HI insult in developing brains, and miR-142-3p/miR-466b-5p/Capn6 pathway might affect the pathogenesis of WMI, providing us new clues for the diagnosis and therapy for WMI.
Collapse
Affiliation(s)
- Xiaojuan Su
- Department of Pediatrics, West China Second University Hospital.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education
| | - Dongqiong Xiao
- Department of Pediatrics, West China Second University Hospital.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education
| | - Lingyi Huang
- West China College of Stomatology, Sichuan University, Chengdu, China
| | - Shiping Li
- Department of Pediatrics, West China Second University Hospital.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education
| | - Yu Tong
- Department of Pediatrics, West China Second University Hospital.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education
| | - Qianghua Ye
- Department of Pediatrics, West China Second University Hospital.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital.,Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education
| |
Collapse
|
18
|
Reply to Peng and Zhao: Loss of endocytic protein TOM1 in Alzheimer's disease. Proc Natl Acad Sci U S A 2020; 117:3917-3919. [PMID: 32047040 PMCID: PMC7049100 DOI: 10.1073/pnas.1917743117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
19
|
Martini AC, Gomez-Arboledas A, Forner S, Rodriguez-Ortiz CJ, McQuade A, Danhash E, Phan J, Javonillo D, Ha JV, Tram M, Trujillo-Estrada L, da Cunha C, Ager RR, Davila JC, Kitazawa M, Blurton-Jones M, Gutierrez A, Baglietto-Vargas D, Medeiros R, LaFerla FM. Amyloid-beta impairs TOM1-mediated IL-1R1 signaling. Proc Natl Acad Sci U S A 2019; 116:21198-21206. [PMID: 31570577 PMCID: PMC6800331 DOI: 10.1073/pnas.1914088116] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Defects in interleukin-1β (IL-1β)-mediated cellular responses contribute to Alzheimer's disease (AD). To decipher the mechanism associated with its pathogenesis, we investigated the molecular events associated with the termination of IL-1β inflammatory responses by focusing on the role played by the target of Myb1 (TOM1), a negative regulator of the interleukin-1β receptor-1 (IL-1R1). We first show that TOM1 steady-state levels are reduced in human AD hippocampi and in the brain of an AD mouse model versus respective controls. Experimentally reducing TOM1 affected microglia activity, substantially increased amyloid-beta levels, and impaired cognition, whereas enhancing its levels was therapeutic. These data show that reparation of the TOM1-signaling pathway represents a therapeutic target for brain inflammatory disorders such as AD. A better understanding of the age-related changes in the immune system will allow us to craft therapies to limit detrimental aspects of inflammation, with the broader purpose of sharply reducing the number of people afflicted by AD.
Collapse
Affiliation(s)
- Alessandra Cadete Martini
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Angela Gomez-Arboledas
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
| | - Stefania Forner
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Carlos J Rodriguez-Ortiz
- Center for Occupational and Environmental Health, School of Medicine, University of California, Irvine, CA 92697
| | - Amanda McQuade
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697
| | - Emma Danhash
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697
| | - Jimmy Phan
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Dominic Javonillo
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Jordan-Vu Ha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Melanie Tram
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Laura Trujillo-Estrada
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
| | - Celia da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Rahasson R Ager
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
| | - Jose C Davila
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
| | - Masashi Kitazawa
- Center for Occupational and Environmental Health, School of Medicine, University of California, Irvine, CA 92697
| | - Mathew Blurton-Jones
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA 92697
| | - Antonia Gutierrez
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
| | - David Baglietto-Vargas
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Instituto de Investigación Biomédica de Málaga-IBIMA, Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), University of Málaga, Málaga 29010, Spain
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
| | - Rodrigo Medeiros
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697;
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA 92697;
- Department of Neurobiology and Behavior, University of California, Irvine, CA 92697
| |
Collapse
|
20
|
Exploratory study on microRNA profiles from plasma-derived extracellular vesicles in Alzheimer's disease and dementia with Lewy bodies. Transl Neurodegener 2019; 8:31. [PMID: 31592314 PMCID: PMC6775659 DOI: 10.1186/s40035-019-0169-5] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022] Open
Abstract
Background Because of the increasing life expectancy in our society, aging-related neurodegenerative disorders are one of the main issues in global health. Most of these diseases are characterized by the deposition of misfolded proteins and a progressive cognitive decline. Among these diseases, Alzheimer’s disease (AD) and dementia with Lewy bodies (DLB) are the most common types of degenerative dementia. Although both show specific features, an important neuropathological and clinical overlap between them hampers their correct diagnosis. In this work, we identified molecular biomarkers aiming to improve the misdiagnosis between both diseases. Methods Plasma extracellular vesicles (EVs) -from DLB, AD and healthy controls- were isolated using size-exclusion chromatography (SEC) and characterized by flow cytometry, Nanoparticle Tracking Analysis (NTA) and cryo-electron microscopy. Next Generation Sequencing (NGS) and related bibliographic search was performed and a selected group of EV-associated microRNAs (miRNAs) was analysed by qPCR. Results Results uncovered two miRNAs (hsa-miR-451a and hsa-miR-21-5p) significantly down-regulated in AD samples respect to DLB patients, and a set of four miRNAs (hsa-miR-23a-3p, hsa-miR-126-3p, hsa-let-7i-5p, and hsa-miR-151a-3p) significantly decreased in AD respect to controls. The two miRNAs showing decreased expression in AD in comparison to DLB provided area under the curve (AUC) values of 0.9 in ROC curve analysis, thus suggesting their possible use as biomarkers to discriminate between both diseases. Target gene analysis of these miRNAs using prediction online tools showed accumulation of phosphorylation enzymes, presence of proteasome-related proteins and genes involved in cell death among others. Conclusion Our data suggest that plasma-EV associated miRNAs may reflect a differential profile for a given dementia-related disorder which, once validated in larger cohorts of patients, could help to improve the differential diagnosis of DLB versus AD. Electronic supplementary material The online version of this article (10.1186/s40035-019-0169-5) contains supplementary material, which is available to authorized users.
Collapse
|
21
|
Quan Q, Qian Y, Li X, Li M. Pioglitazone Reduces β Amyloid Levels via Inhibition of PPARγ Phosphorylation in a Neuronal Model of Alzheimer's Disease. Front Aging Neurosci 2019; 11:178. [PMID: 31379559 PMCID: PMC6650543 DOI: 10.3389/fnagi.2019.00178] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 07/02/2019] [Indexed: 12/28/2022] Open
Abstract
It has been demonstrated that peroxisome proliferator-activated receptor γ (PPARγ) can regulate the transcription of its target gene, insulin-degrading enzyme (IDE), and thus enhance the expression of the IDE protein. The protein can degrade β amyloid (Aβ), a core pathological product of Alzheimer’s disease (AD). PPARγ can also regulate the transcription of other target gene, β-amyloid cleavage enzyme 1 (BACE1), and thus inhibit the expression of the BACE1 protein. BACE1 can hydrolyze amyloid precursor protein (APP), the precursor of Aβ. In adipose tissue, PPARγ agonists can inhibit the phosphorylation of PPARγ by inhibiting cyclin-dependent kinase 5 (CDK5), which in turn affects the expression of target genes regulated by PPARγ. PPARγ agonists may also exert inhibitory effects on the phosphorylation of PPARγ in the brain, thereby affecting the expression of the aforementioned PPARγ target genes and reducing Aβ levels. The present study confirmed this hypothesis by showing that PPARγ agonist pioglitazone attenuated the neuronal apoptosis of primary rat hippocampal neurons induced by Aβ1–42, downregulated CDK5 expression, weakened the binding of CDK5 to PPARγ, reduced PPARγ phosphorylation, increased the expression of PPARγ and IDE, decreased the expression of BACE1, reduced APP production, and downregulated intraneuronal Aβ1–42 levels. These effects were inhibited by the PPARγ antagonist GW9662. After CDK5 silencing with CDK5 shRNA, the above effect of pioglitazone was not observed, except when upregulating the expression of PPARγ in Aβ1–42 treated neurons. In conclusion, this study demonstrated that pioglitazone could inhibit the phosphorylation of PPARγ in vitro by inhibiting CDK5 expression, which in turn affected the expression of PPARγ target genes Ide and Bace1, thereby promoting Aβ degradation and reducing Aβ production. This reduced Aβ levels in the brain, thereby exerting neuroprotective effects in an AD model.
Collapse
Affiliation(s)
- Qiankun Quan
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yihua Qian
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education of China, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xi Li
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ming Li
- Department of Geriatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
22
|
Cardioprotective role of APIP in myocardial infarction through ADORA2B. Cell Death Dis 2019; 10:511. [PMID: 31263105 PMCID: PMC6602929 DOI: 10.1038/s41419-019-1746-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 05/25/2019] [Accepted: 05/28/2019] [Indexed: 12/19/2022]
Abstract
In ischemic human hearts, the induction of adenosine receptor A2B (ADORA2B) is associated with cardioprotection against ischemic heart damage, but the mechanism underlying this association remains unclear. Apaf-1-interacting protein (APIP) and ADORA2B transcript levels in human hearts are substantially higher in patients with heart failure than in controls. Interestingly, the APIP and ADORA2B mRNA levels are highly correlated with each other (R = 0.912). APIP expression was significantly increased in primary neonatal cardiomyocytes under hypoxic conditions and this induction reduced myocardial cell death via the activation of the AKT-HIF1α pathway. Accordingly, infarct sizes of APIP transgenic mice after left anterior descending artery ligation were significantly reduced compared to those of wild-type mice. Strikingly, knockdown of APIP expression impaired the cytoprotective effects of ADORA2B during hypoxic damage. Immunoprecipitation and proximity ligation assays revealed that APIP interacts with ADORA2B, leading to the stabilization of both proteins by interfering with lysosomal degradation, and to the activation of the downstream PKA-CREB signaling pathways. ADORA2B levels in the hearts of APIPTg/Tg, APIPTg/+, and Apip+/- mice were proportionally downregulated. In addition, ADORA2B D296G derived from the rs200741295 polymorphism failed to bind to APIP and did not exert cardioprotective activity during hypoxia. Moreover, Adora2b D296G knock-in mice were more vulnerable than control mice to myocardial infarction and intentional increases in APIP levels overcame the defective protection of the ADORA2B SNP against ischemic injury. Collectively, APIP is crucial for cardioprotection against myocardial infarction by virtue of binding to and stabilizing ADORA2B, thereby dampening ischemic heart injury.
Collapse
|
23
|
Gwon Y, Kim SH, Kim HT, Kam TI, Park J, Lim B, Cha H, Chang HJ, Hong YR, Jung YK. Amelioration of amyloid β-FcγRIIb neurotoxicity and tau pathologies by targeting LYN. FASEB J 2018; 33:4300-4313. [PMID: 30540497 DOI: 10.1096/fj.201800926r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
SRC-family kinases (SFKs) have been implicated in Alzheimer's disease (AD), but their mode of action was scarcely understood. Here, we show that LYN plays an essential role in amyloid β (Aβ)-triggered neurotoxicity and tau hyperphosphorylation by phosphorylating Fcγ receptor IIb2 (FcγRIIb2). We found that enzyme activity of LYN was increased in the brain of AD patients and was promoted in neuronal cells exposed to Aβ 1-42 (Aβ1-42). Knockdown of LYN expression inhibited Aβ1-42-induced neuronal cell death. Of note, LYN interacted with FcγRIIb2 upon exposure to Aβ1-42 and phosphorylated FcγRIIb2 at Tyr273 within immunoreceptor tyrosine-based inhibitory motif in neuronal cells. With the use of the structure-based drug design, we isolated KICG2576, an ATP-competitive inhibitor of LYN. Determination of cocrystal structure illustrated that KICG2576 bound to the cleft in the LYN kinase domain and inhibited LYN with a half-maximal inhibitory concentration value of 0.15 μM. KICG2576 inhibited Aβ- or FcγRIIb2-induced cell death, and this effect was better than pyrazolopyrimidine 1, a widely used inhibitor of SFK. Upon exposure to Aβ, KICG2576 blocked the phosphorylation of FcγRIIb2 and translocation of phosphatidylinositol 3,4,5-trisphosphate 5-phosphatase 2, a binding protein to the phosphorylated FcγRIIb2, to the plasma membrane, resulting in the inhibition of tau hyperphosphorylation, the downstream event of Aβ1-42-FcγRIIb2 binding. Furthermore, intracerebroventricular injection of KICG2576 into mice ameliorated Aβ-induced memory impairment. These results suggest that LYN plays a crucial role in Aβ1-42-mediated neurotoxicity and tau pathology, providing a therapeutic potential of LYN in AD.-Gwon, Y., Kim, S.-H., Kim, H. T., Kam, T.-I., Park, J., Lim, B., Cha, H., Chang, H.-J., Hong, Y. R., Jung, Y.-K. Amelioration of amyloid β-FcγRIIb neurotoxicity and tau pathologies by targeting LYN.
Collapse
Affiliation(s)
- Youngdae Gwon
- School of Biological Sciences, Seoul National University, Seoul, South Korea; and
| | - Seo-Hyun Kim
- School of Biological Sciences, Seoul National University, Seoul, South Korea; and
| | - Hyun Tae Kim
- Crystalgenomics Incorporated, Gyeonggi-do, South Korea
| | - Tae-In Kam
- School of Biological Sciences, Seoul National University, Seoul, South Korea; and
| | - Jisu Park
- School of Biological Sciences, Seoul National University, Seoul, South Korea; and
| | - Bitna Lim
- School of Biological Sciences, Seoul National University, Seoul, South Korea; and
| | - Hyunju Cha
- Crystalgenomics Incorporated, Gyeonggi-do, South Korea
| | - Ho-Jin Chang
- Crystalgenomics Incorporated, Gyeonggi-do, South Korea
| | - Yong Rae Hong
- Crystalgenomics Incorporated, Gyeonggi-do, South Korea
| | - Yong-Keun Jung
- School of Biological Sciences, Seoul National University, Seoul, South Korea; and
| |
Collapse
|