1
|
Awasthi P, Kumar D, Hasan S. Role of 14-3-3 protein family in the pathobiology of EBV in immortalized B cells and Alzheimer's disease. Front Mol Biosci 2024; 11:1353828. [PMID: 39144488 PMCID: PMC11322100 DOI: 10.3389/fmolb.2024.1353828] [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: 12/11/2023] [Accepted: 07/03/2024] [Indexed: 08/16/2024] Open
Abstract
Background and Aims Several studies have revealed that Epstein-Barr virus (EBV) infection raised the likelihood of developing Alzheimer's disease (AD) via infecting B lymphocytes. The purpose of the current investigation was to assess the possible association between EBV infection and AD. Methods The microarray datasets GSE49628, GSE126379, GSE122063, and GSE132903 were utilized to extract DEGs by using the GEO2R tool of the GEO platform. The STRING tool was used to determine the interaction between the DEGs, and Cytoscape was used to visualize the results. The DEGs that were found underwent function analysis, including pathway and GO, using the DAVID 2021 and ClueGo/CluePedia. By using MNC, MCC, Degree, and Radiality of cytoHubba, we identified seven common key genes. Gene co-expression analysis was performed through the GeneMANIA web tool. Furthermore, expression analysis of key genes was performed through GTEx software, which have been identified in various human brain regions. The miRNA-gene interaction was performed through the miRNet v 2.0 tool. DsigDB on the Enrichr platform was utilized to extract therapeutic drugs connected to key genes. Results In GEO2R analysis of datasets with |log2FC|≥ 0.5 and p-value <0.05, 8386, 10,434, 7408, and 759 genes were identified. A total of 141 common DEGs were identified by combining the extracted genes of different datasets. A total of 141 nodes and 207 edges were found during the PPI analysis. The DEG GO analysis with substantial alterations disclosed that they are associated to molecular functions and biological processes, such as positive regulation of neuron death, autophagy regulation of mitochondrion, response of cell to insulin stimulus, calcium signaling regulation, organelle transport along microtubules, protein kinase activity, and phosphoserine binding. Kyoto Encyclopedia of Genes and Genomes analysis discovered the correlation between the DEGs in pathways of neurodegeneration: multiple disease, cell cycle, and cGMP-PKG signaling pathway. Finally, YWHAH, YWHAG, YWHAB, YWHAZ, MAP2K1, PPP2CA, and TUBB genes were identified that are strongly linked to EBV and AD. Three miRNAs, i.e., hsa-mir-15a-5p, hsa-let-7a-5p, and hsa-mir-7-5p, were identified to regulate most of hub genes that are associated with EBV and AD. Further top 10 significant therapeutic drugs were predicted. Conclusion We have discovered new biomarkers and therapeutic targets for AD, as well as the possible biological mechanisms whereby infection with EBV may be involved in AD susceptibility for the first time.
Collapse
Affiliation(s)
- Prankur Awasthi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| | - Dhruv Kumar
- School of Health Sciences and Technology, UPES University Dehradun, Dehradun, India
| | - Saba Hasan
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, India
| |
Collapse
|
2
|
Ganne A, Mainali N, Balasubramaniam M, Atluri R, Pahal S, Asante J, Nagel C, Vallurupalli S, Shmookler Reis RJ, Ayyadevara S. Ezetimibe Lowers Risk of Alzheimer's and Related Dementias over Sevenfold, Reducing Aggregation in Model Systems by Inhibiting 14-3-3G::Hexokinase Interaction. AGING BIOLOGY 2024; 2:20240028. [PMID: 39263528 PMCID: PMC11389752 DOI: 10.59368/agingbio.20240028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Numerous factors predispose to progression of cognitive impairment to Alzheimer's disease and related dementias (ADRD), most notably age, APOE(ε4) alleles, traumatic brain injury, heart disease, hypertension, obesity/diabetes, and Down's syndrome. Protein aggregation is diagnostic for neurodegenerative diseases, and may be causal through promotion of chronic neuroinflammation. We isolated aggregates from postmortem hippocampi of ADRD patients, heart-disease patients, and age-matched controls. Aggregates, characterized by high-resolution proteomics (with or without crosslinking), were significantly elevated in heart-disease and ADRD hippocampi. Hexokinase-1 (HK1) and 14-3-3G/γ proteins, previously implicated in neuronal signaling and neurodegeneration, are especially enriched in ADRD and heart-disease aggregates vs. controls (each P<0.008), and their interaction was implied by extensive crosslinking in both disease groups. Screening the hexokinase-1::14-3-3G interface with FDA-approved drug structures predicted strong affinity for ezetimibe, a benign cholesterol-lowering medication. Diverse cultured human-cell and whole-nematode models of ADRD aggregation showed that this drug potently disrupts HK1::14-3-3G adhesion, reduces disease-associated aggregation, and activates autophagy. Mining clinical databases supports drug reduction of ADRD risk, decreasing it to 0.14 overall (P<0.0001; 95% C.I. 0.06-0.34), and <0.12 in high-risk heart-disease subjects (P<0.006). These results suggest that drug disruption of the 14-3-3G::HK1 interface blocks an early "lynchpin" adhesion, prospectively reducing aggregate accrual and progression of ADRD.
Collapse
Affiliation(s)
- Akshatha Ganne
- Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205
| | - Nirjal Mainali
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock AR 72205
- Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205
| | | | - Ramani Atluri
- Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205
| | - Sonu Pahal
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock AR 72205
- Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205
| | - Joseph Asante
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock AR 72205
- Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205
| | - Corey Nagel
- College of Nursing, University of Arkansas for Medical Sciences, Little Rock AR 72205
| | - Srikanth Vallurupalli
- Central Arkansas Veterans Healthcare System, Little Rock AR 72205
- Department of Internal Medicine, Division of Cardiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Robert J Shmookler Reis
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock AR 72205
- Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205
- Central Arkansas Veterans Healthcare System, Little Rock AR 72205
| | - Srinivas Ayyadevara
- Bioinformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock AR 72205
- Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock AR 72205
- Central Arkansas Veterans Healthcare System, Little Rock AR 72205
| |
Collapse
|
3
|
Sultana R, Butterfield DA. Protein Oxidation in Aging and Alzheimer's Disease Brain. Antioxidants (Basel) 2024; 13:574. [PMID: 38790679 PMCID: PMC11117785 DOI: 10.3390/antiox13050574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 04/28/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Proteins are essential molecules that play crucial roles in maintaining cellular homeostasis and carrying out biological functions such as catalyzing biochemical reactions, structural proteins, immune response, etc. However, proteins also are highly susceptible to damage by reactive oxygen species (ROS) and reactive nitrogen species (RNS). In this review, we summarize the role of protein oxidation in normal aging and Alzheimer's disease (AD). The major emphasis of this review article is on the carbonylation and nitration of proteins in AD and mild cognitive impairment (MCI). The oxidatively modified proteins showed a strong correlation with the reported changes in brain structure, carbohydrate metabolism, synaptic transmission, cellular energetics, etc., of both MCI and AD brains compared to the controls. Some proteins were found to be common targets of oxidation and were observed during the early stages of AD, suggesting that those changes might be critical in the onset of symptoms and/or formation of the pathological hallmarks of AD. Further studies are required to fully elucidate the role of protein oxidation and nitration in the progression and pathogenesis of AD.
Collapse
Affiliation(s)
- Rukhsana Sultana
- Department of Neuroscience, School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 West Campbell Rd., Richardson, TX 75080, USA;
| | - D. Allan Butterfield
- Department of Chemistry, and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA
| |
Collapse
|
4
|
Crha R, Kozeleková A, Hofrová A, Iľkovičová L, Gašparik N, Kadeřávek P, Hritz J. Hiding in plain sight: Complex interaction patterns between Tau and 14-3-3ζ protein variants. Int J Biol Macromol 2024; 266:130802. [PMID: 38492709 DOI: 10.1016/j.ijbiomac.2024.130802] [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: 12/22/2023] [Revised: 03/05/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
Tau protein is an intrinsically disordered protein that plays a key role in Alzheimer's disease (AD). In brains of AD patients, Tau occurs abnormally phosphorylated and aggregated in neurofibrillary tangles (NFTs). Together with Tau, 14-3-3 proteins - abundant cytosolic dimeric proteins - were found colocalized in the NFTs. However, so far, the molecular mechanism of the process leading to pathological changes in Tau structure as well as the direct involvement of 14-3-3 proteins are not well understood. Here, we aimed to reveal the effects of phosphorylation by protein kinase A (PKA) on Tau structural preferences and provide better insight into the interaction between Tau and 14-3-3 proteins. We also addressed the impact of monomerization-inducing phosphorylation of 14-3-3 at S58 on the binding to Tau protein. Using multidimensional nuclear magnetic resonance spectroscopy (NMR), chemical cross-linking analyzed by mass spectrometry (MS) and PAGE, we unveiled differences in their binding affinity, stoichiometry, and interfaces with single-residue resolution. We revealed that the interaction between 14-3-3 and Tau proteins is mediated not only via the 14-3-3 amphipathic binding grooves, but also via less specific interactions with 14-3-3 protein surface and, in the case of monomeric 14-3-3, also partially via the exposed dimeric interface. In addition, the hyperphosphorylation of Tau changes its affinity to 14-3-3 proteins. In conclusion, we propose quite complex interaction mode between the Tau and 14-3-3 proteins.
Collapse
Affiliation(s)
- Radek Crha
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Aneta Kozeleková
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Alena Hofrová
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Lucia Iľkovičová
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Norbert Gašparik
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Pavel Kadeřávek
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Jozef Hritz
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; Department of Chemistry, Faculty of Science, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| |
Collapse
|
5
|
Varlı M, Bhosle SR, Kim E, Yang Y, Taş İ, Zhou R, Pulat S, Gamage CDB, Park SY, Ha HH, Kim H. Usnic Acid Targets 14-3-3 Proteins and Suppresses Cancer Progression by Blocking Substrate Interaction. JACS AU 2024; 4:1521-1537. [PMID: 38665668 PMCID: PMC11040559 DOI: 10.1021/jacsau.3c00774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024]
Abstract
The anticancer therapeutic effects of usnic acid (UA), a lichen secondary metabolite, have been demonstrated in vitro and in vivo. However, the mechanism underlying the anticancer effect of UA remains to be clarified. In this study, the target protein of UA was identified using a UA-linker-Affi-Gel molecule, which showed that UA binds to the 14-3-3 protein. UA binds to 14-3-3, causing the degradation of proteasomal and autophagosomal proteins. The interaction of UA with 14-3-3 isoforms modulated cell invasion, cell cycle progression, aerobic glycolysis, mitochondrial biogenesis, and the Akt/mTOR, JNK, STAT3, NF-κB, and AP-1 signaling pathways in colorectal cancer. A peptide inhibitor of 14-3-3 blocked or regressed the activity of UA and inhibited its effects. The results suggest that UA binds to 14-3-3 isoforms and suppresses cancer progression by affecting 14-3-3 targets and phosphorylated proteins.
Collapse
Affiliation(s)
- Mücahit Varlı
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Suresh R. Bhosle
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Eunae Kim
- College
of Pharmacy, Chosun University, 146 Chosundae-gil, Gwangju 61452, Republic of Korea
| | - Yi Yang
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - İsa Taş
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Rui Zhou
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Sultan Pulat
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Chathurika D. B. Gamage
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - So-Yeon Park
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Hyung-Ho Ha
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| | - Hangun Kim
- College
of Pharmacy, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Republic of Korea
| |
Collapse
|
6
|
Peddinti V, Avaghade MM, Suthar SU, Rout B, Gomte SS, Agnihotri TG, Jain A. Gut instincts: Unveiling the connection between gut microbiota and Alzheimer's disease. Clin Nutr ESPEN 2024; 60:266-280. [PMID: 38479921 DOI: 10.1016/j.clnesp.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/16/2024] [Indexed: 04/13/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder marked by neuroinflammation and gradual cognitive decline. Recent research has revealed that the gut microbiota (GM) plays an important role in the pathogenesis of AD through the microbiota-gut-brain axis. However, the mechanism by which GM and microbial metabolites alter brain function is not clearly understood. GM dysbiosis increases the permeability of the intestine, alters the blood-brain barrier permeability, and elevates proinflammatory mediators causing neurodegeneration. This review article introduced us to the composition and functions of GM along with its repercussions of dysbiosis in relation to AD. We also discussed the importance of the gut-brain axis and its role in communication. Later we focused on the mechanism behind gut dysbiosis and the progression of AD including neuroinflammation, oxidative stress, and changes in neurotransmitter levels. Furthermore, we highlighted recent developments in AD management, such as microbiota-based therapy, dietary interventions like prebiotics, probiotics, and fecal microbiota transplantation. Finally, we concluded with challenges and future directions in AD research based on GM.
Collapse
Affiliation(s)
- Vasu Peddinti
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Manoj Mohan Avaghade
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Sunil Umedmal Suthar
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Biswajit Rout
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Shyam Sudhakar Gomte
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, 382355, Gujarat, India.
| |
Collapse
|
7
|
Qiang Q, Skudder-Hill L, Toyota T, Huang Z, Wei W, Adachi H. CSF 14-3-3β is associated with progressive cognitive decline in Alzheimer's disease. Brain Commun 2023; 5:fcad312. [PMID: 38035365 PMCID: PMC10684297 DOI: 10.1093/braincomms/fcad312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/22/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023] Open
Abstract
Alzheimer's disease is a neurodegenerative disorder characterized pathologically by amyloid-beta plaques, tau tangles and neuronal loss. In clinical practice, the 14-3-3 isoform beta (β) is a biomarker that aids in the diagnosis of sporadic Creutzfeldt-Jakob disease. Recently, a proteomics study found increased CSF 14-3-3β levels in Alzheimer's disease patients, suggesting a potential link between CSF 14-3-3β and Alzheimer's disease. Our present study aimed to further investigate the role of CSF 14-3-3β in Alzheimer's disease by analysing the data of 719 participants with available CSF 14-3-3β measurements from the Alzheimer's Disease Neuroimaging Initiative. Higher CSF 14-3-3β levels were observed in the mild cognitive impairment group compared to the cognitively normal group, with the highest CSF 14-3-3β levels in the Alzheimer's disease dementia group. This study also found significant associations between CSF 14-3-3β levels and CSF biomarkers of p-tau, t-tau, pTau/Aβ42 ratios and GAP-43, as well as other Alzheimer's disease biomarkers such as Aβ-PET. An early increase in CSF 14-3-3β levels was observed prior to Aβ-PET-positive status, and CSF 14-3-3β levels continued to rise after crossing the Aβ-PET positivity threshold before reaching a plateau. The diagnostic accuracy of CSF 14-3-3β (area under the receiver operating characteristic curve = 0.819) was moderate compared to other established Alzheimer's disease biomarkers in distinguishing cognitively normal Aβ pathology-negative individuals from Alzheimer's disease Aβ pathology-positive individuals. Higher baseline CSF 14-3-3β levels were associated with accelerated cognitive decline, reduced hippocampus volumes and declining fluorodeoxyglucose-PET values over a 4-year follow-up period. Patients with mild cognitive impairment and high CSF 14-3-3β levels at baseline had a significantly increased risk [hazard ratio = 2.894 (1.599-5.238), P < 0.001] of progression to Alzheimer's disease dementia during follow-up. These findings indicate that CSF 14-3-3β may be a potential biomarker for Alzheimer's disease and could provide a more comprehensive understanding of the underlying pathological changes of Alzheimer's disease, as well as aid in the diagnosis and monitoring of disease progression.
Collapse
Affiliation(s)
- Qiang Qiang
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital, Fudan University, 200040 Shanghai, China
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, 807-8555 Kitakyushu, Japan
| | - Loren Skudder-Hill
- Yuquan Hospital, Tsinghua University School of Clinical Medicine, 100084 Beijing, China
- School of Medicine, University of Auckland, 1023 Auckland, New Zealand
| | - Tomoko Toyota
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, 807-8555 Kitakyushu, Japan
| | - Zhe Huang
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, 807-8555 Kitakyushu, Japan
| | - Wenshi Wei
- Department of Neurology, Cognitive Disorders Center, Huadong Hospital, Fudan University, 200040 Shanghai, China
| | - Hiroaki Adachi
- Department of Neurology, University of Occupational and Environmental Health School of Medicine, 807-8555 Kitakyushu, Japan
| | | |
Collapse
|
8
|
Leitner DF, Kanshin E, Faustin A, Thierry M, Friedman D, Devore S, Ueberheide B, Devinsky O, Wisniewski T. Localized proteomic differences in the choroid plexus of Alzheimer's disease and epilepsy patients. Front Neurol 2023; 14:1221775. [PMID: 37521285 PMCID: PMC10379643 DOI: 10.3389/fneur.2023.1221775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/22/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Alzheimer's disease (AD) and epilepsy are reciprocally related. Among sporadic AD patients, clinical seizures occur in 10-22% and subclinical epileptiform abnormalities occur in 22-54%. Cognitive deficits, especially short-term memory impairments, occur in most epilepsy patients. Common neurophysiological and molecular mechanisms occur in AD and epilepsy. The choroid plexus undergoes pathological changes in aging, AD, and epilepsy, including decreased CSF turnover, amyloid beta (Aβ), and tau accumulation due to impaired clearance and disrupted CSF amino acid homeostasis. This pathology may contribute to synaptic dysfunction in AD and epilepsy. Methods We evaluated control (n = 8), severe AD (n = 8; A3, B3, C3 neuropathology), and epilepsy autopsy cases (n = 12) using laser capture microdissection (LCM) followed by label-free quantitative mass spectrometry on the choroid plexus adjacent to the hippocampus at the lateral geniculate nucleus level. Results Proteomics identified 2,459 proteins in the choroid plexus. At a 5% false discovery rate (FDR), 616 proteins were differentially expressed in AD vs. control, 1 protein in epilepsy vs. control, and 438 proteins in AD vs. epilepsy. There was more variability in the epilepsy group across syndromes. The top 20 signaling pathways associated with differentially expressed proteins in AD vs. control included cell metabolism pathways; activated fatty acid beta-oxidation (p = 2.00 x 10-7, z = 3.00), and inhibited glycolysis (p = 1.00 x 10-12, z = -3.46). For AD vs. epilepsy, the altered pathways included cell metabolism pathways, activated complement system (p = 5.62 x 10-5, z = 2.00), and pathogen-induced cytokine storm (p = 2.19 x 10-2, z = 3.61). Of the 617 altered proteins in AD and epilepsy vs. controls, 497 (81%) were positively correlated (p < 0.0001, R2 = 0.27). Discussion We found altered signaling pathways in the choroid plexus of severe AD cases and many correlated changes in the protein expression of cell metabolism pathways in AD and epilepsy cases. The shared molecular mechanisms should be investigated further to distinguish primary pathogenic changes from the secondary ones. These mechanisms could inform novel therapeutic strategies to prevent disease progression or restore normal function. A focus on dual-diagnosed AD/epilepsy cases, specific epilepsy syndromes, such as temporal lobe epilepsy, and changes across different severity levels in AD and epilepsy would add to our understanding.
Collapse
Affiliation(s)
- Dominique F. Leitner
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, United States
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Evgeny Kanshin
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, United States
| | - Arline Faustin
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
| | - Manon Thierry
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Daniel Friedman
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Sasha Devore
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Beatrix Ueberheide
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Proteomics Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, United States
| | - Orrin Devinsky
- Comprehensive Epilepsy Center, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
| | - Thomas Wisniewski
- Center for Cognitive Neurology, Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Neurology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, New York University Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
9
|
Liu YQ, Liang CQ, Chen ZW, Hu J, Hu JJ, Luo YY, Chen YX, Li YM. 14-3-3ζ Participates in the Phase Separation of Phosphorylated and Glycated Tau and Modulates the Physiological and Pathological Functions of Tau. ACS Chem Neurosci 2023; 14:1220-1225. [PMID: 36939323 DOI: 10.1021/acschemneuro.3c00034] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
Tau plays a major role in Alzheimer's disease (AD) and several other neurodegenerative diseases. Tau undergoing liquid-liquid phase separation (LLPS) performs specific physiological functions, induces pathological processes, and contributes to neurodegeneration. Regulating Tau phase separation helps maintain physiological functions of Tau and inhibits pathological aggregation. Here, we show that the 14-3-3 zeta isoform (14-3-3ζ) participates in Tau LLPS. 14-3-3ζ can undergo co-phase separation with WT Tau, participate in and stabilize Tau droplets, and inhibit Tau droplet-driven tubulin assembly. On the other hand, 14-3-3ζ disrupts the LLPS of phosphorylated and glycated Tau, thereby inhibiting the amyloid aggregation initiated by LLPS.
Collapse
Affiliation(s)
- Yu-Qing Liu
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Chu-Qiao Liang
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Zhi-Wei Chen
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Jun Hu
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Jin-Jian Hu
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Yun-Yi Luo
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Yong-Xiang Chen
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China
| | - Yan-Mei Li
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, P. R. China.,Beijing Institute for Brain Disorders, Beijing 100069, P. R. China.,Center for Synthetic and System Biology, Tsinghua University, Beijing 100084, P. R. China
| |
Collapse
|
10
|
Betters RK, Luhmann E, Gottschalk AC, Xu Z, Shin MR, Ptak CP, Fiock KL, Radoshevich LC, Hefti MM. Characterization of the Tau Interactome in Human Brain Reveals Isoform-Dependent Interaction with 14-3-3 Family Proteins. eNeuro 2023; 10:ENEURO.0503-22.2023. [PMID: 36898832 PMCID: PMC10035768 DOI: 10.1523/eneuro.0503-22.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/20/2023] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
Despite exhibiting tau phosphorylation similar to Alzheimer's disease (AD), the human fetal brain is remarkably resilient to tau aggregation and toxicity. To identify potential mechanisms for this resilience, we used co-immunoprecipitation (co-IP) with mass spectrometry to characterize the tau interactome in human fetal, adult, and Alzheimer's disease brains. We found significant differences between the tau interactome in fetal and AD brain tissue, with little difference between adult and AD, although these findings are limited by the low throughput and small sample size of these experiments. Differentially interacting proteins were enriched for 14-3-3 domains, and we found that the 14-3-3-β, η, and γ isoforms interacted with phosphorylated tau in Alzheimer's disease but not the fetal brain. Since long isoform (4R) tau is only seen in the adult brain and this is one of the major differences between fetal and AD tau, we tested the ability of our strongest hit (14-3-3-β) to interact with 3R and 4R tau using co-immunoprecipitation, mass photometry, and nuclear magnetic resonance (NMR). We found that 14-3-3-β interacts preferentially with phosphorylated 4R tau, forming a complex consisting of two 14-3-3-β molecules to one tau. By NMR, we mapped 14-3-3 binding regions on tau that span the second microtubule binding repeat, which is unique to 4R tau. Our findings suggest that there are isoform-driven differences between the phospho-tau interactome in fetal and Alzheimer's disease brain, including differences in interaction with the critical 14-3-3 family of protein chaperones, which may explain, in part, the resilience of fetal brain to tau toxicity.
Collapse
Affiliation(s)
- Ryan K Betters
- Department of Pathology
- Interdisciplinary Neuroscience Graduate Program
| | | | | | - Zhen Xu
- Protein and Crystallography Facility
| | - Mallory R Shin
- Department of Pathology
- Interdisciplinary Neuroscience Graduate Program
| | | | | | | | - Marco M Hefti
- Department of Pathology
- Interdisciplinary Neuroscience Graduate Program
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA
| |
Collapse
|
11
|
Cho E, Park J, Hwang EM, Kim HW, Park JY. 14-3-3γ haploinsufficiency leads to altered dopamine pathway and Parkinson's disease-like motor incoordination in mice. Mol Brain 2023; 16:2. [PMID: 36604743 PMCID: PMC9817279 DOI: 10.1186/s13041-022-00990-z] [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: 06/21/2022] [Accepted: 12/19/2022] [Indexed: 01/07/2023] Open
Abstract
The 14-3-3 protein family with seven isoforms found in mammals is widely expressed in the brain and plays various roles in cellular processes. Several studies have reported that 14-3-3γ, one of the 14-3-3 protein isoforms, is associated with neurological and psychiatric disorders, but the role of 14-3-3γ in the pathophysiology of brain diseases is unclear. Although studies have been conducted on the relationship between 14-3-3γ protein and Parkinson's disease (PD), a common neurodegenerative disorder with severe motor symptoms such as bradykinesia and rigidity, a direct connection remains to be elucidated. We recently showed that adult heterozygous 14-3-3γ knockout mice are hyperactive and exhibit anxiety-like behavior. In this study, we further characterized the molecular and behavioral changes in aged 14-3-3γ heterozygous mice to investigate the role of 14-3-3γ in the brain. We observed decreased dopamine levels and altered dopamine metabolism in the brains of these mice, including changes in the phosphorylation of proteins implicated in PD pathology. Furthermore, we confirmed that they displayed PD symptom-like behavioral deficits, such as impaired motor coordination and decreased ability to the nest-building activity. These findings suggest an association between 14-3-3γ dysfunction and PD pathophysiology.
Collapse
Affiliation(s)
- Eunsil Cho
- grid.222754.40000 0001 0840 2678Department of Integrated Biomedical and Life Sciences, Korea University, Seoul, 02708 Korea ,grid.222754.40000 0001 0840 2678BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, 02841 Korea
| | - Jinsil Park
- grid.263333.40000 0001 0727 6358College of Life Sciences, Sejong University, Seoul, 05006 Korea
| | - Eun Mi Hwang
- grid.35541.360000000121053345Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, 02792 Korea
| | - Hyung Wook Kim
- grid.263333.40000 0001 0727 6358College of Life Sciences, Sejong University, Seoul, 05006 Korea
| | - Jae-Yong Park
- grid.222754.40000 0001 0840 2678Department of Integrated Biomedical and Life Sciences, Korea University, Seoul, 02708 Korea ,grid.222754.40000 0001 0840 2678BK21FOUR R&E Center for Learning Health Systems, Korea University, Seoul, 02841 Korea ,ASTRION, Seoul, 02842 Korea
| |
Collapse
|
12
|
Chen S, Chang Y, Li L, Acosta D, Li Y, Guo Q, Wang C, Turkes E, Morrison C, Julian D, Hester ME, Scharre DW, Santiskulvong C, Song SX, Plummer JT, Serrano GE, Beach TG, Duff KE, Ma Q, Fu H. Spatially resolved transcriptomics reveals genes associated with the vulnerability of middle temporal gyrus in Alzheimer's disease. Acta Neuropathol Commun 2022; 10:188. [PMID: 36544231 PMCID: PMC9773466 DOI: 10.1186/s40478-022-01494-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/11/2022] [Indexed: 12/24/2022] Open
Abstract
Human middle temporal gyrus (MTG) is a vulnerable brain region in early Alzheimer's disease (AD), but little is known about the molecular mechanisms underlying this regional vulnerability. Here we utilize the 10 × Visium platform to define the spatial transcriptomic profile in both AD and control (CT) MTG. We identify unique marker genes for cortical layers and the white matter, and layer-specific differentially expressed genes (DEGs) in human AD compared to CT. Deconvolution of the Visium spots showcases the significant difference in particular cell types among cortical layers and the white matter. Gene co-expression analyses reveal eight gene modules, four of which have significantly altered co-expression patterns in the presence of AD pathology. The co-expression patterns of hub genes and enriched pathways in the presence of AD pathology indicate an important role of cell-cell-communications among microglia, oligodendrocytes, astrocytes, and neurons, which may contribute to the cellular and regional vulnerability in early AD. Using single-molecule fluorescent in situ hybridization, we validated the cell-type-specific expression of three novel DEGs (e.g., KIF5A, PAQR6, and SLC1A3) and eleven previously reported DEGs associated with AD pathology (i.e., amyloid beta plaques and intraneuronal neurofibrillary tangles or neuropil threads) at the single cell level. Our results may contribute to the understanding of the complex architecture and neuronal and glial response to AD pathology of this vulnerable brain region.
Collapse
Affiliation(s)
- Shuo Chen
- grid.261331.40000 0001 2285 7943Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH 43210 USA ,grid.261331.40000 0001 2285 7943Biomedical Sciences Graduate Program, Ohio State University, Columbus, OH 43210 USA
| | - Yuzhou Chang
- grid.261331.40000 0001 2285 7943Biomedical Sciences Graduate Program, Ohio State University, Columbus, OH 43210 USA ,grid.261331.40000 0001 2285 7943Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Liangping Li
- grid.261331.40000 0001 2285 7943Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Diana Acosta
- grid.261331.40000 0001 2285 7943Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Yang Li
- grid.261331.40000 0001 2285 7943Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Qi Guo
- grid.261331.40000 0001 2285 7943Biomedical Sciences Graduate Program, Ohio State University, Columbus, OH 43210 USA ,grid.261331.40000 0001 2285 7943Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Cankun Wang
- grid.261331.40000 0001 2285 7943Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Emir Turkes
- grid.83440.3b0000000121901201UK Dementia Research Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Cody Morrison
- grid.261331.40000 0001 2285 7943Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Dominic Julian
- grid.240344.50000 0004 0392 3476The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Mark E. Hester
- grid.240344.50000 0004 0392 3476The Steve and Cindy Rasmussen Institute for Genomic Medicine, Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205 USA
| | - Douglas W. Scharre
- grid.261331.40000 0001 2285 7943Department of Neurology, Center for Cognitive and Memory Disorders, Center for Neuromodulation, Ohio State University, Columbus, OH 43210 USA
| | - Chintda Santiskulvong
- grid.50956.3f0000 0001 2152 9905Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Sarah XueYing Song
- grid.50956.3f0000 0001 2152 9905Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Jasmine T. Plummer
- grid.50956.3f0000 0001 2152 9905Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Geidy E. Serrano
- grid.414208.b0000 0004 0619 8759Banner Sun Health Research Institute, Sun City, AZ 85351 USA
| | - Thomas G. Beach
- grid.414208.b0000 0004 0619 8759Banner Sun Health Research Institute, Sun City, AZ 85351 USA
| | - Karen E. Duff
- grid.83440.3b0000000121901201UK Dementia Research Institute, UCL Queen Square Institute of Neurology, London, UK
| | - Qin Ma
- grid.261331.40000 0001 2285 7943Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Hongjun Fu
- grid.261331.40000 0001 2285 7943Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| |
Collapse
|
13
|
Obsilova V, Obsil T. Structural insights into the functional roles of 14-3-3 proteins. Front Mol Biosci 2022; 9:1016071. [PMID: 36188227 PMCID: PMC9523730 DOI: 10.3389/fmolb.2022.1016071] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/02/2022] [Indexed: 12/02/2022] Open
Abstract
Signal transduction cascades efficiently transmit chemical and/or physical signals from the extracellular environment to intracellular compartments, thereby eliciting an appropriate cellular response. Most often, these signaling processes are mediated by specific protein-protein interactions involving hundreds of different receptors, enzymes, transcription factors, and signaling, adaptor and scaffolding proteins. Among them, 14-3-3 proteins are a family of highly conserved scaffolding molecules expressed in all eukaryotes, where they modulate the function of other proteins, primarily in a phosphorylation-dependent manner. Through these binding interactions, 14-3-3 proteins participate in key cellular processes, such as cell-cycle control, apoptosis, signal transduction, energy metabolism, and protein trafficking. To date, several hundreds of 14-3-3 binding partners have been identified, including protein kinases, phosphatases, receptors and transcription factors, which have been implicated in the onset of various diseases. As such, 14-3-3 proteins are promising targets for pharmaceutical interventions. However, despite intensive research into their protein-protein interactions, our understanding of the molecular mechanisms whereby 14-3-3 proteins regulate the functions of their binding partners remains insufficient. This review article provides an overview of the current state of the art of the molecular mechanisms whereby 14-3-3 proteins regulate their binding partners, focusing on recent structural studies of 14-3-3 protein complexes.
Collapse
Affiliation(s)
- Veronika Obsilova
- Institute of Physiology of the Czech Academy of Sciences, Laboratory of Structural Biology of Signaling Proteins, Division BIOCEV, Vestec, Czechia
- *Correspondence: Veronika Obsilova, ; Tomas Obsil,
| | - Tomas Obsil
- Department of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Prague, Czechia
- *Correspondence: Veronika Obsilova, ; Tomas Obsil,
| |
Collapse
|
14
|
Lu Y. Early increase of cerebrospinal fluid 14-3-3ζ protein in the alzheimer's disease continuum. Front Aging Neurosci 2022; 14:941927. [PMID: 35966774 PMCID: PMC9372587 DOI: 10.3389/fnagi.2022.941927] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe earlier research has shown that the 14-3-3ζ is increased in neurofibrillary tangles (NFTs) of human Alzheimer's disease (AD) brains and stimulates the tau phosphorylation. Cerebrospinal fluid (CSF) 14-3-3ζ along the AD continuum remains to be explored.MethodsWe analyzed 113 cognitive normal (CN) controls, 372 patients with mild cognitive impairment (MCI), and 225 patients with AD dementia from the Alzheimer's Disease Neuroimaging Initiative database. CSF 14-3-3ζ protein was measured by Mass Spectrometry.ResultsWe observed higher CSF 14-3-3ζ in the MCI group vs. the CN group and in the AD group vs. the MCI or CN group. The 14-3-3ζ was able to distinguish AD from CN and MCI. High 14-3-3ζ predicted conversion from MCI to AD. In CSF, phosphorylated tau at threonine 181 and total-tau were associated with 14-3-3ζ in MCI and AD groups, and beta-amyloid (Aβ) 42 correlated with 14-3-3ζ in the MCI group. Baseline high 14-3-3ζ was associated with cognitive decline, brain atrophy, glucose hypometabolism, and Aβ deposition in MCI and AD at baseline and follow-up.ConclusionOur findings revealed the potential diagnostic and prognostic utility of CSF 14-3-3ζ in the AD continuum. The 14-3-3ζ could be a promising therapeutic target for the intervention of AD.
Collapse
|
15
|
Herod SG, Dyatel A, Hodapp S, Jovanovic M, Berchowitz LE. Clearance of an amyloid-like translational repressor is governed by 14-3-3 proteins. Cell Rep 2022; 39:110753. [PMID: 35508136 PMCID: PMC9156962 DOI: 10.1016/j.celrep.2022.110753] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/24/2022] [Accepted: 04/06/2022] [Indexed: 11/13/2022] Open
Abstract
Amyloids are fibrous protein aggregates associated with age-related diseases. While these aggregates are typically described as irreversible and pathogenic, some cells use reversible amyloid-like structures that serve important functions. The RNA-binding protein Rim4 forms amyloid-like assemblies that are essential for translational control during Saccharomyces cerevisiae meiosis. Rim4 amyloid-like assemblies are disassembled in a phosphorylation-dependent manner at meiosis II onset. By investigating Rim4 clearance, we elucidate co-factors that mediate clearance of amyloid-like assemblies in a physiological setting. We demonstrate that yeast 14-3-3 proteins bind to Rim4 assemblies and facilitate their subsequent phosphorylation and timely clearance. Furthermore, distinct 14-3-3 proteins play non-redundant roles in facilitating phosphorylation and clearance of amyloid-like Rim4. Additionally, we find that 14-3-3 proteins contribute to global protein aggregate homeostasis. Based on the role of 14-3-3 proteins in aggregate homeostasis and their interactions with disease-associated assemblies, we propose that these proteins may protect against pathological protein aggregates.
Collapse
Affiliation(s)
- S Grace Herod
- Department of Genetics and Development, Hammer Health Sciences Center, Columbia University Irving Medical Center, New York, NY, USA; Taub Institute for Research on Alzheimer's and the Aging Brain, New York, NY, USA
| | - Annie Dyatel
- Department of Genetics and Development, Hammer Health Sciences Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Stefanie Hodapp
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Marko Jovanovic
- Department of Biological Sciences, Columbia University, New York, NY, USA
| | - Luke E Berchowitz
- Department of Genetics and Development, Hammer Health Sciences Center, Columbia University Irving Medical Center, New York, NY, USA; Taub Institute for Research on Alzheimer's and the Aging Brain, New York, NY, USA.
| |
Collapse
|
16
|
Han Y, Ye H, Li P, Zeng Y, Yang J, Gao M, Su Z, Huang Y. In vitro characterization and molecular dynamics simulation reveal mechanism of 14-3-3ζ regulated phase separation of the tau protein. Int J Biol Macromol 2022; 208:1072-1081. [PMID: 35381286 DOI: 10.1016/j.ijbiomac.2022.03.215] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/10/2022] [Accepted: 03/31/2022] [Indexed: 11/29/2022]
Abstract
As a major microtubule-associated protein, tau is involved in the assembly of microtubules in the central nervous system. However, under pathological conditions tau assembles into amyloid filaments. Liquid droplets formed by liquid-liquid phase separation (LLPS) are a recently identified assembly state of tau and may have a major effect on the physiological function of tau and the formation of tau aggregates. 14-3-3 proteins are ubiquitously expressed in various tissues and regulate a wide variety of biological processes. In this work, we demonstrate that 14-3-3ζ is recruited into tau droplets and regulates tau LLPS by in vitro assays. While the mobility of tau molecules inside the droplets is not affected in the presence of 14-3-3ζ, the amount and size of droplets can vary significantly. Mechanistic studies reveal that 14-3-3ζ regulates tau LLPS by electrostatic interactions and hydrophobic interactions with the proline-rich domain and the microtubule-binding domain of tau. Surprisingly, the disordered C-terminal tail rather than the amphipathic binding groove of 14-3-3ζ plays a key role. Our findings not only provide a novel dimension to understand the interactions between 14 and 3-3 proteins and tau, but also suggest that 14-3-3 proteins may play an important role in regulating the LLPS of their binding partners.
Collapse
Affiliation(s)
- Yue Han
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; Department of Biological Engineering, Hubei University of Technology, Wuhan 430068, China
| | - Haiqiong Ye
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; Department of Biological Engineering, Hubei University of Technology, Wuhan 430068, China
| | - Ping Li
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; Department of Biological Engineering, Hubei University of Technology, Wuhan 430068, China
| | - Yifan Zeng
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; Department of Biological Engineering, Hubei University of Technology, Wuhan 430068, China
| | - Jing Yang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; Department of Biological Engineering, Hubei University of Technology, Wuhan 430068, China
| | - Meng Gao
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; Department of Biological Engineering, Hubei University of Technology, Wuhan 430068, China.
| | - Zhengding Su
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; Department of Biological Engineering, Hubei University of Technology, Wuhan 430068, China
| | - Yongqi Huang
- Key Laboratory of Industrial Fermentation (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan 430068, China; Department of Biological Engineering, Hubei University of Technology, Wuhan 430068, China.
| |
Collapse
|
17
|
Sandebring-Matton A, Axenhus M, Bogdanovic N, Winblad B, Schedin-Weiss S, Nilsson P, Tjernberg LO. Microdissected Pyramidal Cell Proteomics of Alzheimer Brain Reveals Alterations in Creatine Kinase B-Type, 14-3-3-γ, and Heat Shock Cognate 71. Front Aging Neurosci 2021; 13:735334. [PMID: 34867272 PMCID: PMC8641652 DOI: 10.3389/fnagi.2021.735334] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/18/2021] [Indexed: 11/30/2022] Open
Abstract
Novel insights on proteins involved in Alzheimer’s disease (AD) are needed. Since multiple cell types and matrix components are altered in AD, bulk analysis of brain tissue maybe difficult to interpret. In the current study, we isolated pyramidal cells from the cornu ammonis 1 (CA1) region of the hippocampus from five AD and five neurologically healthy donors using laser capture microdissection (LCM). The samples were analyzed by proteomics using 18O-labeled internal standard and nano-high-performance liquid chromatography coupled to tandem mass spectrometry (HPLC-MS/MS) for relative quantification. Fold change between AD and control was calculated for the proteins that were identified in at least two individual proteomes from each group. From the 10 cases analyzed, 62 proteins were identified in at least two AD cases and two control cases. Creatine kinase B-type (CKB), 14-3-3-γ, and heat shock cognate 71 (Hsc71), which have not been extensively studied in the context of the human AD brain previously, were selected for further studies by immunohistochemistry (IHC). In hippocampus, semi-quantitative measures of IHC staining of the three proteins confirmed the findings from our proteomic analysis. Studies of the same proteins in the frontal cortex revealed that the alterations remained for CKB and 14-3-3-γ but not for Hsc71. Protein upregulation in CA1 neurons of final stage AD is either a result of detrimental, pathological effects, or from cell-specific protective response mechanisms in surviving neurons. Based on previous findings from experimental studies, CKB and Hsc71 likely exhibit protective effects, whereas 14-3-3-γ may represent a detrimental pathway. These new players could reflect pathways of importance for the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Anna Sandebring-Matton
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Ageing Epidemiology (AGE) Research Unit, School of Public Health, Imperial College London, London, United Kingdom
| | - Michael Axenhus
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Nenad Bogdanovic
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Sophia Schedin-Weiss
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Per Nilsson
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Lars O Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden.,Clinical Chemistry, Karolinska University Hospital, Solna, Sweden
| |
Collapse
|
18
|
Kitoka K, Skrabana R, Gasparik N, Hritz J, Jaudzems K. NMR Studies of Tau Protein in Tauopathies. Front Mol Biosci 2021; 8:761227. [PMID: 34859051 PMCID: PMC8632555 DOI: 10.3389/fmolb.2021.761227] [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: 08/19/2021] [Accepted: 10/25/2021] [Indexed: 11/13/2022] Open
Abstract
Tauopathies, including Alzheimer's disease (AD), are the most troublesome of all age-related chronic conditions, as there are no well-established disease-modifying therapies for their prevention and treatment. Spatio-temporal distribution of tau protein pathology correlates with cognitive decline and severity of the disease, therefore, tau protein has become an appealing target for therapy. Current knowledge of the pathological effects and significance of specific species in the tau aggregation pathway is incomplete although more and more structural and mechanistic insights are being gained using biophysical techniques. Here, we review the application of NMR to structural studies of various tau forms that appear in its aggregation process, focusing on results obtained from solid-state NMR. Furthermore, we discuss implications from these studies and their prospective contribution to the development of new tauopathy therapies.
Collapse
Affiliation(s)
- Kristine Kitoka
- Laboratory of Physical Organic Chemistry, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Rostislav Skrabana
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Norbert Gasparik
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Faculty of Science, National Centre for Biomolecular Research, Masaryk University, Brno, Czech Republic
| | - Jozef Hritz
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Chemistry, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Kristaps Jaudzems
- Laboratory of Physical Organic Chemistry, Latvian Institute of Organic Synthesis, Riga, Latvia
- Faculty of Chemistry, University of Latvia, Riga, Latvia
| |
Collapse
|
19
|
Jin M, Jin X, Homma H, Fujita K, Tanaka H, Murayama S, Akatsu H, Tagawa K, Okazawa H. Prediction and verification of the AD-FTLD common pathomechanism based on dynamic molecular network analysis. Commun Biol 2021; 4:961. [PMID: 34385591 PMCID: PMC8361101 DOI: 10.1038/s42003-021-02475-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 07/23/2021] [Indexed: 12/12/2022] Open
Abstract
Multiple gene mutations cause familial frontotemporal lobar degeneration (FTLD) while no single gene mutations exists in sporadic FTLD. Various proteins aggregate in variable regions of the brain, leading to multiple pathological and clinical prototypes. The heterogeneity of FTLD could be one of the reasons preventing development of disease-modifying therapy. We newly develop a mathematical method to analyze chronological changes of PPI networks with sequential big data from comprehensive phosphoproteome of four FTLD knock-in (KI) mouse models (PGRNR504X-KI, TDP43N267S-KI, VCPT262A-KI and CHMP2BQ165X-KI mice) together with four transgenic mouse models of Alzheimer's disease (AD) and with APPKM670/671NL-KI mice at multiple time points. The new method reveals the common core pathological network across FTLD and AD, which is shared by mouse models and human postmortem brains. Based on the prediction, we performed therapeutic intervention of the FTLD models, and confirmed amelioration of pathologies and symptoms of four FTLD mouse models by interruption of the core molecule HMGB1, verifying the new mathematical method to predict dynamic molecular networks.
Collapse
Affiliation(s)
- Meihua Jin
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Xiaocen Jin
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Hidenori Homma
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan.
| | - Kyota Fujita
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Hikari Tanaka
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Shigeo Murayama
- Department of Neuropathology, Brain Bank for Aging Research, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo, Japan
- Brain Bank for Neurodevelopmental, Neurological and Psychiatric Disorders, Molecular Research Center for Children's Mental Development, United Graduate School of Child Development, Osaka University, Suita, Osaka, Japan
| | - Hiroyasu Akatsu
- Department of Medicine for Aging in Place and Community-Based Medical Education, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Kazuhiko Tagawa
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Hitoshi Okazawa
- Department of Neuropathology, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan.
- Center for Brain Integration Research, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
20
|
Fang Y, He Q, Cao J. Targeted protein degradation and regulation with molecular glue: past and recent discoveries. Curr Med Chem 2021; 29:2490-2503. [PMID: 34365941 DOI: 10.2174/0929867328666210806113949] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/10/2021] [Accepted: 06/15/2021] [Indexed: 11/22/2022]
Abstract
The evolution in research and clinical settings of targeted therapies has been inspired by the progress of cancer chemotherapy to use small molecules and monoclonal antibodies for targeting specific disease-associated genes and proteins for noninfectious chronic diseases. In addition to conventional protein inhibition and activation strategies as drug discovery modalities, new methods of targeted protein degradation and regulation using molecular glues have become an attractive approach for drug discovery. Mechanistically, molecular glues trigger interactions between the proteins that originally did not interact by forming ternary complexes as protein-protein interaction (PPI) modulators. New molecular glues and their mechanisms of action have been actively investigated in the past decades. An immunomodulatory imide drug, thalidomide, and its derivatives have been used in the clinic and are a class of molecular glue that induces degradation of several neo-substrates. In this review, we summarize the development of molecular glues and share our opinions on the identification of novel molecular glues in an attempt to promote the concept and inspire further investigations.
Collapse
Affiliation(s)
- Yizheng Fang
- College of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing. China
| | - Qiaojun He
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou. China
| | - Ji Cao
- The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou. China
| |
Collapse
|
21
|
Underwood R, Gannon M, Pathak A, Kapa N, Chandra S, Klop A, Yacoubian TA. 14-3-3 mitigates alpha-synuclein aggregation and toxicity in the in vivo preformed fibril model. Acta Neuropathol Commun 2021; 9:13. [PMID: 33413679 PMCID: PMC7792107 DOI: 10.1186/s40478-020-01110-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/19/2020] [Indexed: 12/26/2022] Open
Abstract
Alpha-synuclein (αsyn) is the key component of proteinaceous aggregates termed Lewy Bodies that pathologically define a group of disorders known as synucleinopathies, including Parkinson's Disease (PD) and Dementia with Lewy Bodies. αSyn is hypothesized to misfold and spread throughout the brain in a prion-like fashion. Transmission of αsyn necessitates the release of misfolded αsyn from one cell and the uptake of that αsyn by another, in which it can template the misfolding of endogenous αsyn upon cell internalization. 14-3-3 proteins are a family of highly expressed brain proteins that are neuroprotective in multiple PD models. We have previously shown that 14-3-3θ acts as a chaperone to reduce αsyn aggregation, cell-to-cell transmission, and neurotoxicity in the in vitro pre-formed fibril (PFF) model. In this study, we expanded our studies to test the impact of 14-3-3s on αsyn toxicity in the in vivo αsyn PFF model. We used both transgenic expression models and adenovirus associated virus (AAV)-mediated expression to examine whether 14-3-3 manipulation impacts behavioral deficits, αsyn aggregation, and neuronal counts in the PFF model. 14-3-3θ transgene overexpression in cortical and amygdala regions rescued social dominance deficits induced by PFFs at 6 months post injection, whereas 14-3-3 inhibition by transgene expression of the competitive 14-3-3 peptide inhibitor difopein in the cortex and amygdala accelerated social dominance deficits. The behavioral rescue by 14-3-3θ overexpression was associated with delayed αsyn aggregation induced by PFFs in these brain regions. Conversely, 14-3-3 inhibition by difopein in the cortex and amygdala accelerated αsyn aggregation and reduction in NECAB1-positive neuron counts induced by PFFs. 14-3-3θ overexpression by AAV in the substantia nigra (SN) also delayed αsyn aggregation in the SN and partially rescued PFF-induced reduction in tyrosine hydroxylase (TH)-positive dopaminergic cells in the SN. 14-3-3 inhibition in the SN accelerated nigral αsyn aggregation and enhanced PFF-induced reduction in TH-positive dopaminergic cells. These data indicate a neuroprotective role for 14-3-3θ against αsyn toxicity in vivo.
Collapse
Affiliation(s)
- Rachel Underwood
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294 USA
- Center for Neurodegenerative Disease Research, Perelman School of Medicine at the University of Pennsylvania, Maloney Building, 3rd Floor, 3600 Spruce Street, Philadelphia, PA 19104-2676 USA
| | - Mary Gannon
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Aneesh Pathak
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Navya Kapa
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Sidhanth Chandra
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294 USA
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, IL 60611 USA
| | - Alyssa Klop
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Talene A. Yacoubian
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294 USA
- Civitan International Research Center, Room 510A, 1719 Sixth Avenue South, Birmingham, AL 35294 USA
| |
Collapse
|
22
|
Abstract
14-3-3 proteins are mostly expressed in the brain and are closely involved in numerous brain functions and various brain disorders. Among the isotypes of the 14-3-3 proteins, 14-3-3γ is mainly expressed in neurons and is highly produced during brain development, which could indicate that it has a significance in neural development. Furthermore, the distinctive levels of temporally and locally regulated 14-3-3γ expression in various brain disorders suggest that it could play a substantial role in brain plasticity of the diseased states. In this review, we introduce the various brain disorders reported to be involved with 14-3-3γ, and summarize the changes of 14-3-3γ expression in each brain disease. We also discuss the potential of 14-3-3γ for treatment and the importance of research on specific 14-3-3 isotypes for an effective therapeutic approach.
Collapse
Affiliation(s)
- Eunsil Cho
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul 02708, Korea
| |
Collapse
|
23
|
Neves JF, Petrvalská O, Bosica F, Cantrelle FX, Merzougui H, O'Mahony G, Hanoulle X, Obšil T, Landrieu I. Phosphorylated full-length Tau interacts with 14-3-3 proteins via two short phosphorylated sequences, each occupying a binding groove of 14-3-3 dimer. FEBS J 2020; 288:1918-1934. [PMID: 32979285 DOI: 10.1111/febs.15574] [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] [Received: 11/04/2019] [Revised: 08/07/2020] [Accepted: 09/07/2020] [Indexed: 01/22/2023]
Abstract
Protein-protein interactions (PPIs) remain poorly explored targets for the treatment of Alzheimer's disease. The interaction of 14-3-3 proteins with Tau was shown to be linked to Tau pathology. This PPI is therefore seen as a potential target for Alzheimer's disease. When Tau is phosphorylated by PKA (Tau-PKA), several phosphorylation sites are generated, including two known 14-3-3 binding sites, surrounding the phosphorylated serines 214 and 324 of Tau. The crystal structures of 14-3-3 in complex with peptides surrounding these Tau phosphosites show that both these motifs are anchored in the amphipathic binding groove of 14-3-3. However, in the absence of structural data with the full-length Tau protein, the stoichiometry of the complex or the interface and affinity of the partners is still unclear. In this work, we addressed these points, using a broad range of biophysical techniques. The interaction of the long and disordered Tau-PKA protein with 14-3-3σ is restricted to two short sequences, containing phosphorylated serines, which bind in the amphipathic binding groove of 14-3-3σ. Phosphorylation of Tau is fundamental for the formation of this stable complex, and the affinity of the Tau-PKA/14-3-3σ interaction is in the 1-10 micromolar range. Each monomer of the 14-3-3σ dimer binds one of two different phosphorylated peptides of Tau-PKA, suggesting a 14-3-3/Tau-PKA stoichiometry of 2 : 1, confirmed by analytical ultracentrifugation. These results contribute to a better understanding of this PPI and provide useful insights for drug discovery projects aiming at the modulation of this interaction.
Collapse
Affiliation(s)
- João Filipe Neves
- CNRS ERL9002 Integrative Structural Biology, Lille, France.,Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Univ. Lille, Lille, France
| | - Olivia Petrvalská
- Department of Structural Biology of Signaling Proteins, Division BIOCEV, Institute of Physiology of the Czech Academy of Sciences, Vestec, Czech Republic.,Department of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Francesco Bosica
- Medicinal Chemistry, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.,Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, The Netherlands
| | - François-Xavier Cantrelle
- CNRS ERL9002 Integrative Structural Biology, Lille, France.,Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Univ. Lille, Lille, France
| | - Hamida Merzougui
- CNRS ERL9002 Integrative Structural Biology, Lille, France.,Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Univ. Lille, Lille, France
| | - Gavin O'Mahony
- Medicinal Chemistry, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Xavier Hanoulle
- CNRS ERL9002 Integrative Structural Biology, Lille, France.,Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Univ. Lille, Lille, France
| | - Tomáš Obšil
- Department of Structural Biology of Signaling Proteins, Division BIOCEV, Institute of Physiology of the Czech Academy of Sciences, Vestec, Czech Republic.,Department of Physical and Macromolecular Chemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Isabelle Landrieu
- CNRS ERL9002 Integrative Structural Biology, Lille, France.,Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Risk Factors and Molecular Determinants of Aging-Related Diseases, Univ. Lille, Lille, France
| |
Collapse
|
24
|
Ke PC, Zhou R, Serpell LC, Riek R, Knowles TPJ, Lashuel HA, Gazit E, Hamley IW, Davis TP, Fändrich M, Otzen DE, Chapman MR, Dobson CM, Eisenberg DS, Mezzenga R. Half a century of amyloids: past, present and future. Chem Soc Rev 2020; 49:5473-5509. [PMID: 32632432 PMCID: PMC7445747 DOI: 10.1039/c9cs00199a] [Citation(s) in RCA: 304] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Amyloid diseases are global epidemics with profound health, social and economic implications and yet remain without a cure. This dire situation calls for research into the origin and pathological manifestations of amyloidosis to stimulate continued development of new therapeutics. In basic science and engineering, the cross-β architecture has been a constant thread underlying the structural characteristics of pathological and functional amyloids, and realizing that amyloid structures can be both pathological and functional in nature has fuelled innovations in artificial amyloids, whose use today ranges from water purification to 3D printing. At the conclusion of a half century since Eanes and Glenner's seminal study of amyloids in humans, this review commemorates the occasion by documenting the major milestones in amyloid research to date, from the perspectives of structural biology, biophysics, medicine, microbiology, engineering and nanotechnology. We also discuss new challenges and opportunities to drive this interdisciplinary field moving forward.
Collapse
Affiliation(s)
- Pu Chun Ke
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Zhongshan Hospital, Fudan University, 111 Yixueyuan Rd, Xuhui District, Shanghai, China
| | - Ruhong Zhou
- Institute of Quantitative Biology, Zhejiang University, Hangzhou 310058, China; Department of Chemistry, Columbia University, New York, New York, 10027, USA
| | - Louise C. Serpell
- School of Life Sciences, University of Sussex, Falmer, East Sussex BN1 9QG, UK
| | - Roland Riek
- Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, ETH Zurich, Wolfgang-Pauli-Str. 10, 8093 Zurich, Switzerland
| | - Tuomas P. J. Knowles
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
- Cavendish Laboratory, University of Cambridge, J J Thomson Avenue, CB3 0HE, Cambridge, UK
| | - Hilal A. Lashuel
- Laboratory of Molecular Neurobiology and Neuroproteomics, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Ehud Gazit
- Department of Molecular Microbiology and Biotechnology, George S. Wise Faculty of Life Sciences; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Ian W. Hamley
- School of Chemistry, Food Biosciences and Pharmacy, University of Reading, Whiteknights, Reading RG6 6AD, UK
| | - Thomas P. Davis
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane Qld 4072, Australia
| | - Marcus Fändrich
- Institute of Protein Biochemistry, Ulm University, 89081, Ulm, Germany
| | - Daniel Erik Otzen
- Department of Molecular Biology, Center for Insoluble Protein Structures (inSPIN), Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | - Matthew R. Chapman
- Department of Molecular, Cellular and Developmental Biology, Centre for Microbial Research, University of Michigan, Ann Arbor, MI 48109-1048, USA
| | - Christopher M. Dobson
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK
| | - David S. Eisenberg
- Departments of Chemistry and Biochemistry and Biological Chemistry, UCLA-DOE Institute and Howard Hughes Medical Institute, UCLA, Los Angeles, CA, USA
| | - Raffaele Mezzenga
- Department of Health Science & Technology, ETH Zurich, Schmelzbergstrasse 9, LFO, E23, 8092 Zurich, Switzerland
- Department of Materials, ETH Zurich, Wolfgang Pauli Strasse 10, 8093 Zurich, Switzerland
| |
Collapse
|
25
|
Chaudhuri P, Prajapati KP, Anand BG, Dubey K, Kar K. Amyloid cross-seeding raises new dimensions to understanding of amyloidogenesis mechanism. Ageing Res Rev 2019; 56:100937. [PMID: 31430565 DOI: 10.1016/j.arr.2019.100937] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/21/2019] [Accepted: 07/23/2019] [Indexed: 12/12/2022]
Abstract
Hallmarks of most of the amyloid pathologies are surprisingly found to be heterocomponent entities such as inclusions and plaques which contain diverse essential proteins and metabolites. Experimental studies have already revealed the occurrence of coaggregation and cross-seeding during amyloid formation of several proteins and peptides, yielding multicomponent assemblies of amyloid nature. Further, research reports on the co-occurrence of more than one type of amyloid-linked pathologies in the same individual suggest the possible cross-talk among the disease related amyloidogenic protein species during their amyloid growth. In this review paper, we have tried to gain more insight into the process of coaggregation and cross-seeding during amyloid aggregation of proteins, particularly focusing on their relevance to the pathogenesis of the protein misfolding diseases. Revelation of amyloid cross-seeding and coaggregation seems to open new dimensions in our mechanistic understanding of amyloidogenesis and such knowledge may possibly inspire better designing of anti-amyloid therapeutics.
Collapse
|
26
|
Butterfield DA, Boyd-Kimball D. Redox proteomics and amyloid β-peptide: insights into Alzheimer disease. J Neurochem 2019; 151:459-487. [PMID: 30216447 PMCID: PMC6417976 DOI: 10.1111/jnc.14589] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/15/2018] [Accepted: 09/07/2018] [Indexed: 12/12/2022]
Abstract
Alzheimer disease (AD) is a progressive neurodegenerative disorder associated with aging and characterized pathologically by the presence of senile plaques, neurofibrillary tangles, and neurite and synapse loss. Amyloid beta-peptide (1-42) [Aβ(1-42)], a major component of senile plaques, is neurotoxic and induces oxidative stress in vitro and in vivo. Redox proteomics has been used to identify proteins oxidatively modified by Aβ(1-42) in vitro and in vivo. In this review, we discuss these proteins in the context of those identified to be oxidatively modified in animal models of AD, and human studies including familial AD, pre-clinical AD (PCAD), mild cognitive impairment (MCI), early AD, late AD, Down syndrome (DS), and DS with AD (DS/AD). These redox proteomics studies indicate that Aβ(1-42)-mediated oxidative stress occurs early in AD pathogenesis and results in altered antioxidant and cellular detoxification defenses, decreased energy yielding metabolism and mitochondrial dysfunction, excitotoxicity, loss of synaptic plasticity and cell structure, neuroinflammation, impaired protein folding and degradation, and altered signal transduction. Improved access to biomarker imaging and the identification of lifestyle interventions or treatments to reduce Aβ production could be beneficial in preventing or delaying the progression of AD. This article is part of the special issue "Proteomics".
Collapse
Affiliation(s)
- D. Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| | - Debra Boyd-Kimball
- Department of Chemistry and Biochemistry, University of Mount Union, Alliance, OH 44601
| |
Collapse
|
27
|
Gu Q, Cuevas E, Raymick J, Kanungo J, Sarkar S. Downregulation of 14-3-3 Proteins in Alzheimer’s Disease. Mol Neurobiol 2019; 57:32-40. [DOI: 10.1007/s12035-019-01754-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 08/29/2019] [Indexed: 01/03/2023]
|
28
|
Barbier P, Zejneli O, Martinho M, Lasorsa A, Belle V, Smet-Nocca C, Tsvetkov PO, Devred F, Landrieu I. Role of Tau as a Microtubule-Associated Protein: Structural and Functional Aspects. Front Aging Neurosci 2019; 11:204. [PMID: 31447664 PMCID: PMC6692637 DOI: 10.3389/fnagi.2019.00204] [Citation(s) in RCA: 282] [Impact Index Per Article: 56.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/18/2019] [Indexed: 12/24/2022] Open
Abstract
Microtubules (MTs) play a fundamental role in many vital processes such as cell division and neuronal activity. They are key structural and functional elements in axons, supporting neurite differentiation and growth, as well as transporting motor proteins along the axons, which use MTs as support tracks. Tau is a stabilizing MT associated protein, whose functions are mainly regulated by phosphorylation. A disruption of the MT network, which might be caused by Tau loss of function, is observed in a group of related diseases called tauopathies, which includes Alzheimer’s disease (AD). Tau is found hyperphosphorylated in AD, which might account for its loss of MT stabilizing capacity. Since destabilization of MTs after dissociation of Tau could contribute to toxicity in neurodegenerative diseases, a molecular understanding of this interaction and its regulation is essential.
Collapse
Affiliation(s)
- Pascale Barbier
- Fac Pharm, Aix Marseille Univ., Centre National de la Recherche Scientifique (CNRS), Inst Neurophysiopathol (INP), Fac Pharm, Marseille, France
| | - Orgeta Zejneli
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), Lille, France.,Univ. Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), CHU-Lille, UMR-S 1172, Centre de Recherche Jean-Pierre AUBERT (JPArc), Lille, France
| | - Marlène Martinho
- Aix Marseille Univ., Centre National de la Recherche Scientifique (CNRS), UMR 7281, Bioénergétique et Ingénierie des Protéines (BIP), Marseille, France
| | - Alessia Lasorsa
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), Lille, France
| | - Valérie Belle
- Aix Marseille Univ., Centre National de la Recherche Scientifique (CNRS), UMR 7281, Bioénergétique et Ingénierie des Protéines (BIP), Marseille, France
| | - Caroline Smet-Nocca
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), Lille, France
| | - Philipp O Tsvetkov
- Fac Pharm, Aix Marseille Univ., Centre National de la Recherche Scientifique (CNRS), Inst Neurophysiopathol (INP), Fac Pharm, Marseille, France
| | - François Devred
- Fac Pharm, Aix Marseille Univ., Centre National de la Recherche Scientifique (CNRS), Inst Neurophysiopathol (INP), Fac Pharm, Marseille, France
| | - Isabelle Landrieu
- Univ. Lille, Centre National de la Recherche Scientifique (CNRS), UMR 8576, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF), Lille, France
| |
Collapse
|
29
|
Ke YD, Chan G, Stefanoska K, Au C, Bi M, Müller J, Przybyla M, Feiten A, Prikas E, Halliday GM, Piguet O, Kiernan MC, Kassiou M, Hodges JR, Loy CT, Mattick JS, Ittner A, Kril JJ, Sutherland GT, Ittner LM. CNS cell type-specific gene profiling of P301S tau transgenic mice identifies genes dysregulated by progressive tau accumulation. J Biol Chem 2019; 294:14149-14162. [PMID: 31366728 DOI: 10.1074/jbc.ra118.005263] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 07/24/2019] [Indexed: 12/20/2022] Open
Abstract
The microtubule-associated protein tau undergoes aberrant modification resulting in insoluble brain deposits in various neurodegenerative diseases, including frontotemporal dementia (FTD), progressive supranuclear palsy, and corticobasal degeneration. Tau aggregates can form in different cell types of the central nervous system (CNS) but are most prevalent in neurons. We have previously recapitulated aspects of human FTD in mouse models by overexpressing mutant human tau in CNS neurons, including a P301S tau variant in TAU58/2 mice, characterized by early-onset and progressive behavioral deficits and FTD-like neuropathology. The molecular mechanisms underlying the functional deficits of TAU58/2 mice remain mostly elusive. Here, we employed functional genomics (i.e. RNAseq) to determine differentially expressed genes in young and aged TAU58/2 mice to identify alterations in cellular processes that may contribute to neuropathy. We identified genes in cortical brain samples differentially regulated between young and old TAU58/2 mice relative to nontransgenic littermates and by comparative analysis with a dataset of CNS cell type-specific genes expressed in nontransgenic mice. Most differentially-regulated genes had known or putative roles in neurons and included presynaptic and excitatory genes. Specifically, we observed changes in presynaptic factors, glutamatergic signaling, and protein scaffolding. Moreover, in the aged mice, expression levels of several genes whose expression was annotated to occur in other brain cell types were altered. Immunoblotting and immunostaining of brain samples from the TAU58/2 mice confirmed altered expression and localization of identified and network-linked proteins. Our results have revealed genes dysregulated by progressive tau accumulation in an FTD mouse model.
Collapse
Affiliation(s)
- Yazi D Ke
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Gabriella Chan
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Kristie Stefanoska
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Carol Au
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Mian Bi
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Julius Müller
- The Jenner Institute, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford OX3 7DQ, United Kingdom
| | - Magdalena Przybyla
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Astrid Feiten
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Emmanuel Prikas
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales 2005, Australia
| | - Olivier Piguet
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales 2005, Australia.,School of Psychology, University of Sydney, Sydney, New South Wales 2005, Australia.,ARC Centre of Excellence in Cognition and Its Disorders, University of Sydney, Sydney, New South Wales 2005, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales 2005, Australia.,Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Sydney, New South Wales 2005, Australia
| | - Michael Kassiou
- School of Chemistry, University of Sydney, Sydney, New South Wales 2005, Australia
| | - John R Hodges
- Brain and Mind Centre, University of Sydney, Sydney, New South Wales 2005, Australia
| | - Clement T Loy
- Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, New South Wales 2010, Australia.,Sydney School of Public Health, University of Sydney, New South Wales 2006, Australia
| | - John S Mattick
- Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.,St. Vincent's Clinical School, Faculty of Medicine, University of New South Wales, New South Wales 2010, Australia
| | - Arne Ittner
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Jillian J Kril
- Charles Perkins Centre and Discipline of Pathology, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales 2005, Australia
| | - Greg T Sutherland
- Charles Perkins Centre and Discipline of Pathology, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales 2005, Australia
| | - Lars M Ittner
- Dementia Research Centre and Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, New South Wales 2109, Australia
| |
Collapse
|
30
|
Zhang KQ, Wen HS, Li JF, Qi X, Fan HY, Zhang XY, Tian Y, Liu Y, Wang HL, Li Y. 14-3-3 gene family in spotted sea bass (Lateolabrax maculatus): Genome-wide identification, phylogenetic analysis and expression profiles after salinity stress. Comp Biochem Physiol A Mol Integr Physiol 2019; 235:1-11. [PMID: 31082484 DOI: 10.1016/j.cbpa.2019.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 05/03/2019] [Accepted: 05/06/2019] [Indexed: 12/23/2022]
Abstract
The tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation (14-3-3) proteins are a group of highly conserved homologous and heterologous proteins involved in a wild range of physiological processes, including the regulation of many molecular phenomena under different environmental salinities. In this study, we identified eleven 14-3-3 genes from the spotted sea bass (Lateolabrax maculatus) genome and transcriptomic databases and verified their identities by conducting phylogenetic, syntenic and gene structure analyses. The spotted sea bass 14-3-3 genes are highly conserved based on sequence alignment, conserved domains and motifs, and tertiary structural feature. Quantitative real-time polymerase chain reaction (qRT-PCR) analysis of 14-3-3 genes in gill of spotted sea bass under normal physiological conditions indicated that the expression level of 14-3-3 zeta was the highest among tested genes, followed by 14-3-3 theta. Furthermore, expression profiles of 14-3-3 genes in gill tissue (in vivo and in vitro) indicated that the 14-3-3 zeta and 14-3-3 theta genes were significantly induced by different environmental salinities in spotted sea bass, suggesting their potential involvement in response to salinity challenge. Our findings may lay the foundation for future functional studies on the 14-3-3 gene family in euryhaline teleosts.
Collapse
Affiliation(s)
- Kai-Qiang Zhang
- College of Fisheries, Ocean University of China, Qingdao, China; Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao, China
| | - Hai-Shen Wen
- College of Fisheries, Ocean University of China, Qingdao, China; Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao, China
| | - Ji-Fang Li
- College of Fisheries, Ocean University of China, Qingdao, China; Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao, China
| | - Xin Qi
- College of Fisheries, Ocean University of China, Qingdao, China; Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao, China
| | - Hong-Ying Fan
- College of Fisheries, Ocean University of China, Qingdao, China; Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao, China
| | - Xiao-Yan Zhang
- College of Fisheries, Ocean University of China, Qingdao, China; Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao, China
| | - Yuan Tian
- College of Fisheries, Ocean University of China, Qingdao, China; Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao, China
| | - Yang Liu
- College of Fisheries, Ocean University of China, Qingdao, China; Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao, China
| | - Hao-Long Wang
- College of Fisheries, Ocean University of China, Qingdao, China; Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao, China
| | - Yun Li
- College of Fisheries, Ocean University of China, Qingdao, China; Key Laboratory of Mariculture (Ocean University of China), Ministry of Education, Qingdao, China.
| |
Collapse
|
31
|
Smani D, Sarkar S, Raymick J, Kanungo J, Paule MG, Gu Q. Downregulation of 14-3-3 Proteins in a Kainic Acid-Induced Neurotoxicity Model. Mol Neurobiol 2019; 55:122-129. [PMID: 28840498 DOI: 10.1007/s12035-017-0724-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The 14-3-3 proteins are among the most abundant proteins expressed in the brain, comprising about 1% of the total amount of soluble brain proteins. Through phosphoserine- and phosphothreonine-binding motifs, 14-3-3 proteins regulate many signaling proteins and cellular processes including cell death. In the present study, we utilized a well-known kainic acid (KA)-induced excitotoxicity rat model and examined the expression of 14-3-3 and its isoforms in the frontal cortex of KA-treated and control animals. Among the different 14-3-3 isoforms, abundant levels of eta and tau were detected in the frontal cortex, followed by sigma, epsilon, and gamma, while the expression levels of alpha/beta and zeta/delta isoforms were low. Compared to the control animals, KA treatment induced a significant downregulation of the overall 14-3-3 protein level as well as the levels of the abundant isoforms eta, tau, epsilon, and gamma. We also investigated two 14-3-3-interacting proteins that are involved in the cell death process: Bcl-2-associated X (BAX) and extracellular signal-regulated kinase (ERK). Both BAX and phosphorylated ERK showed increased levels following KA treatment. Together, these findings demonstrate an abundance of several 14-3-3 isoforms in the frontal cortex and that KA treatment can cause a downregulation of 14-3-3 expression and an upregulation of 14-3-3-interacting proteins BAX and phospho-ERK. Thus, downregulation of 14-3-3 proteins could be one of the early molecular events associated with excitotoxicity. This could lead to subsequent upregulation of 14-3-3-binding proteins such as BAX and phospho-ERK that contribute to further downstream apoptosis processes, eventually leading to cell death. Maintaining sufficient levels of 14-3-3 expression and function may become a target of therapeutic intervention for excitotoxicity-induced neurodegeneration.
Collapse
Affiliation(s)
- Danyal Smani
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA
| | - Sumit Sarkar
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA
| | - James Raymick
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA
| | - Jyotshna Kanungo
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA
| | - Merle G Paule
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA
| | - Qiang Gu
- Division of Neurotoxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Rd, Jefferson, AR, 72079, USA.
| |
Collapse
|
32
|
14-3-3/Tau Interaction and Tau Amyloidogenesis. J Mol Neurosci 2019; 68:620-630. [PMID: 31062171 DOI: 10.1007/s12031-019-01325-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 04/22/2019] [Indexed: 01/02/2023]
Abstract
The major function of microtubule-associated protein tau is to promote microtubule assembly in the central nervous system. However, aggregation of abnormally phosphorylated tau is a hallmark of tauopathies. Although the molecular mechanisms of conformational transitions and assembling of tau molecules into amyloid fibril remain largely unknown, several factors have been shown to promote tau aggregation, including mutations, polyanions, phosphorylation, and interactions with other proteins. 14-3-3 proteins are a family of highly conserved, multifunctional proteins that are mainly expressed in the central nervous system. Being a scaffolding protein, 14-3-3 proteins interact with tau and regulate tau phosphorylation by bridging tau with various protein kinases. 14-3-3 proteins also directly regulate tau aggregation via specific and non-specific interactions with tau. In this review, we summarize recent advances in characterization of tau conformation and tau/14-3-3 interaction. We discuss the connection between 14-3-3 binding and tau aggregation with a special emphasis on the regulatory role of 14-3-3 on tau conformation.
Collapse
|
33
|
Sluchanko NN, Bustos DM. Intrinsic disorder associated with 14-3-3 proteins and their partners. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 166:19-61. [PMID: 31521232 DOI: 10.1016/bs.pmbts.2019.03.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Protein-protein interactions (PPIs) mediate a variety of cellular processes and form complex networks, where connectivity is achieved owing to the "hub" proteins whose interaction with multiple protein partners is facilitated by the intrinsically disordered protein regions (IDPRs) and posttranslational modifications (PTMs). Universal regulatory proteins of the eukaryotic 14-3-3 family nicely exemplify these concepts and are the focus of this chapter. The extremely wide interactome of 14-3-3 proteins is characterized by high levels of intrinsic disorder (ID) enabling protein phosphorylation and consequent specific binding to the well-structured 14-3-3 dimers, one of the first phosphoserine/phosphothreonine binding modules discovered. However, high ID enrichment also challenges structural studies, thereby limiting the progress in the development of small molecule modulators of the key 14-3-3 PPIs of increased medical importance. Besides the well-known structural flexibility of their variable C-terminal tails, recent studies revealed the strong and conserved ID propensity hidden in the N-terminal segment of 14-3-3 proteins (~40 residues), normally forming the α-helical dimerization region, that may have a potential role for the dimer/monomer dynamics and recently reported moonlighting chaperone-like activity of these proteins. We review the role of ID in the 14-3-3 structure, their interactome, and also in selected 14-3-3 complexes. In addition, we discuss approaches that, in the future, may help minimize the disproportion between the large amount of known 14-3-3 partners and the small number of 14-3-3 complexes characterized with atomic precision, to unleash the whole potential of 14-3-3 PPIs as drug targets.
Collapse
Affiliation(s)
- Nikolai N Sluchanko
- A.N. Bach Institute of Biochemistry, Federal Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation; Department of Biophysics, Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russian Federation.
| | - Diego M Bustos
- Instituto de Histología y Embriología (IHEM) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), CC56, Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo (UNCuyo), Mendoza, Argentina
| |
Collapse
|
34
|
Wakabayashi K, Umahara T, Hirokawa K, Hanyu H, Uchihara T. 14-3-3 protein sigma isoform co-localizes with phosphorylated α-synuclein in Lewy bodies and Lewy neurites in patients with Lewy body disease. Neurosci Lett 2018; 674:171-175. [DOI: 10.1016/j.neulet.2018.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/22/2018] [Accepted: 03/06/2018] [Indexed: 01/12/2023]
|
35
|
Stevers LM, Sijbesma E, Botta M, MacKintosh C, Obsil T, Landrieu I, Cau Y, Wilson AJ, Karawajczyk A, Eickhoff J, Davis J, Hann M, O'Mahony G, Doveston RG, Brunsveld L, Ottmann C. Modulators of 14-3-3 Protein-Protein Interactions. J Med Chem 2017; 61:3755-3778. [PMID: 28968506 PMCID: PMC5949722 DOI: 10.1021/acs.jmedchem.7b00574] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
Direct
interactions between proteins are essential for the regulation
of their functions in biological pathways. Targeting the complex network
of protein–protein interactions (PPIs) has now been widely
recognized as an attractive means to therapeutically intervene in
disease states. Even though this is a challenging endeavor and PPIs
have long been regarded as “undruggable” targets, the
last two decades have seen an increasing number of successful examples
of PPI modulators, resulting in growing interest in this field. PPI
modulation requires novel approaches and the integrated efforts of
multiple disciplines to be a fruitful strategy. This perspective focuses
on the hub-protein 14-3-3, which has several hundred identified protein
interaction partners, and is therefore involved in a wide range of
cellular processes and diseases. Here, we aim to provide an integrated
overview of the approaches explored for the modulation of 14-3-3 PPIs
and review the examples resulting from these efforts in both inhibiting
and stabilizing specific 14-3-3 protein complexes by small molecules,
peptide mimetics, and natural products.
Collapse
Affiliation(s)
- Loes M Stevers
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS) , Eindhoven University of Technology , P.O. Box 513, 5600 MB , Eindhoven , The Netherlands
| | - Eline Sijbesma
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS) , Eindhoven University of Technology , P.O. Box 513, 5600 MB , Eindhoven , The Netherlands
| | - Maurizio Botta
- Department of Biotechnology, Chemistry and Pharmacy , University of Siena , Via Aldo Moro 2 , 53100 Siena , Italy
| | - Carol MacKintosh
- Division of Cell and Developmental Biology, School of Life Sciences , University of Dundee , Dundee DD1 4HN , United Kingdom
| | - Tomas Obsil
- Department of Physical and Macromolecular Chemistry, Faculty of Science , Charles University , Prague 116 36 , Czech Republic
| | | | - Ylenia Cau
- Department of Biotechnology, Chemistry and Pharmacy , University of Siena , Via Aldo Moro 2 , 53100 Siena , Italy
| | - Andrew J Wilson
- School of Chemistry , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , United Kingdom.,Astbury Center For Structural Molecular Biology , University of Leeds , Woodhouse Lane , Leeds LS2 9JT , United Kingdom
| | | | - Jan Eickhoff
- Lead Discovery Center GmbH , Dortmund 44227 , Germany
| | - Jeremy Davis
- UCB Celltech , 216 Bath Road , Slough SL1 3WE , United Kingdom
| | - Michael Hann
- GlaxoSmithKline , Gunnels Wood Road , Stevenage, Hertfordshire SG1 2NY , United Kingdom
| | - Gavin O'Mahony
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit , AstraZeneca Gothenburg , Pepparedsleden 1 , SE-431 83 Mölndal , Sweden
| | - Richard G Doveston
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS) , Eindhoven University of Technology , P.O. Box 513, 5600 MB , Eindhoven , The Netherlands
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS) , Eindhoven University of Technology , P.O. Box 513, 5600 MB , Eindhoven , The Netherlands
| | - Christian Ottmann
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS) , Eindhoven University of Technology , P.O. Box 513, 5600 MB , Eindhoven , The Netherlands.,Department of Chemistry , University of Duisburg-Essen , Universitätstraße 7 , 45141 Essen , Germany
| |
Collapse
|
36
|
Bacterial co-expression of human Tau protein with protein kinase A and 14-3-3 for studies of 14-3-3/phospho-Tau interaction. PLoS One 2017; 12:e0178933. [PMID: 28575131 PMCID: PMC5456370 DOI: 10.1371/journal.pone.0178933] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 05/22/2017] [Indexed: 11/19/2022] Open
Abstract
Abundant regulatory 14-3-3 proteins have an extremely wide interactome and coordinate multiple cellular events via interaction with specifically phosphorylated partner proteins. Notwithstanding the key role of 14-3-3/phosphotarget interactions in many physiological and pathological processes, they are dramatically underexplored. Here, we focused on the 14-3-3 interaction with human Tau protein associated with the development of several neurodegenerative disorders, including Alzheimer’s and Parkinson’s diseases. Among many known phosphorylation sites within Tau, protein kinase A (PKA) phosphorylates several key residues of Tau and induces its tight interaction with 14-3-3 proteins. However, the stoichiometry and mechanism of 14-3-3 interaction with phosphorylated Tau (pTau) are not clearly elucidated. In this work, we describe a simple bacterial co-expression system aimed to facilitate biochemical and structural studies on the 14-3-3/pTau interaction. We show that dual co-expression of human fetal Tau with PKA in Escherichia coli results in multisite Tau phosphorylation including also naturally occurring sites which were not previously considered in the context of 14-3-3 binding. Tau protein co-expressed with PKA displays tight functional interaction with 14-3-3 isoforms of a different type. Upon triple co-expression with 14-3-3 and PKA, Tau protein could be co-purified with 14-3-3 and demonstrates complex which is similar to that formed in vitro between individual 14-3-3 and pTau obtained from dual co-expression. Although used in this study for the specific case of the previously known 14-3-3/pTau interaction, our co-expression system may be useful to study of other selected 14-3-3/phosphotarget interactions and for validations of 14-3-3 complexes identified by other methods.
Collapse
|
37
|
Physiological and Pathological Roles of 15-Deoxy-Δ12,14-Prostaglandin J2 in the Central Nervous System and Neurological Diseases. Mol Neurobiol 2017; 55:2227-2248. [DOI: 10.1007/s12035-017-0435-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 02/03/2017] [Indexed: 12/29/2022]
|
38
|
Sluchanko NN, Gusev NB. Moonlighting chaperone‐like activity of the universal regulatory 14‐3‐3 proteins. FEBS J 2017; 284:1279-1295. [DOI: 10.1111/febs.13986] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 11/20/2016] [Accepted: 12/06/2016] [Indexed: 12/25/2022]
Affiliation(s)
- Nikolai N. Sluchanko
- Laboratory of Structural Biochemistry of Proteins A. N. Bach Institute of Biochemistry Federal Research Center of Biotechnology of the Russian Academy of Sciences Moscow Russia
| | - Nikolai B. Gusev
- Department of Biochemistry School of Biology Moscow State University Russia
| |
Collapse
|
39
|
Mi Z, Halfter W, Abrahamson EE, Klunk WE, Mathis CA, Mufson EJ, Ikonomovic MD. Tenascin-C Is Associated with Cored Amyloid-β Plaques in Alzheimer Disease and Pathology Burdened Cognitively Normal Elderly. J Neuropathol Exp Neurol 2016; 75:868-76. [PMID: 27444354 PMCID: PMC5909866 DOI: 10.1093/jnen/nlw062] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Tenascin-C (TN-C) is an extracellular matrix glycoprotein linked to inflammatory processes in pathological conditions including Alzheimer disease (AD). We examined the distribution of TN-C immunoreactivity (ir) in relation to amyloid-β (Aβ) plaques and vascular Aβ deposits in autopsy brain tissues from 14 patients with clinical and neuropathological AD and 10 aged-matched controls with no cognitive impairment; 5 of the controls had Aβ plaques and 5 did not. TN-C ir was abundant in cortical white matter and subpial cerebral gray matter in all cases, whereas TN-C ir was weak in blood vessels. In all cases with Aβ plaques but not in plaque-free controls, TN-C ir was detected as large (>100 µm in diameter) diffuse extracellular deposits in cortical grey matter. TN-C plaques completely overlapped and surrounded cored Aβ plaques labeled with X-34, a fluorescent derivative of Congo red, and they were associated with reactive astrocytes astrocytes, microglia and phosphorylated tau-containing dystrophic neurites. Diffuse Aβ plaques lacking amyloid cores, reactive glia or dystrophic neurites showed no TN-C ir. In cases with cerebral amyloid angiopathy, TN-C ir in vessel walls did not spread into the surrounding neuropil. These results suggest a role for TN-C in Aβ plaque pathogenesis and its potential as a biomarker and therapy target.
Collapse
Affiliation(s)
- Zhiping Mi
- From the Departments of Neurology (ZM, EEA, WEK, MDI)Department of Neurobiology (WH)Department of Psychiatry (WEK, MDI)Department of Radiology, University of Pittsburgh (CAM)Department of Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System (ZM, EEA, MDI)Department of Neurobiology, Barrow Neurological Institute, Pittsburgh, PA, USA (EJM)
| | - Willi Halfter
- From the Departments of Neurology (ZM, EEA, WEK, MDI)Department of Neurobiology (WH)Department of Psychiatry (WEK, MDI)Department of Radiology, University of Pittsburgh (CAM)Department of Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System (ZM, EEA, MDI)Department of Neurobiology, Barrow Neurological Institute, Pittsburgh, PA, USA (EJM)
| | - Eric E Abrahamson
- From the Departments of Neurology (ZM, EEA, WEK, MDI)Department of Neurobiology (WH)Department of Psychiatry (WEK, MDI)Department of Radiology, University of Pittsburgh (CAM)Department of Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System (ZM, EEA, MDI)Department of Neurobiology, Barrow Neurological Institute, Pittsburgh, PA, USA (EJM)
| | - William E Klunk
- From the Departments of Neurology (ZM, EEA, WEK, MDI)Department of Neurobiology (WH)Department of Psychiatry (WEK, MDI)Department of Radiology, University of Pittsburgh (CAM)Department of Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System (ZM, EEA, MDI)Department of Neurobiology, Barrow Neurological Institute, Pittsburgh, PA, USA (EJM)
| | - Chester A Mathis
- From the Departments of Neurology (ZM, EEA, WEK, MDI)Department of Neurobiology (WH)Department of Psychiatry (WEK, MDI)Department of Radiology, University of Pittsburgh (CAM)Department of Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System (ZM, EEA, MDI)Department of Neurobiology, Barrow Neurological Institute, Pittsburgh, PA, USA (EJM)
| | - Elliott J Mufson
- From the Departments of Neurology (ZM, EEA, WEK, MDI)Department of Neurobiology (WH)Department of Psychiatry (WEK, MDI)Department of Radiology, University of Pittsburgh (CAM)Department of Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System (ZM, EEA, MDI)Department of Neurobiology, Barrow Neurological Institute, Pittsburgh, PA, USA (EJM)
| | - Milos D Ikonomovic
- From the Departments of Neurology (ZM, EEA, WEK, MDI)Department of Neurobiology (WH)Department of Psychiatry (WEK, MDI)Department of Radiology, University of Pittsburgh (CAM)Department of Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System (ZM, EEA, MDI)Department of Neurobiology, Barrow Neurological Institute, Pittsburgh, PA, USA (EJM)
| |
Collapse
|
40
|
Subramanian S, Mahadevan A, Satishchandra P, Shankar SK. Development of a dot blot assay with antibodies to recombinant "core" 14-3-3 protein: Evaluation of its usefulness in diagnosis of Creutzfeldt-Jakob disease. Ann Indian Acad Neurol 2016; 19:205-10. [PMID: 27293331 PMCID: PMC4888683 DOI: 10.4103/0972-2327.176867] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND AND PURPOSE Definitive diagnosis of Creutzfeldt-Jakob disease (CJD) requires demonstration of infective prion protein (PrP(Sc)) in brain tissues by immunohistochemistry or immunoblot, making antemortem diagnosis of CJD difficult. The World Health Organization (WHO) recommends detection of 14-3-3 protein in cerebrospinal fluid (CSF) in cases of dementia, with clinical correlation, as a useful diagnostic marker for CJD, obviating the need for brain biopsy. This facility is currently available in only a few specialized centers in the West and no commercial kit is available for clinical diagnostic use in India. Hence the objective of this study was to develop an in-house sensitive assay for quantitation of 14-3-3 protein and to evaluate its diagnostic potential to detect 14-3-3 proteins in CSF as a biomarker in suspected cases of CJD. MATERIALS AND METHODS A minigene expressing the "core" 14-3-3 protein was synthesized by overlapping polymerase chain reaction (PCR) and the recombinant protein was produced by employing a bacterial expression system. Polyclonal antibodies raised in rabbit against the purified recombinant protein were used for developing a dot blot assay with avidin-biotin technology for signal amplification and quantitation of 14-3-3 protein in CSF. RESULTS The results in the present study suggest the diagnostic potential of the dot blot method with about 10-fold difference (P< 0.001) in the CSF levels of 14-3-3 protein between the CJD cases (N= 50) and disease controls (N= 70). The receiver operating characteristic (ROC) analysis of the results suggested an optimal cutoff value of 2 ng/mL. CONCLUSIONS We have developed an indigenous, economical, and sensitive dot blot method for the quantitation of 14-3-3 protein in CSF.
Collapse
Affiliation(s)
- Sarada Subramanian
- Department of Neurochemistry, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | | | - Susarla Krishna Shankar
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| |
Collapse
|
41
|
CSF biomarkers in neurodegenerative and vascular dementias. Prog Neurobiol 2016; 138-140:36-53. [DOI: 10.1016/j.pneurobio.2016.03.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/12/2016] [Accepted: 03/14/2016] [Indexed: 12/14/2022]
|
42
|
Ivanov AS, Ershov PV, Molnar AA, Mezentsev YV, Kaluzhskiy LA, Yablokov EO, Florinskaya AV, Gnedenko OV, Medvedev AE, Kozin SA, Mitkevich VA, Makarov AA, Gilep AA, Luschik AY, Gaidukevich IV, Usanov SA. Direct molecular fishing in molecular partners investigation in protein–protein and protein–peptide interactions. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2016. [DOI: 10.1134/s1068162016010052] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
43
|
Yamamoto Y, Koma H, Yagami T. Localization of 14-3-3δ/ξ on the neuronal cell surface. Exp Cell Res 2015; 338:149-61. [PMID: 26363361 DOI: 10.1016/j.yexcr.2015.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 09/02/2015] [Accepted: 09/02/2015] [Indexed: 10/23/2022]
Abstract
14-3-3 proteins are intracellularly expressed as ubiquitous adaptor proteins. Here, we found localization of 14-3-3δ/ξ on the neuronal cell surface. 14-3-3δ/ξ was identified as a membrane target for 15-deoxy-Δ(12,14)-prostaglandin J2 (15d-PGJ2). 15d-PGJ2 is a pathological mediator of neurodegenerative diseases including Alzheimer's disease (AD). A causative peptide for AD, amyloid β, is one of binding partner of 14-3-3δ/ξ. Non-permeabilized neurons were used to avoid the intracellular effects of anti-14-3-3δ/ξ antibody in the present study. The plasmalemmal 14-3-3δ/ξ, but not the cytosolic one, was stimulated by its specific antibody, resulting in neuronal cell death. The neurotoxicity of anti-14-3-3δ/ξ antibody was suppressed by an antioxidant, catalase. Catalase prevented neurons from anti-14-3-3δ/ξ antibody-generating neurotoxic H2O2. The neuroprotective effect of catalase was also detected with the post-treatment of neurons after the application of anti-14-3-3δ/ξ antibody. Activation of mitogen-activated protein kinase signaling cascade is a down-stream consequence of H2O2 exposure. A c-Jun N-terminal kinase inhibitor suppressed anti-14-3-3δ/ξ antibody-induced neuronal cell death. To my knowledge, this is the first report that the antibody-stimulated plasmalemmal 14-3-3δ/ξ induced neuronal cell death. Furthermore, H2O2 and JNK contributed to the neurotoxicity of anti-14-3-3δ/ξ antibody as well as those of amyloid β and 15d-PGJ2.
Collapse
Affiliation(s)
- Yasuhiro Yamamoto
- Division of Physiology, Department of Pharmaceutical Health Care, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 2-1, kami-ohno 7-Chome, Himeji, Hyogo 670-8524, Japan
| | - Hiromi Koma
- Division of Physiology, Department of Pharmaceutical Health Care, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 2-1, kami-ohno 7-Chome, Himeji, Hyogo 670-8524, Japan
| | - Tatsurou Yagami
- Division of Physiology, Department of Pharmaceutical Health Care, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, 2-1, kami-ohno 7-Chome, Himeji, Hyogo 670-8524, Japan.
| |
Collapse
|
44
|
Joo Y, Schumacher B, Landrieu I, Bartel M, Smet-Nocca C, Jang A, Choi HS, Jeon NL, Chang KA, Kim HS, Ottmann C, Suh YH. Involvement of 14-3-3 in tubulin instability and impaired axon development is mediated by Tau. FASEB J 2015; 29:4133-44. [PMID: 26103986 DOI: 10.1096/fj.14-265009] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 06/15/2015] [Indexed: 01/06/2023]
Abstract
14-3-3 proteins act as adapters that exert their function by interacting with their various protein partners. 14-3-3 proteins have been implicated in a variety of human diseases including neurodegenerative diseases. 14-3-3 proteins have recently been reported to be abundant in the neurofibrillary tangles (NFTs) observed inside the neurons of brains affected by Alzheimer's disease (AD). These NFTs are mainly constituted of phosphorylated Tau protein, a microtubule-associated protein known to bind 14-3-3. Despite this indication of 14-3-3 protein involvement in the AD pathogenesis, the role of 14-3-3 in the Tauopathy remains to be clarified. In the present study, we shed light on the role of 14-3-3 proteins in the molecular pathways leading to Tauopathies. Overexpression of the 14-3-3σ isoform resulted in a disruption of the tubulin cytoskeleton and prevented neuritic outgrowth in neurons. NMR studies validated the phosphorylated residues pSer214 and pSer324 in Tau as the 2 primary sites for 14-3-3 binding, with the crystal structure of 14-3-3σ in complex with Tau-pSer214 and Tau-pSer324 revealing the molecular details of the interaction. These data suggest a rationale for a possible pharmacologic intervention of the Tau/14-3-3 interaction.
Collapse
Affiliation(s)
- Yuyoung Joo
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Benjamin Schumacher
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Isabelle Landrieu
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Maria Bartel
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Caroline Smet-Nocca
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Ahram Jang
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hee Soon Choi
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Noo Li Jeon
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Keun-A Chang
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Hye-Sun Kim
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Christian Ottmann
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| | - Yoo-Hun Suh
- *Department of Pharmacology, College of Medicine and Neuroscience Research Institute, Medical Research Council, and School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea; Department of Pharmacology, College of Medicine, Gachon University, Incheon, Republic of Korea; Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany; Unité Mixte de Recherche 8576, Centre National de la Recherche Scientifique-University of Lille, Villeneuve d'Ascq, France; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands; and Korea Brain Research Institute, Daegu, Republic of Korea
| |
Collapse
|
45
|
Homeostatic regulation of T cell trafficking by a B cell-derived peptide is impaired in autoimmune and chronic inflammatory disease. Nat Med 2015; 21:467-475. [PMID: 25894827 PMCID: PMC4425550 DOI: 10.1038/nm.3842] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 03/18/2015] [Indexed: 12/16/2022]
Abstract
During an inflammatory response, lymphocyte recruitment into tissue must be tightly controlled because dysregulated trafficking contributes to the pathogenesis of chronic disease. Here we show that during inflammation and in response to adiponectin, B cells tonically inhibit T cell trafficking by secreting a peptide (PEPITEM) proteolytically derived from 14.3.3.ζδ protein. PEPITEM binds cadherin-15 on endothelial cells, promoting synthesis and release of sphingosine-1 phosphate, which inhibits trafficking of T cells without affecting recruitment of other leukocytes. Expression of adiponectin receptors on B cells and adiponectin induced PEPITEM secretion wanes with age, implying immune senescence of the pathway. Additionally, these changes are evident in individuals with type-1-diabetes or rheumatoid arthritis, and circulating PEPITEM in patient serum is reduced compared to healthy age matched donors. In both diseases, tonic inhibition of T cell trafficking across inflamed endothelium is lost. Importantly, control of patient T cell trafficking is re-established by exogenous PEPITEM. Moreover, in animal models of peritonitis, hepatic I/R injury, Salmonella infection, Uveitis and Sjögren’s Syndrome, PEPITEM could reduce T cell recruitment into inflamed tissues.
Collapse
|
46
|
Regulation of nicotinic acetylcholine receptors in Alzheimer׳s disease: a possible role of chaperones. Eur J Pharmacol 2015; 755:34-41. [PMID: 25771456 DOI: 10.1016/j.ejphar.2015.02.047] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/15/2015] [Accepted: 02/22/2015] [Indexed: 12/25/2022]
Abstract
Nicotinic acetylcholine receptors (nAChRs) seem to play an integral role in the progress and/or prevention of Alzheimer׳s diseases (AD). Functional abnormalities and problems in biogenesis and trafficking of nAChRs are two major culprits in AD; on the other hand, chaperones modulate post-translational changes in nAChRs. Moreover, they indirectly regulate nAChRs by controlling AD-related proteins such as tau and amyloid beta (Aβ). In this review, we go through recent studies which are showing that chaperones modulate the expression of nAChRs in a subtype-specific manner and explain how AD progress is affected by nAChRs chaperoning.
Collapse
|
47
|
Iwamoto M, Miura Y, Tsumoto H, Tanaka Y, Morisawa H, Endo T, Toda T. Antioxidant effects of carnitine supplementation on 14-3-3 protein isoforms in the aged rat hippocampus detected using fully automated two-dimensional chip gel electrophoresis. Free Radic Res 2014; 48:1409-16. [PMID: 25179439 DOI: 10.3109/10715762.2014.960411] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We here described the antioxidant effects of carnitine supplementation on 14-3-3 protein isoforms in the aged rat hippocampus detected using the fully automated two-dimensional chip gel electrophoresis system (Auto2D). This system was easy and convenient to use, and the resolution obtained was more sensitive and higher than that of conventional two-dimensional polyacrylamide gel electrophoresis (2-D PAGE). We separated and identified five isoforms of the 14-3-3 protein (beta/alpha, gamma, epsilon, zeta/delta, and eta) using the Auto2D system. We then examined the antioxidant effects of carnitine supplementation on the protein profiles of the cytosolic fraction in the aged rat hippocampus, demonstrating that carnitine supplementation suppressed the oxidation of methionine residues in these isoforms. Since methionine residues are easily oxidized to methionine sulfoxide, the convenient and high-resolution 2-D PAGE system can be available to analyze methionine oxidation avoiding artifactual oxidation. We showed here that the Auto2D system was a very useful tool for studying antioxidant effects through proteomic analysis of protein oxidation.
Collapse
Affiliation(s)
- M Iwamoto
- Research Team for Mechanism of Aging, Tokyo Metropolitan Institute of Gerontology , Tokyo , Japan
| | | | | | | | | | | | | |
Collapse
|
48
|
Hoyo NLD, López-Begines S, Rosa JL, Chen J, Méndez A. Functional EF-hands in neuronal calcium sensor GCAP2 determine its phosphorylation state and subcellular distribution in vivo, and are essential for photoreceptor cell integrity. PLoS Genet 2014; 10:e1004480. [PMID: 25058152 PMCID: PMC4109901 DOI: 10.1371/journal.pgen.1004480] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 05/17/2014] [Indexed: 11/18/2022] Open
Abstract
The neuronal calcium sensor proteins GCAPs (guanylate cyclase activating proteins) switch between Ca2+-free and Ca2+-bound conformational states and confer calcium sensitivity to guanylate cyclase at retinal photoreceptor cells. They play a fundamental role in light adaptation by coupling the rate of cGMP synthesis to the intracellular concentration of calcium. Mutations in GCAPs lead to blindness. The importance of functional EF-hands in GCAP1 for photoreceptor cell integrity has been well established. Mutations in GCAP1 that diminish its Ca2+ binding affinity lead to cell damage by causing unabated cGMP synthesis and accumulation of toxic levels of free cGMP and Ca2+. We here investigate the relevance of GCAP2 functional EF-hands for photoreceptor cell integrity. By characterizing transgenic mice expressing a mutant form of GCAP2 with all EF-hands inactivated (EF−GCAP2), we show that GCAP2 locked in its Ca2+-free conformation leads to a rapid retinal degeneration that is not due to unabated cGMP synthesis. We unveil that when locked in its Ca2+-free conformation in vivo, GCAP2 is phosphorylated at Ser201 and results in phospho-dependent binding to the chaperone 14-3-3 and retention at the inner segment and proximal cell compartments. Accumulation of phosphorylated EF−GCAP2 at the inner segment results in severe toxicity. We show that in wildtype mice under physiological conditions, 50% of GCAP2 is phosphorylated correlating with the 50% of the protein being retained at the inner segment. Raising mice under constant light exposure, however, drastically increases the retention of GCAP2 in its Ca2+-free form at the inner segment. This study identifies a new mechanism governing GCAP2 subcellular distribution in vivo, closely related to disease. It also identifies a pathway by which a sustained reduction in intracellular free Ca2+ could result in photoreceptor damage, relevant for light damage and for those genetic disorders resulting in “equivalent-light” scenarios. Visual perception is initiated at retinal photoreceptor cells, where light activates an enzymatic cascade that reduces free cGMP. As cGMP drops, cGMP-channels close and reduce the inward current –including Ca2+ influx– so that photoreceptors hyperpolarize and emit a signal. As the light extinguishes, cGMP levels are restored to reestablish sensitivity. cGMP synthesis relies on guanylate cyclase/guanylate cyclase activating protein (RetGC/GCAP) complexes. GCAPs link the rate of cGMP synthesis to intracellular Ca2+ levels, by switching between a Ca2+-free state that activates cGMP synthesis during light exposure, and a Ca2+-bound state that arrests cGMP synthesis in the dark. It is established that GCAP1 mutations linked to adCORD disrupt this tight Ca2+ control of the cGMP levels. We here show that a GCAP2 functional transition from the Ca2+-free to the Ca2+-loaded form is essential for photoreceptor cell integrity, by a non-related mechanism. We show that GCAP2 locked in its Ca2+-free form is retained by phosphorylation and 14-3-3 binding to the proximal rod compartments, causing severe cell damage. This study identifies a pathway by which a sustained reduction in intracellular free Ca2+ could result in photoreceptor damage, relevant for light damage and for those genetic disorders resulting in “equivalent-light” scenarios.
Collapse
Affiliation(s)
| | | | - Jose Luis Rosa
- Department of Physiological Sciences II, University of Barcelona-Bellvitge Health Science Campus, Barcelona, Spain
| | - Jeannie Chen
- Department of Cell and Neurobiology, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, United States of America
| | - Ana Méndez
- Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
- Department of Pathology and Experimental Therapeutics, University of Barcelona-Bellvitge Health Science Campus, Barcelona, Spain
- * E-mail:
| |
Collapse
|
49
|
Höhn A, Jung T, Grune T. Pathophysiological importance of aggregated damaged proteins. Free Radic Biol Med 2014; 71:70-89. [PMID: 24632383 DOI: 10.1016/j.freeradbiomed.2014.02.028] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Revised: 02/28/2014] [Accepted: 02/28/2014] [Indexed: 02/06/2023]
Abstract
Reactive oxygen species (ROS) are formed continuously in the organism even under physiological conditions. If the level of ROS in cells exceeds the cellular defense capacity, components such as RNA/DNA, lipids, and proteins are damaged and modified, thus affecting the functionality of organelles as well. Proteins are especially prominent targets of various modifications such as oxidation, glycation, or conjugation with products of lipid peroxidation, leading to the alteration of their biological function, nonspecific interactions, and the production of high-molecular-weight protein aggregates. To ensure the maintenance of cellular functions, two proteolytic systems are responsible for the removal of oxidized and modified proteins, especially the proteasome and organelles, mainly the autophagy-lysosomal systems. Furthermore, increased protein oxidation and oxidation-dependent impairment of proteolytic systems lead to an accumulation of oxidized proteins and finally to the formation of nondegradable protein aggregates. Accordingly, the cellular homeostasis cannot be maintained and the cellular metabolism is negatively affected. Here we address the current knowledge of protein aggregation during oxidative stress, aging, and disease.
Collapse
Affiliation(s)
- Annika Höhn
- Department of Nutritional Toxicology, Institute of Nutrition, Friedrich-Schiller-University Jena, 07743 Jena, Germany
| | - Tobias Jung
- Department of Nutritional Toxicology, Institute of Nutrition, Friedrich-Schiller-University Jena, 07743 Jena, Germany
| | - Tilman Grune
- Department of Nutritional Toxicology, Institute of Nutrition, Friedrich-Schiller-University Jena, 07743 Jena, Germany.
| |
Collapse
|
50
|
Jia B, Wu Y, Zhou Y. 14-3-3 and aggresome formation: implications in neurodegenerative diseases. Prion 2014; 8:28123. [PMID: 24549097 DOI: 10.4161/pri.28123] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Protein misfolding and aggregation underlie the pathogenesis of many neurodegenerative diseases. In addition to chaperone-mediated refolding and proteasomal degradation, the aggresome-macroautophagy pathway has emerged as another defense mechanism for sequestration and clearance of toxic protein aggregates in cells. Previously, the 14-3-3 proteins were shown to be indispensable for the formation of aggresomes induced by mutant huntingtin proteins. In a recent study, we have determined that 14-3-3 functions as a molecular adaptor to recruit chaperone-associated misfolded proteins to dynein motors for transport to aggresomes. This molecular complex involves a dimeric binding of 14-3-3 to both the dynein-intermediate chain (DIC) and an Hsp70 co-chaperone Bcl-2-associated athanogene 3 (BAG3). As 14-3-3 has been implicated in various neurodegenerative diseases, our findings may provide mechanistic insights into its role in managing misfolded protein stress during the process of neurodegeneration.
Collapse
Affiliation(s)
- Baohui Jia
- Department of Biomedical Sciences; Florida State University College of Medicine; Tallahassee, FL USA; Guang An Men Hospital; Beijing PR China
| | - Yuying Wu
- Department of Biomedical Sciences; Florida State University College of Medicine; Tallahassee, FL USA
| | - Yi Zhou
- Department of Biomedical Sciences; Florida State University College of Medicine; Tallahassee, FL USA
| |
Collapse
|