1
|
Zhou D, Liu B, Liu L, Liu G, Zhu F, Huang Z, Zhang S, He Z, Fan L. Essential Regulation of Spermatogonial Stem Cell Fate Decisions and Male Fertility by APBB1 via Interaction with KAT5 and GDF15 in Humans and Mice. RESEARCH (WASHINGTON, D.C.) 2025; 8:0647. [PMID: 40151319 PMCID: PMC11948500 DOI: 10.34133/research.0647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 12/09/2024] [Accepted: 03/08/2025] [Indexed: 03/29/2025]
Abstract
Spermatogonial stem cells (SSCs) are essential for initiating and maintaining normal spermatogenesis, and notably, they have important applications in both reproduction and regenerative medicine. Nevertheless, the molecular mechanisms controlling the fate determinations of human SSCs remain elusive. In this study, we identified a selective expression of APBB1 in dormant human SSCs. We demonstrated for the first time that APBB1 interacted with KAT5, which led to the suppression of GDF15 expression and consequent inhibition of human SSC proliferation. Intriguingly, Apbb1-/- mice assumed the disrupted spermatogenesis and markedly reduced fertility. SSC transplantation assays revealed that Apbb1 silencing enhanced SSC colonization and impeded their differentiation, which resulted in the impaired spermatogenesis. Notably, 4 deleterious APBB1 mutation sites were identified in 2,047 patients with non-obstructive azoospermia (NOA), and patients with the c.1940C>G mutation had a similar testicular phenotype with Apbb1-/- mice. Additionally, we observed lower expression levels of APBB1 in NOA patients with spermatogenic arrest than in obstructive azoospermia patients with normal spermatogenesis. Collectively, our findings highlight an essential role of APBB1/KAT5/GDF15 in governing human SSC fate decisions and maintaining normal spermatogenesis and underscore them as therapeutic targets for treating male infertility.
Collapse
Affiliation(s)
- Dai Zhou
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science,
Central South University, Changsha, Hunan 410000, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province; Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Institute of Interdisciplinary Studies, Hunan Normal University, Hunan 410013, China
- Hainan Academy of Medical Sciences,
Hainan Medical University, Hainan 570311, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| | - Bang Liu
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Lvjun Liu
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Guangmin Liu
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science,
Central South University, Changsha, Hunan 410000, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| | - Fang Zhu
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science,
Central South University, Changsha, Hunan 410000, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| | - Zenghui Huang
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science,
Central South University, Changsha, Hunan 410000, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| | - Shusheng Zhang
- Hunan Provincial Key Laboratory of Regional Hereditary Birth Defect Prevention and Control, Changsha Hospital for Maternal and Child Health Care Affiliated to Hunan Normal University, Changsha, Hunan 410000, China
| | - Zuping He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province; Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Institute of Interdisciplinary Studies, Hunan Normal University, Hunan 410013, China
- Hainan Academy of Medical Sciences,
Hainan Medical University, Hainan 570311, China
| | - Liqing Fan
- Institute of Reproduction and Stem Cell Engineering, School of Basic Medicine Science,
Central South University, Changsha, Hunan 410000, China
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan 410000, China
| |
Collapse
|
2
|
Chan WWR, Chow J, Chau DDL, Zhai Y, Lau KF. Beclin 1-Mediated Autophagy Is Potentiated by an Interaction with the Neuronal Adaptor FE65. BIOLOGY 2025; 14:97. [PMID: 39857327 PMCID: PMC11763304 DOI: 10.3390/biology14010097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Autophagy is a vital cellular pathway in eukaryotic cells, including neurons, where it plays significant roles in neurodevelopment and maintenance. A crucial step in autophagy is the formation of the class III phosphatidylinositol 3-kinase complex 1 (PI3KC3-C1), which is essential for initiating autophagosome biogenesis. Beclin 1 is the key component of PI3KC3-C1, and its interactors have been reported to affect autophagy. The brain-enriched adaptor protein FE65 has been shown to interact with Alzheimer's disease amyloid precursor protein (APP) to alter the processing of APP. Additionally, FE65 has been implicated in various cellular pathways, including autophagy. We demonstrate here that FE65 positively regulates autophagy. FE65, through its C-terminus, has been shown to interact with Beclin 1. Notably, the overexpression of FE65 enhances Beclin 1-mediated autophagy, whereas this process is attenuated in FE65 knockout cells. Moreover, the stimulatory effect of FE65 on Beclin 1-mediated autophagy is diminished by an FE65 C-terminus deletion mutant that disrupts the FE65-Beclin 1 interaction. Lastly, we have found that the FE65-Beclin 1 interaction modulates the kinase activity of the PI3KC3-C1 complex. Together, we have identified FE65 as a novel Beclin 1 interactor, and this interaction potentiates autophagy.
Collapse
Affiliation(s)
| | | | | | | | - Kwok-Fai Lau
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China; (W.W.R.C.); (J.C.); (D.D.-L.C.); (Y.Z.)
| |
Collapse
|
3
|
Ng LLH, Chow J, Lau KF. The AICD interactome: implications in neurodevelopment and neurodegeneration. Biochem Soc Trans 2024; 52:2539-2556. [PMID: 39670668 PMCID: PMC11668293 DOI: 10.1042/bst20241510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/14/2024]
Abstract
The pathophysiological mechanism involving the proteolytic processing of amyloid precursor protein (APP) and the generation of amyloid plaques is of significant interest in research on Alzheimer's disease (AD). The increasing significance of the downstream AD-related pathophysiological mechanisms has sparked research interest in other products of the APP processing cascades, including the APP intracellular domain (AICD). The potential importance of AICD in various cellular processes in the central nervous system has been established through the identification of its interactors. The interaction between AICD and its physiological binding partners is implicated in cellular events including regulation of transcriptional activity, cytoskeletal dynamics, neuronal growth, APP processing and cellular apoptosis. On the contrary, AICD is also implicated in neurodegeneration, which is a potential outcome of the functional fluctuation of AICD-mediated neuronal processes within the neuronal network. In this review, we summarize the neuronal functions and pathological manifestations of the dynamic AICD interaction network.
Collapse
Affiliation(s)
- Laura Lok-Haang Ng
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jessica Chow
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kwok-Fai Lau
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
4
|
Chiu J, Krupa JM, Seah C, Pasternak SH. Small GTPases control macropinocytosis of amyloid precursor protein and cleavage to amyloid-β. Heliyon 2024; 10:e31077. [PMID: 38799759 PMCID: PMC11126852 DOI: 10.1016/j.heliyon.2024.e31077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/03/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024] Open
Abstract
The overproduction of the toxic peptide amyloid-beta (Aβ) generated from the cleavage of amyloid precursor protein (APP) is proposed to be a critical event in the development of Alzheimer's disease. Evidence suggests that the cleavage of APP occurs after its internalization from the cell surface. Previously, we identified a novel pathway for APP internalization, which trafficks cell surface APP directly to lysosomes by macropinocytosis, leading to its processing into Aβ. We also demonstrated that ADP-ribosylation factor 6 (Arf6) is required for the macropinocytosis of APP. Here, we characterized the roles of Arf6's downstream effectors Rac1, Cdc42 and RhoA. Both pharmacological inhibition and siRNA knockdown of these proteins reduced the amount of APP colocalized with LAMP1-labeled lysosomes without affecting APP transport to early endosomes. Decreases in the production of both Aβ40 and Aβ42 were also observed by ELISA in response to inhibitor treatment. These findings together demonstrate that Rac1, Cdc42 and RhoA are components of the mechanism regulating the macropinocytosis of APP and targeting these components can reduce the production of Aβ.
Collapse
Affiliation(s)
- Justin Chiu
- Department of Physiology and Pharmacology, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Jordan M. Krupa
- Neuroscience Program, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Claudia Seah
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Stephen H. Pasternak
- Department of Physiology and Pharmacology, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Neuroscience Program, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Robarts Research Institute, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Department of Clinical Neurological Sciences, The Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
5
|
Revol RS, Koistinen NA, Menon PK, Chicote-Gonzàlez A, Iverfeldt K, Ström AL. Alpha-secretase dependent nuclear localization of the amyloid-β precursor protein-binding protein Fe65 promotes DNA repair. Mol Cell Neurosci 2023; 127:103903. [PMID: 37918552 DOI: 10.1016/j.mcn.2023.103903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/29/2023] [Accepted: 10/29/2023] [Indexed: 11/04/2023] Open
Abstract
Fe65 is a brain enriched adaptor protein involved in various cellular processes, including actin cytoskeleton regulation, DNA repair and transcription. A well-studied interacting partner of Fe65 is the transmembrane amyloid-β precursor protein (APP), which can undergo regulated intramembrane proteolysis (RIP). Following β- and γ-secretase-mediated RIP, the released APP intracellular domain (AICD) together with Fe65 can translocate to the nucleus and regulate transcription. In this study, we investigated if Fe65 nuclear localization can also be regulated by different α-secretases, also known to participate in RIP of APP and other transmembrane proteins. We found that in both Phorbol 12-myristate 13-acetate and all-trans retinoic acid differentiated neuroblastoma cells a strong negative impact on Fe65 nuclear localization, equal to the effect observed upon γ-secretase inhibition, could be detected following inhibition of all three (ADAM9, ADAM10 and ADAM17) α-secretases. Moreover, using the comet assay and analysis of Fe65 dependent DNA repair associated posttranslational modifications of histones, we could show that inhibition of α-secretase-mediated Fe65 nuclear translocation resulted in impaired capacity of the cells to repair DNA damage. Taken together this suggests that α-secretase processing of APP and/or other Fe65 interacting transmembrane proteins play an important role in regulating Fe65 nuclear translocation and DNA repair.
Collapse
Affiliation(s)
- Rebecca S Revol
- Stockholm University, Department of Biochemistry and Biophysics, 106 91 Stockholm, Sweden
| | - Niina A Koistinen
- Stockholm University, Department of Biochemistry and Biophysics, 106 91 Stockholm, Sweden
| | - Preeti K Menon
- Stockholm University, Department of Biochemistry and Biophysics, 106 91 Stockholm, Sweden
| | | | - Kerstin Iverfeldt
- Stockholm University, Department of Biochemistry and Biophysics, 106 91 Stockholm, Sweden
| | - Anna-Lena Ström
- Stockholm University, Department of Biochemistry and Biophysics, 106 91 Stockholm, Sweden.
| |
Collapse
|
6
|
Chau DDL, Ng LLH, Zhai Y, Lau KF. Amyloid precursor protein and its interacting proteins in neurodevelopment. Biochem Soc Trans 2023; 51:1647-1659. [PMID: 37387352 PMCID: PMC10629809 DOI: 10.1042/bst20221527] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/22/2023] [Accepted: 06/26/2023] [Indexed: 07/01/2023]
Abstract
Amyloid precursor protein (APP) is a key molecule in the pathogenesis of Alzheimer's disease (AD) as the pathogenic amyloid-β peptide is derived from it. Two closely related APP family proteins (APPs) have also been identified in mammals. Current knowledge, including genetic analyses of gain- and loss-of-function mutants, highlights the importance of APPs in various physiological functions. Notably, APPs consist of multiple extracellular and intracellular protein-binding regions/domains. Protein-protein interactions are crucial for many cellular processes. In past decades, many APPs interactors have been identified which assist the revelation of the putative roles of APPs. Importantly, some of these interactors have been shown to influence several APPs-mediated neuronal processes which are found defective in AD and other neurodegenerative disorders. Studying APPs-interactor complexes would not only advance our understanding of the physiological roles of APPs but also provide further insights into the association of these processes to neurodegeneration, which may lead to the development of novel therapies. In this mini-review, we summarize the roles of APPs-interactor complexes in neurodevelopmental processes including neurogenesis, neurite outgrowth, axonal guidance and synaptogenesis.
Collapse
Affiliation(s)
- Dennis Dik-Long Chau
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Laura Lok-Haang Ng
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yuqi Zhai
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kwok-Fai Lau
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
7
|
Justo AFO, Toscano ECDB, Farias-Itao DS, Suemoto CK. The action of phosphodiesterase-5 inhibitors on β-amyloid pathology and cognition in experimental Alzheimer's disease: A systematic review. Life Sci 2023; 320:121570. [PMID: 36921685 DOI: 10.1016/j.lfs.2023.121570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/09/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023]
Abstract
Alzheimer's disease (AD) is the most frequent cause of dementia worldwide. The etiology of AD is partially explained by the deposition of β-amyloid in the brain. Despite extensive research on the pathogenesis of AD, the current treatments are ineffective. Here, we systematically reviewed studies that investigated whether phosphodiesterase 5 inhibitors (PDE5i) are efficient in reducing the β-amyloid load in hippocampi and improving cognitive decline in rodent models with β-amyloid accumulation. We identified ten original studies, which used rodent models with β-amyloid accumulation, were treated with PDE5i, and β-amyloid was measured in the hippocampi. PDE5i was efficient in reducing the β-amyloid levels, except for one study that exclusively used female rodents and the treatment did not affect β-amyloid levels. Interestingly, PDE5i prevented cognitive decline in all studies. This study supports the potential therapeutic use of PDE5i for the reduction of the β-amyloid load in hippocampi and cognitive decline. However, we highlight the importance of conducting additional experimental studies to evaluate the PDE5i-related molecular mechanisms involved in β-amyloid removal in male and female animals.
Collapse
Affiliation(s)
- Alberto Fernando Oliveira Justo
- Physiopathology in Aging Laboratory (LIM-22), Department of Internal Medicine, University of São Paulo Medical School, São Paulo, Brazil.
| | - Eliana Cristina de Brito Toscano
- Physiopathology in Aging Laboratory (LIM-22), Department of Internal Medicine, University of São Paulo Medical School, São Paulo, Brazil; Department of Pathology, Federal University of Juiz de Fora Medical School, Juiz de Fora, Brazil; Post-graduation Program in Health, Federal University of Juiz de Fora Medical School, Juiz de Fora, Brazil.
| | | | - Claudia Kimie Suemoto
- Physiopathology in Aging Laboratory (LIM-22), Department of Internal Medicine, University of São Paulo Medical School, São Paulo, Brazil; Division of Geriatrics, Department of Internal Medicine, University of São Paulo Medical School, São Paulo, Brazil.
| |
Collapse
|
8
|
Brandimarti R, Irollo E, Meucci O. The US9-Derived Protein gPTB9TM Modulates APP Processing Without Targeting Secretase Activities. Mol Neurobiol 2023; 60:1811-1825. [PMID: 36576708 PMCID: PMC9984340 DOI: 10.1007/s12035-022-03153-2] [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: 06/01/2022] [Accepted: 11/29/2022] [Indexed: 12/29/2022]
Abstract
Alteration of neuronal protein processing is often associated with neurological disorders and is highly dependent on cellular protein trafficking. A prime example is the amyloidogenic processing of amyloid precursor protein (APP) in intracellular vesicles, which plays a key role in age-related cognitive impairment. Most approaches to correct this altered processing aim to limit enzymatic activities that lead to toxic products, such as protein cleavage by β-secretase and the resulting amyloid β production. A viable alternative is to direct APP to cellular compartments where non-amyloidogenic mechanisms are favored. To this end, we exploited the molecular properties of the herpes simplex virus 1 (HSV-1) transport protein US9 to guide APP interaction with preferred endogenous targets. Specifically, we generated a US9 chimeric construct that facilitates APP processing through the non-amyloidogenic pathway and tested it in primary cortical neurons. In addition to reducing amyloid β production, our approach controls other APP-dependent biochemical steps that lead to neuronal deficits, including phosphorylation of APP and tau proteins. Notably, it also promotes the release of neuroprotective soluble αAPP. In contrast to other neuroprotective strategies, these US9-driven effects rely on the activity of endogenous neuronal proteins, which lends itself well to the study of fundamental mechanisms of APP processing/trafficking. Overall, this work introduces a new method to limit APP misprocessing and its cellular consequences without directly targeting secretase activity, offering a novel tool to reduce cognitive decline in pathologies such as Alzheimer's disease and HIV-associated neurocognitive disorders.
Collapse
Affiliation(s)
- Renato Brandimarti
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N.15th Street, Philadelphia, PA, 19102, USA.,Center for Neuroimmunology and CNS Therapeutics, Drexel University College of Medicine, 245 N.15th Street, Philadelphia, PA, 19102, USA.,Department of Pharmacy and Biotechnology, University of Bologna, Via San Giacomo,14, 40126, Bologna, Italy
| | - Elena Irollo
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N.15th Street, Philadelphia, PA, 19102, USA.,Center for Neuroimmunology and CNS Therapeutics, Drexel University College of Medicine, 245 N.15th Street, Philadelphia, PA, 19102, USA
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N.15th Street, Philadelphia, PA, 19102, USA. .,Center for Neuroimmunology and CNS Therapeutics, Drexel University College of Medicine, 245 N.15th Street, Philadelphia, PA, 19102, USA. .,Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N.15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
9
|
FSTL1-knockdown improves neural oscillation via decreasing neuronal-inflammation regulating apoptosis in Aβ 1-42 induced AD model mice. Exp Neurol 2023; 359:114231. [PMID: 36162512 DOI: 10.1016/j.expneurol.2022.114231] [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: 05/05/2022] [Revised: 09/04/2022] [Accepted: 09/19/2022] [Indexed: 12/30/2022]
Abstract
Follistatin like protein 1 (FSTL1) is a famous growth regulatory protein. FSTL1 has been noticed in many diseases, including heart and lung ischemia, cerebral ischemia, glioma, schizophrenia, and Autism. The role of FSTL1 has been declared in the genetics and development of the central nervous system. Therefore, we designed this study to investigate the function and the role of FSTL1 in Alzheimer's disease. Firstly, we noticed upregulated expression level of FSTL1 among four to six-month-old 5XFAD AD mice. Accordingly, we hypothesized that FSTL1-Knockdown improved AD model mice's cognitive function and recover from Alzheimer's disease. Thus, AD model mice were made by single intracerebroventricular injections of Aβ1-42 peptides in FSTL1+/- and CON mice. Next, our results concluded that FSTL1-knockdown effectively improved cognitive functions. FSTL1-knockdown enhanced the pattern of neural oscillations, and synaptic plasticity in Aβ1-42 treated FSTL1-Knockdown mice compared to Aβ1-42 induced AD model mice. Next, FSTL1-Knockdown inhibited the activation of microglia and binding of TLR-4 with microglia. Further, inactivated microglia stopped the formation of MyD88. Thus, our data revealed that FSTL1-Knockdown is slowing down the caspase/BAX/Bcl-2/TLR-4 regulating apoptosis pathway, and the expression of inflammatory cytokines in the hippocampus of Aβ1-42 inserted FSTL1-Knockdown mice. Overall, all these data illuminate the clinical significance role of down-regulated FSTL1. FSTL1-Knockdown reduced the amyloid-beta by affecting microglia, neural-inflammation and apoptosis in AD-like model mice. Finally, down regulation of FSTL1 improved synaptic plasticity, neural oscillations, and cognitive behaviours in the Aβ1-42 induced AD model mice.
Collapse
|
10
|
Kamal M, Tokmakjian L, Knox J, Mastrangelo P, Ji J, Cai H, Wojciechowski JW, Hughes MP, Takács K, Chu X, Pei J, Grolmusz V, Kotulska M, Forman-Kay JD, Roy PJ. A spatiotemporal reconstruction of the C. elegans pharyngeal cuticle reveals a structure rich in phase-separating proteins. eLife 2022; 11:e79396. [PMID: 36259463 PMCID: PMC9629831 DOI: 10.7554/elife.79396] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 10/11/2022] [Indexed: 11/19/2022] Open
Abstract
How the cuticles of the roughly 4.5 million species of ecdysozoan animals are constructed is not well understood. Here, we systematically mine gene expression datasets to uncover the spatiotemporal blueprint for how the chitin-based pharyngeal cuticle of the nematode Caenorhabditis elegans is built. We demonstrate that the blueprint correctly predicts expression patterns and functional relevance to cuticle development. We find that as larvae prepare to molt, catabolic enzymes are upregulated and the genes that encode chitin synthase, chitin cross-linkers, and homologs of amyloid regulators subsequently peak in expression. Forty-eight percent of the gene products secreted during the molt are predicted to be intrinsically disordered proteins (IDPs), many of which belong to four distinct families whose transcripts are expressed in overlapping waves. These include the IDPAs, IDPBs, and IDPCs, which are introduced for the first time here. All four families have sequence properties that drive phase separation and we demonstrate phase separation for one exemplar in vitro. This systematic analysis represents the first blueprint for cuticle construction and highlights the massive contribution that phase-separating materials make to the structure.
Collapse
Affiliation(s)
- Muntasir Kamal
- Department of Molecular Genetics, University of TorontoTorontoCanada
- The Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
| | - Levon Tokmakjian
- The Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
- Department of Pharmacology and Toxicology, University of TorontoTorontoCanada
| | - Jessica Knox
- Department of Molecular Genetics, University of TorontoTorontoCanada
- The Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
| | - Peter Mastrangelo
- Department of Molecular Genetics, University of TorontoTorontoCanada
- The Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
| | - Jingxiu Ji
- Department of Molecular Genetics, University of TorontoTorontoCanada
- The Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
| | - Hao Cai
- Molecular Medicine Program, The Hospital for Sick ChildrenTorontoCanada
| | - Jakub W Wojciechowski
- Wroclaw University of Science and Technology, Faculty of Fundamental Problems of Technology, Department of Biomedical EngineeringWroclawPoland
| | - Michael P Hughes
- Department of Cell and Molecular Biology, St. Jude Children’s Research HospitalMemphisUnited States
| | - Kristóf Takács
- PIT Bioinformatics Group, Institute of Mathematics, Eötvös UniversityBudapestHungary
| | - Xiaoquan Chu
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking UniversityBeijingChina
| | - Jianfeng Pei
- Department of Computer Science and Technology, Tsinghua UniversityBeijingChina
| | - Vince Grolmusz
- PIT Bioinformatics Group, Institute of Mathematics, Eötvös UniversityBudapestHungary
| | - Malgorzata Kotulska
- Wroclaw University of Science and Technology, Faculty of Fundamental Problems of Technology, Department of Biomedical EngineeringWroclawPoland
| | - Julie Deborah Forman-Kay
- Molecular Medicine Program, The Hospital for Sick ChildrenTorontoCanada
- Department of Biochemistry, University of TorontoTorontoCanada
| | - Peter J Roy
- Department of Molecular Genetics, University of TorontoTorontoCanada
- The Donnelly Centre for Cellular and Biomolecular Research, University of TorontoTorontoCanada
- Department of Pharmacology and Toxicology, University of TorontoTorontoCanada
| |
Collapse
|
11
|
Wurz AI, Schulz AM, O’Bryant CT, Sharp JF, Hughes RM. Cytoskeletal dysregulation and neurodegenerative disease: Formation, monitoring, and inhibition of cofilin-actin rods. Front Cell Neurosci 2022; 16:982074. [PMID: 36212686 PMCID: PMC9535683 DOI: 10.3389/fncel.2022.982074] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/31/2022] [Indexed: 12/04/2022] Open
Abstract
The presence of atypical cytoskeletal dynamics, structures, and associated morphologies is a common theme uniting numerous diseases and developmental disorders. In particular, cytoskeletal dysregulation is a common cellular feature of Alzheimer’s disease, Parkinson’s disease, and Huntington’s disease. While the numerous activators and inhibitors of dysregulation present complexities for characterizing these elements as byproducts or initiators of the disease state, it is increasingly clear that a better understanding of these anomalies is critical for advancing the state of knowledge and plan of therapeutic attack. In this review, we focus on the hallmarks of cytoskeletal dysregulation that are associated with cofilin-linked actin regulation, with a particular emphasis on the formation, monitoring, and inhibition of cofilin-actin rods. We also review actin-associated proteins other than cofilin with links to cytoskeleton-associated neurodegenerative processes, recognizing that cofilin-actin rods comprise one strand of a vast web of interactions that occur as a result of cytoskeletal dysregulation. Our aim is to present a current perspective on cytoskeletal dysregulation, connecting recent developments in our understanding with emerging strategies for biosensing and biomimicry that will help shape future directions of the field.
Collapse
Affiliation(s)
- Anna I. Wurz
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Anna M. Schulz
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Collin T. O’Bryant
- Department of Chemistry, East Carolina University, Greenville, NC, United States
| | - Josephine F. Sharp
- Department of Chemistry, Notre Dame College, South Euclid, OH, United States
| | - Robert M. Hughes
- Department of Chemistry, East Carolina University, Greenville, NC, United States
- *Correspondence: Robert M. Hughes,
| |
Collapse
|
12
|
Examination of Longitudinal Alterations in Alzheimer’s Disease-Related Neurogenesis in an APP/PS1 Transgenic Mouse Model, and the Effects of P33, a Putative Neuroprotective Agent Thereon. Int J Mol Sci 2022; 23:ijms231810364. [PMID: 36142277 PMCID: PMC9499399 DOI: 10.3390/ijms231810364] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/05/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Neurogenesis plays a crucial role in cognitive processes. During aging and in Alzheimer’s disease (AD), altered neurogenesis and neuroinflammation are evident both in C57BL/6J, APPSwe/PS1dE9 (Tg) mice and humans. AD pathology may slow down upon drug treatment, for example, in a previous study of our group P33, a putative neuroprotective agent was found to exert advantageous effects on the elevated levels of APP, Aβ, and neuroinflammation. In the present study, we aimed to examine longitudinal alterations in neurogenesis, neuroinflammation and AD pathology in a transgenic (Tg) mouse model, and assessed the putative beneficial effects of long-term P33 treatment on AD-specific neurological alterations. Hippocampal cell proliferation and differentiation were significantly reduced between 8 and 12 months of age. Regarding neuroinflammation, significantly elevated astrogliosis and microglial activation were observed in 6- to 7-month-old Tg animals. The amounts of the molecules involved in the amyloidogenic pathway were altered from 4 months of age in Tg animals. P33-treatment led to significantly increased neurogenesis in 9-month-old animals. Our data support the hypothesis that altered neurogenesis may be a consequence of AD pathology. Based on our findings in the transgenic animal model, early pharmacological treatment before the manifestation of AD symptoms might ameliorate neurological decline.
Collapse
|
13
|
Zhang Y, Yang X, Liu Y, Ge L, Wang J, Sun X, Wu B, Wang J. Vav2 is a novel APP-interacting protein that regulates APP protein level. Sci Rep 2022; 12:12752. [PMID: 35882892 PMCID: PMC9325707 DOI: 10.1038/s41598-022-16883-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/18/2022] [Indexed: 11/24/2022] Open
Abstract
Amyloid precursor protein (APP) is a transmembrane protein that plays critical role in the pathogenesis of Alzheimer's disease (AD). It is also involved in many types of cancers. Increasing evidence has shown that the tyrosine phosphorylation site Y682 in the intracellular tail of APP is crucial for APP function. Here, we report that Vav2, a guanine nucleotide exchange factor (GEF) for Rho family GTPase, is a novel interaction partner of APP. We found that Vav2-SH2 domain was able to bind directly to the Y682-phosphorylated intracellular tail of APP through isothermal titration calorimetry and NMR titrating experiments. The crystal structure of Vav2-SH2 in complex with an APP-derived phosphopeptide was determined to understand the structural basis of this recognition specificity. The interaction of APP and Vav2 in a full-length manner was further confirmed in cells by GST pull-down, co-immunoprecipitation and immunofluorescence staining experiments. In addition, we found overexpression of Vav2 could inhibit APP degradation and markedly increase the protein levels of APP and its cleavage productions in 20E2 cells, and this function of Vav2 required a functional SH2 domain.
Collapse
Affiliation(s)
- Youjia Zhang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.,University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaxin Yang
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, China.,Brain Research Institute, Qilu Hospital of Shandong University, Jinan, China
| | - Yongrui Liu
- University of Science and Technology of China, Hefei, Anhui, China
| | - Liang Ge
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Jiarong Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China
| | - Xiulian Sun
- Brain Research Institute, Qilu Hospital of Shandong University, Jinan, China. .,The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission, Qilu Hospital of Shandong University, Jinan, China. .,NHC Key Laboratory of Otorhinolaryngology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Bo Wu
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China.
| | - Junfeng Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui, China. .,University of Science and Technology of China, Hefei, Anhui, China. .,Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, China.
| |
Collapse
|
14
|
Xu J, Iwabuchi E, Miki Y, Kanai A, Takagi K, Suzuki T, Ishida T, Sasano H. FE65 defines the efficacy of tamoxifen treatment via osteopontin expression in estrogen receptor-positive breast cancer. Pathol Res Pract 2022; 234:153898. [DOI: 10.1016/j.prp.2022.153898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 10/18/2022]
|
15
|
FE65 in breast cancer and its clinicopathological significance. Breast Cancer 2021; 29:144-155. [PMID: 34498219 DOI: 10.1007/s12282-021-01291-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 08/31/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Transcription coregulator adapter protein FE65 is well known to play pivotal roles in pathogenesis of Alzheimer's disease by regulating amyloid precursor protein (APP) expression and processing. APP was recently reported to be also involved in development of human malignancies. Therefore, in this study, we studied FE65 status in different subtypes of human breast cancer and correlated the results with cell proliferation and migration of carcinoma cells and clinicopathological features of breast cancer patients to explore its biological and clinical significance in breast cancer. METHODS We first immunolocalized FE65 and APP in 138 breast cancer patients and correlated the results with their tumor grade. Then, we did further exploration by proximity ligation assay, WST-8, and wound-healing assay. RESULTS FE65 immunoreactivity in carcinoma cells was significantly associated with lymph-node metastasis, ERα, and high pathological N factor. APP immunoreactivity was significantly positively correlated with high pathological N factor. FE65, APP, and p-APP were all significantly correlated with shorter disease-free survival of breast cancer patients. In addition, the status of FE65 was significantly associated with overall survival. Results of in vitro analysis revealed that FE65 promoted the migration and proliferation of T-47D and ZR-75-1 breast carcinoma cells. In situ proximity ligation assay revealed that FE65 could bind to APP in the cytoplasm. FE65 was also associated with APP and ERα in carcinoma cells, suggesting their cooperativity in promoting carcinoma cell proliferation and migration. APP was also significantly associated with adverse clinical outcome of the patients. CONCLUSIONS This is the first study to explore the clinical significance of FE65 in human breast cancer. The significant positive correlation of FE65 with poor clinical outcome, direct binding to APP, and promotion of carcinoma cell proliferation and migration indicated that FE65-APP pathway could serve as the potential candidate of therapeutic intervention in breast cancer patients.
Collapse
|
16
|
Augustin V, Kins S. Fe65: A Scaffolding Protein of Actin Regulators. Cells 2021; 10:cells10071599. [PMID: 34202290 PMCID: PMC8304848 DOI: 10.3390/cells10071599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 01/19/2023] Open
Abstract
The scaffolding protein family Fe65, composed of Fe65, Fe65L1, and Fe65L2, was identified as an interaction partner of the amyloid precursor protein (APP), which plays a key function in Alzheimer’s disease. All three Fe65 family members possess three highly conserved interaction domains, forming complexes with diverse binding partners that can be assigned to different cellular functions, such as transactivation of genes in the nucleus, modulation of calcium homeostasis and lipid metabolism, and regulation of the actin cytoskeleton. In this article, we rule out putative new intracellular signaling mechanisms of the APP-interacting protein Fe65 in the regulation of actin cytoskeleton dynamics in the context of various neuronal functions, such as cell migration, neurite outgrowth, and synaptic plasticity.
Collapse
|
17
|
Yao Y, Kang SS, Xia Y, Wang ZH, Liu X, Muller T, Sun YE, Ye K. A delta-secretase-truncated APP fragment activates CEBPB, mediating Alzheimer's disease pathologies. Brain 2021; 144:1833-1852. [PMID: 33880508 DOI: 10.1093/brain/awab062] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 12/10/2020] [Accepted: 12/10/2020] [Indexed: 11/13/2022] Open
Abstract
Amyloid-β precursor protein (APP) is sequentially cleaved by secretases and generates amyloid-β, the major components in senile plaques in Alzheimer's disease. APP is upregulated in human Alzheimer's disease brains. However, the molecular mechanism of how APP contributes to Alzheimer's disease pathogenesis remains incompletely understood. Here we show that truncated APP C586-695 fragment generated by δ-secretase directly binds to CCAAT/enhancer-binding protein beta (CEBPB), an inflammatory transcription factor, and enhances its transcriptional activity, escalating Alzheimer's disease-related gene expression and pathogenesis. The APP C586-695 fragment, but not full-length APP, strongly associates with CEBPB and elicits its nuclear translocation and augments the transcriptional activities on APP itself, MAPT (microtubule-associated protein tau), δ-secretase and inflammatory cytokine mRNA expression, finally triggering Alzheimer's disease pathology and cognitive disorder in a viral overexpression mouse model. Blockade of δ-secretase cleavage of APP by mutating the cleavage sites reduces its stimulatory effect on CEBPB, alleviating amyloid pathology and cognitive dysfunctions. Clearance of APP C586-695 from 5xFAD mice by antibody administration mitigates Alzheimer's disease pathologies and restores cognitive functions. Thus, in addition to the sequestration of amyloid-β, APP implicates in Alzheimer's disease pathology by activating CEBPB upon δ-secretase cleavage.
Collapse
Affiliation(s)
- Yinan Yao
- Tongji Hospital, Tongji University School of Medicine, Shanghai 20065, P.R. China.,Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Seong Su Kang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yiyuan Xia
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Zhi-Hao Wang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Thorsten Muller
- Department of Molecular Biochemistry, Cell Signalling, Ruhr-University Bochum, 44801 Bochum, Germany.,Institute of Psychiatric Phenomics and Genomics (IPPG), University Hospital, LMU Munich, Munich 80336, Germany
| | - Yi E Sun
- Tongji Hospital, Tongji University School of Medicine, Shanghai 20065, P.R. China.,Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
18
|
Li Z, Rasmussen LJ. TIP60 in aging and neurodegeneration. Ageing Res Rev 2020; 64:101195. [PMID: 33091598 DOI: 10.1016/j.arr.2020.101195] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 09/29/2020] [Accepted: 10/14/2020] [Indexed: 02/06/2023]
Abstract
Epigenetic modification of chromatin, including histone methylation and acetylation, plays critical roles in eukaryotic cells and has a significant impact on chromatin structure/accessibility, gene regulation and, susceptibility to aging, neurodegenerative disease, cancer, and other age-related diseases. This article reviews the current advances on TIP60/KAT5, a major histone acetyltransferase with diverse functions in eukaryotes, with emphasis on its regulation of autophagy, proteasome-dependent protein turnover, RNA transcription, DNA repair, circadian rhythms, learning and memory, and other neurological functions implicated in aging and neurodegeneration. Moreover, the promising therapeutic potential of TIP60 is discussed to target Alzheimer's disease and other neurological diseases.
Collapse
|
19
|
Gasterstädt I, Jack A, Stahlhut T, Rennau LM, Gonda S, Wahle P. Genetically Encoded Calcium Indicators Can Impair Dendrite Growth of Cortical Neurons. Front Cell Neurosci 2020; 14:570596. [PMID: 33192315 PMCID: PMC7606991 DOI: 10.3389/fncel.2020.570596] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022] Open
Abstract
A battery of genetically encoded calcium indicators (GECIs) with different binding kinetics and calcium affinities was developed over the recent years to permit long-term calcium imaging. GECIs are calcium buffers and therefore, expression of GECIs may interfere with calcium homeostasis and signaling pathways important for neuronal differentiation and survival. Our objective was to investigate if the biolistically induced expression of five commonly used GECIs at two postnatal time points (days 14 and 22–25) could affect the morphological maturation of cortical neurons in organotypic slice cultures of rat visual cortex. Expression of GCaMP3 in both time windows, and of GCaMP5G and TN-XXL in the later time window impaired apical and /or basal dendrite growth of pyramidal neurons. With time, the proportion of GECI transfectants with nuclear filling increased, but an only prolonged expression of TN-XXL caused higher levels of neurodegeneration. In multipolar interneurons, only GCaMP3 evoked a transient growth delay during the early time window. GCaMP6m and GCaMP6m-XC were quite “neuron-friendly.” Since growth-impaired neurons might not have the physiological responses typical of age-matched wildtype neurons the results obtained after prolonged developmental expression of certain GECIs might need to be interpreted with caution.
Collapse
Affiliation(s)
- Ina Gasterstädt
- Developmental Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Alexander Jack
- Developmental Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Tobias Stahlhut
- Developmental Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Lisa-Marie Rennau
- Developmental Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Steffen Gonda
- Developmental Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| | - Petra Wahle
- Developmental Neurobiology, Faculty of Biology and Biotechnology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
20
|
Probst S, Krüger M, Kägi L, Thöni S, Schuppli D, Nitsch RM, Konietzko U. Fe65 is the sole member of its family that mediates transcription regulated by the amyloid precursor protein. J Cell Sci 2020; 133:jcs242917. [PMID: 32843577 DOI: 10.1242/jcs.242917] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 07/11/2020] [Indexed: 08/31/2023] Open
Abstract
The amyloid precursor protein (APP), a central molecule in Alzheimer's disease (AD), has physiological roles in cell adhesion and signaling, migration, neurite outgrowth and synaptogenesis. Intracellular adapter proteins mediate the function of transmembrane proteins. Fe65 (also known as APBB1) is a major APP-binding protein. Regulated intramembrane proteolysis (RIP) by γ-secretase releases the APP intracellular domain (AICD), together with the interacting proteins, from the membrane. We studied the impact of the Fe65 family (Fe65, and its homologs Fe65L1 and Fe65L2, also known as APBB2 and APBB3, respectively) on the nuclear signaling function of the AICD. All Fe65 family members increased amyloidogenic processing of APP, generating higher levels of β-cleaved APP stubs and AICD. However, Fe65 was the only family member supporting AICD translocation to nuclear spots and its transcriptional activity. Using a recently established transcription assay, we dissected the transcriptional activity of Fe65 and provide strong evidence that Fe65 represents a transcription factor. We show that Fe65 relies on the lysine acetyltransferase Tip60 (also known as KAT5) for nuclear translocation. Furthermore, inhibition of APP cleavage reduces nuclear Tip60 levels, but this does not occur in Fe65-knockout cells. The rate of APP cleavage therefore regulates the nuclear translocation of AICD-Fe65-Tip60 (AFT) complexes, to promote transcription by Fe65.
Collapse
Affiliation(s)
- Sabine Probst
- Institute for Regenerative Medicine - IREM, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952 Schlieren - Zurich, Switzerland
| | - Maik Krüger
- Institute for Regenerative Medicine - IREM, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952 Schlieren - Zurich, Switzerland
| | - Larissa Kägi
- Institute for Regenerative Medicine - IREM, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952 Schlieren - Zurich, Switzerland
| | - Sarina Thöni
- Institute for Regenerative Medicine - IREM, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952 Schlieren - Zurich, Switzerland
| | - Daniel Schuppli
- Institute for Regenerative Medicine - IREM, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952 Schlieren - Zurich, Switzerland
| | - Roger M Nitsch
- Institute for Regenerative Medicine - IREM, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952 Schlieren - Zurich, Switzerland
| | - Uwe Konietzko
- Institute for Regenerative Medicine - IREM, University of Zurich, Campus Schlieren, Wagistrasse 12, 8952 Schlieren - Zurich, Switzerland
| |
Collapse
|
21
|
Iannuzzi F, Sirabella R, Canu N, Maier TJ, Annunziato L, Matrone C. Fyn Tyrosine Kinase Elicits Amyloid Precursor Protein Tyr682 Phosphorylation in Neurons from Alzheimer's Disease Patients. Cells 2020; 9:E1807. [PMID: 32751526 PMCID: PMC7463977 DOI: 10.3390/cells9081807] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disorder with a few early detection strategies. We previously proposed the amyloid precursor protein (APP) tyrosine 682 (Tyr682) residue as a valuable target for the development of new innovative pharmacologic or diagnostic interventions in AD. Indeed, when APP is phosphorylated at Tyr682, it is forced into acidic neuronal compartments where it is processed to generate neurotoxic amyloid β peptides. Of interest, Fyn tyrosine kinase (TK) interaction with APP Tyr682 residue increases in AD neurons. Here we proved that when Fyn TK was overexpressed it elicited APP Tyr682 phosphorylation in neurons from healthy donors and promoted the amyloidogenic APP processing with Aβ peptides accumulation and neuronal death. Phosphorylation of APP at Tyr (pAPP-Tyr) increased in neurons of AD patients and AD neurons that exhibited high pAPP-Tyr also had higher Fyn TK activity. Fyn TK inhibition abolished the pAPP-Tyr and reduced Aβ42 secretion in AD neurons. In addition, the multidomain adaptor protein Fe65 controlled the Fyn-mediated pAPP-Tyr, warranting the possibility of targeting the Fe65-APP-Fyn pathway to develop innovative strategies in AD. Altogether, these results strongly emphasize the relevance of focusing on pAPP Tyr682 either for diagnostic purposes, as an early biomarker of the disease, or for pharmacological targeting, using Fyn TKI.
Collapse
Affiliation(s)
- Filomena Iannuzzi
- Department of Biomedicine, Aarhus University, Aarhus C, 8000 Aarhus, Denmark;
| | - Rossana Sirabella
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
| | - Nadia Canu
- Department of System Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
- Institute of Biochemistry and Cell Biology, CNR, 00015 Monterotondo, Rome, Italy
| | - Thorsten J. Maier
- Paul-Ehrlich-Institut, (Federal Institute for Vaccines and Biomedicines), 63225 Langen, Germany;
| | - Lucio Annunziato
- SDN Research Institute Diagnostics and Nuclear (IRCCS SDN), Gianturco, 80131 Naples, Italy
| | - Carmela Matrone
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
| |
Collapse
|
22
|
Unravelling the Role of Glycogen Synthase Kinase-3 in Alzheimer's Disease-Related Epileptic Seizures. Int J Mol Sci 2020; 21:ijms21103676. [PMID: 32456185 PMCID: PMC7279454 DOI: 10.3390/ijms21103676] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/20/2020] [Accepted: 05/20/2020] [Indexed: 12/21/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. An increasing body of evidence describes an elevated incidence of epilepsy in patients with AD, and many transgenic animal models of AD also exhibit seizures and susceptibility to epilepsy. However, the biological mechanisms that underlie the occurrence of seizure or increased susceptibility to seizures in AD is unknown. Glycogen synthase kinase-3 (GSK-3) is a serine/threonine kinase that regulates various cellular signaling pathways, and plays a crucial role in the pathogenesis of AD. It has been suggested that GSK-3 might be a key factor that drives epileptogenesis in AD by interacting with the pathological hallmarks of AD, amyloid precursor protein (APP) and tau. Furthermore, seizures may also contribute to the progression of AD through GSK-3. In this way, GSK-3 might be involved in initiating a vicious cycle between AD and seizures. This review aims to summarise the possible role of GSK-3 in the link between AD and seizures. Understanding the role of GSK-3 in AD-associated seizures and epilepsy may help researchers develop new therapeutic approach that can manage seizure and epilepsy in AD patients as well as decelerate the progression of AD.
Collapse
|
23
|
Abstract
The WW domain is a modular protein structure that recognizes the proline-rich Pro-Pro-x-Tyr (PPxY) motif contained in specific target proteins. The compact modular nature of the WW domain makes it ideal for mediating interactions between proteins in complex networks and signaling pathways of the cell (e.g. the Hippo pathway). As a result, WW domains play key roles in a plethora of both normal and disease processes. Intriguingly, RNA and DNA viruses have evolved strategies to hijack cellular WW domain-containing proteins and thereby exploit the modular functions of these host proteins for various steps of the virus life cycle, including entry, replication, and egress. In this review, we summarize key findings in this rapidly expanding field, in which new virus-host interactions continue to be identified. Further unraveling of the molecular aspects of these crucial virus-host interactions will continue to enhance our fundamental understanding of the biology and pathogenesis of these viruses. We anticipate that additional insights into these interactions will help support strategies to develop a new class of small-molecule inhibitors of viral PPxY-host WW-domain interactions that could be used as antiviral therapeutics.
Collapse
Affiliation(s)
- Ariel Shepley-McTaggart
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Hao Fan
- Bioinformatics Institute, Agency for Science, Technology, and Research (A*STAR), 30 Biopolis Street, Matrix #07-01, Singapore 138671.,Department of Biological Sciences (DBS), National University of Singapore, Singapore 119077.,Center for Computational Biology, DUKE-NUS Medical School, Singapore 169857
| | - Marius Sudol
- Department of Physiology, National University of Singapore, Singapore 119077.,Laboratory of Cancer Signaling and Domainopathies, Yong Loo Li School of Medicine, Block MD9, 2 Medical Drive #04-01, Singapore 117597.,Mechanobiology Institute, T-Lab, 5A Engineering Drive 1, Singapore 117411.,Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Ronald N Harty
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
24
|
McLean FH, Campbell FM, Sergi D, Grant C, Morris AC, Hay EA, MacKenzie A, Mayer CD, Langston RF, Williams LM. Early and reversible changes to the hippocampal proteome in mice on a high-fat diet. Nutr Metab (Lond) 2019; 16:57. [PMID: 31462902 PMCID: PMC6708244 DOI: 10.1186/s12986-019-0387-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 08/16/2019] [Indexed: 12/11/2022] Open
Abstract
Background The rise in global obesity makes it crucial to understand how diet drives obesity-related health conditions, such as premature cognitive decline and Alzheimer's disease (AD). In AD hippocampal-dependent episodic memory is one of the first types of memory to be impaired. Previous studies have shown that in mice fed a high-fat diet (HFD) episodic memory is rapidly but reversibly impaired. Methods In this study we use hippocampal proteomics to investigate the effects of HFD in the hippocampus. Mice were fed either a low-fat diet (LFD) or HFD containing either 10% or 60% (Kcal) from fat for 3 days, 1 week or 2 weeks. One group of mice were fed the HFD for 1 week and then returned to the LFD for a further week. Primary hippocampal cultures were challenged with palmitic acid (PA), the most common long-chain saturated FA in the Western diet, and with the anti-inflammatory, n-3 polyunsaturated FA, docosahexaenoic acid (DHA), or a combination of the two to ascertain effects of these fatty acids on dendritic structure. Results HFD-induced changes occur in hippocampal proteins involved in metabolism, inflammation, cell stress, cell signalling, and the cytoskeleton after 3 days, 1 week and 2 weeks of HFD. Replacement of the HFD after 1 week by a low-fat diet (LFD) for a further week resulted in partial recovery of the hippocampal proteome. Microtubule-associated protein 2 (MAP2), one of the earliest proteins changed, was used to investigate the impact of fatty acids (FAs) on hippocampal neuronal morphology. PA challenge resulted in shorter and less arborised dendrites while DHA had no effect when applied alone but counteracted the effects of PA when FAs were used in combination. Dendritic morphology recovered when PA was removed from the cell culture media. Conclusion This study provides evidence for the rapid and reversible effects of diet on the hippocampal proteome and the impact of PA and DHA on dendritic structure.
Collapse
Affiliation(s)
- Fiona H McLean
- 1Division of Neuroscience, University of Dundee, Ninewells Hospital & Medical School, Dundee, DD1 9SY UK.,2Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD UK
| | - Fiona M Campbell
- 2Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD UK
| | - Domenico Sergi
- 2Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD UK
| | - Christine Grant
- 2Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD UK
| | - Amanda C Morris
- 2Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD UK
| | - Elizabeth A Hay
- 3Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD UK
| | - Alasdair MacKenzie
- 3Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD UK
| | - Claus D Mayer
- 4Biomathematics and Statistics Scotland, University of Aberdeen, Aberdeen, AB25 2ZD UK
| | - Rosamund F Langston
- 1Division of Neuroscience, University of Dundee, Ninewells Hospital & Medical School, Dundee, DD1 9SY UK
| | - Lynda M Williams
- 2Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD UK
| |
Collapse
|
25
|
Matrone C, Iannuzzi F, Annunziato L. The Y 682ENPTY 687 motif of APP: Progress and insights toward a targeted therapy for Alzheimer's disease patients. Ageing Res Rev 2019; 52:120-128. [PMID: 31039414 DOI: 10.1016/j.arr.2019.04.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/04/2019] [Accepted: 04/10/2019] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder for which no curative treatments, disease modifying strategies or effective symptomatic therapies exist. Current pharmacologic treatments for AD can only decelerate the progression of the disease for a short time, often at the cost of severe side effects. Therefore, there is an urgent need for biomarkers able to diagnose AD at its earliest stages, to conclusively track disease progression, and to accelerate the clinical development of innovative therapies. Scientific research and economic efforts for the development of pharmacotherapies have recently homed in on the hypothesis that neurotoxic β-amyloid (Aβ) peptides in their oligomeric or fibrillary forms are primarily responsible for the cognitive impairment and neuronal death seen in AD. As such, modern pharmacologic approaches are largely based on reducing production by inhibiting β and γ secretase cleavage of the amyloid precursor protein (APP) or on dissolving existing cerebral Aβ plaques or to favor Aβ clearance from the brain. The following short review aims to persuade the reader of the idea that APP plays a much larger role in AD pathogenesis. APP plays a greater role in AD pathogenesis than its role as the precursor for Aβ peptides: both the abnormal cleavage of APP leading to Aβ peptide accumulation and the disruption of APP physiological functions contribute to AD pathogenesis. We summarize our recent results on the role played by the C-terminal APP motif -the Y682ENPTY68 motif- in APP function and dysfunction, and we provide insights into targeting the Tyr682 residue of APP as putative novel strategy in AD.
Collapse
|
26
|
Effects of the Pentapeptide P33 on Memory and Synaptic Plasticity in APP/PS1 Transgenic Mice: A Novel Mechanism Presenting the Protein Fe65 as a Target. Int J Mol Sci 2019; 20:ijms20123050. [PMID: 31234498 PMCID: PMC6627374 DOI: 10.3390/ijms20123050] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/17/2019] [Accepted: 06/21/2019] [Indexed: 01/09/2023] Open
Abstract
Regulated intramembrane proteolysis (RIP) of the amyloid precursor protein (APP) leads to the formation of fragments, among which the intracellular domain of APP (AICD) was also identified to be a causative of early pathological events. AICD-counteracting proteins, such as Fe65, may serve as alternative therapeutic targets of Alzheimer’s disease (AD). The detection of elevated levels of Fe65 in the brains of both human patients and APP transgenic mice may further strengthen the hypothesis that influencing the interaction between Fe65 and APP may have a beneficial effect on the course of AD. Based on a PXP motif, proven to bind to the WW domain of Fe65, a new pentapeptide was designed and tested. The impedimental effect of P33 on the production of beta amyloid (Aβ) (soluble fraction and aggregated plaques) and on the typical features of the AD pathology (decreased dendritic spine density, synaptic markers, elevated inflammatory reactions) was also demonstrated. Significant enhancements of both learning ability and memory function were observed in a Morris water maze paradigm. The results led us to formulate the theory that P33 acts by altering the conformation of Fe65 via binding to its WW domain, consequently hindering any interactions between Fe65 and key members involved in APP processing.
Collapse
|
27
|
Li W, Tam KMV, Chan WWR, Koon AC, Ngo JCK, Chan HYE, Lau KF. Neuronal adaptor FE65 stimulates Rac1-mediated neurite outgrowth by recruiting and activating ELMO1. J Biol Chem 2018; 293:7674-7688. [PMID: 29615491 DOI: 10.1074/jbc.ra117.000505] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 03/12/2018] [Indexed: 12/25/2022] Open
Abstract
Neurite outgrowth is a crucial process in developing neurons for neural network formation. Understanding the regulatory mechanisms of neurite outgrowth is essential for developing strategies to stimulate neurite regeneration after nerve injury and in neurodegenerative disorders. FE65 is a brain-enriched adaptor that stimulates Rac1-mediated neurite elongation. However, the precise mechanism by which FE65 promotes the process remains elusive. Here, we show that ELMO1, a subunit of ELMO1-DOCK180 bipartite Rac1 guanine nucleotide exchange factor (GEF), interacts with the FE65 N-terminal region. Overexpression of FE65 and/or ELMO1 enhances, whereas knockdown of FE65 or ELMO1 inhibits, neurite outgrowth and Rac1 activation. The effect of FE65 alone or together with ELMO1 is attenuated by an FE65 double mutation that disrupts FE65-ELMO1 interaction. Notably, FE65 is found to activate ELMO1 by diminishing ELMO1 intramolecular autoinhibitory interaction and to promote the targeting of ELMO1 to the plasma membrane, where Rac1 is activated. We also show that FE65, ELMO1, and DOCK180 form a tripartite complex. Knockdown of DOCK180 reduces the stimulatory effect of FE65-ELMO1 on Rac1 activation and neurite outgrowth. Thus, we identify a novel mechanism by which FE65 stimulates Rac1-mediated neurite outgrowth by recruiting and activating ELMO1.
Collapse
Affiliation(s)
- Wen Li
- From the School of Life Sciences, Faculty of Science, Chinese University of Hong Kong, Shatin New Territories, Hong Kong
| | - Ka Ming Vincent Tam
- From the School of Life Sciences, Faculty of Science, Chinese University of Hong Kong, Shatin New Territories, Hong Kong
| | - Wai Wa Ray Chan
- From the School of Life Sciences, Faculty of Science, Chinese University of Hong Kong, Shatin New Territories, Hong Kong
| | - Alex Chun Koon
- From the School of Life Sciences, Faculty of Science, Chinese University of Hong Kong, Shatin New Territories, Hong Kong
| | - Jacky Chi Ki Ngo
- From the School of Life Sciences, Faculty of Science, Chinese University of Hong Kong, Shatin New Territories, Hong Kong
| | - Ho Yin Edwin Chan
- From the School of Life Sciences, Faculty of Science, Chinese University of Hong Kong, Shatin New Territories, Hong Kong
| | - Kwok-Fai Lau
- From the School of Life Sciences, Faculty of Science, Chinese University of Hong Kong, Shatin New Territories, Hong Kong
| |
Collapse
|
28
|
Wang Q, Li WX, Dai SX, Guo YC, Han FF, Zheng JJ, Li GH, Huang JF. Meta-Analysis of Parkinson's Disease and Alzheimer's Disease Revealed Commonly Impaired Pathways and Dysregulation of NRF2-Dependent Genes. J Alzheimers Dis 2018; 56:1525-1539. [PMID: 28222515 DOI: 10.3233/jad-161032] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Many lines of evidence suggest that Parkinson's disease (PD) and Alzheimer's disease (AD) have common characteristics, such as mitochondrial dysfunction and oxidative stress. As the underlying molecular mechanisms are unclear, we perform a meta-analysis with 9 microarray datasets of PD studies and 7 of AD studies to explore it. Functional enrichment analysis revealed that PD and AD both showed dysfunction in the synaptic vesicle cycle, GABAergic synapses, phagosomes, oxidative phosphorylation, and TCA cycle pathways, and AD had more enriched genes. Comparing the differentially expressed genes between AD and PD, we identified 54 common genes shared by more than six tissues. Among them, 31 downregulated genes contained the antioxidant response element (ARE) consensus sequence bound by NRF2. NRF2 is a transcription factor, which protects cells against oxidative stress through coordinated upregulation of ARE-driven genes. To our surprise, although NRF2 was upregulated, its target genes were all downregulated. Further exploration found that MAFF was upregulated in all tissues and significantly negatively correlated with the 31 NRF2-dependent genes in diseased conditions. Previous studies have demonstrated over-expressed small MAFs can form homodimers and act as transcriptional repressors. Therefore, MAFF might play an important role in dysfunction of NRF2 regulatory network in PD and AD.
Collapse
Affiliation(s)
- Qian Wang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Wen-Xing Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Institute of Health Sciences, Anhui University, Hefei, Anhui, China
| | - Shao-Xing Dai
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yi-Cheng Guo
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Fei-Fei Han
- Immuno-Metabolic Computational Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jun-Juan Zheng
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Gong-Hua Li
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Jing-Fei Huang
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China.,KIZ-SU Joint Laboratory of Animal Models and Drug Development, College of Pharmaceutical Sciences, Soochow University, Kunming, Yunnan, China.,Collaborative Innovation Center for Natural Products and Biological Drugs of Yunnan, Kunming, Yunnan, China.,Chinese University of Hong Kong Joint Research Center for Bio-resources and Human Disease Mechanisms, Kunming, Yunnan, China
| |
Collapse
|
29
|
Phosphorylation of FE65 at threonine 579 by GSK3β stimulates amyloid precursor protein processing. Sci Rep 2017; 7:12456. [PMID: 28963516 PMCID: PMC5622059 DOI: 10.1038/s41598-017-12334-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/06/2017] [Indexed: 02/01/2023] Open
Abstract
Excessive generation of amyloid-β peptide (Aβ) by aberrant proteolysis of amyloid precursor protein (APP) is a key event in Alzheimer's disease (AD) pathogenesis. FE65 is a brain-enriched phospho-adaptor protein that interacts with APP and has been shown to modulate APP processing. However, the mechanism(s) that FE65 alters APP processing is still not fully understood. In the present study, we demonstrate that FE65 is phosphorylated at threonine 579 (T579) by glycogen synthase kinase 3β (GSK3β). Moreover, FE65 T579 phosphorylation potentiates γ- and β-secretases-mediated APP processing and Aβ liberation. Additionally, the phosphorylation suppresses FE65 PTB2 intermolecular dimerization but enhances FE65/APP complex formation. Hence, our findings reveal a novel mechanism that GSK3β stimulates amyloidogenic processing of APP by phosphorylation of FE65 at T579.
Collapse
|
30
|
Small things matter: Implications of APP intracellular domain AICD nuclear signaling in the progression and pathogenesis of Alzheimer’s disease. Prog Neurobiol 2017; 156:189-213. [DOI: 10.1016/j.pneurobio.2017.05.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/25/2017] [Accepted: 05/30/2017] [Indexed: 01/08/2023]
|
31
|
Feilen LP, Haubrich K, Strecker P, Probst S, Eggert S, Stier G, Sinning I, Konietzko U, Kins S, Simon B, Wild K. Fe65-PTB2 Dimerization Mimics Fe65-APP Interaction. Front Mol Neurosci 2017; 10:140. [PMID: 28553201 PMCID: PMC5425604 DOI: 10.3389/fnmol.2017.00140] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/25/2017] [Indexed: 01/21/2023] Open
Abstract
Physiological function and pathology of the Alzheimer’s disease causing amyloid precursor protein (APP) are correlated with its cytosolic adaptor Fe65 encompassing a WW and two phosphotyrosine-binding domains (PTBs). The C-terminal Fe65-PTB2 binds a large portion of the APP intracellular domain (AICD) including the GYENPTY internalization sequence fingerprint. AICD binding to Fe65-PTB2 opens an intra-molecular interaction causing a structural change and altering Fe65 activity. Here we show that in the absence of the AICD, Fe65-PTB2 forms a homodimer in solution and determine its crystal structure at 2.6 Å resolution. Dimerization involves the unwinding of a C-terminal α-helix that mimics binding of the AICD internalization sequence, thus shielding the hydrophobic binding pocket. Specific dimer formation is validated by nuclear magnetic resonance (NMR) techniques and cell-based analyses reveal that Fe65-PTB2 together with the WW domain are necessary and sufficient for dimerization. Together, our data demonstrate that Fe65 dimerizes via its APP interaction site, suggesting that besides intra- also intermolecular interactions between Fe65 molecules contribute to homeostatic regulation of APP mediated signaling.
Collapse
Affiliation(s)
- Lukas P Feilen
- Heidelberg University Biochemistry Center (BZH), University of HeidelbergHeidelberg, Germany
| | - Kevin Haubrich
- Heidelberg University Biochemistry Center (BZH), University of HeidelbergHeidelberg, Germany.,European Molecular Biology Laboratory (EMBL), Structural and Computational BiologyHeidelberg, Germany
| | - Paul Strecker
- Division of Human Biology and Human Genetics, University of KaiserslauternKaiserslautern, Germany
| | - Sabine Probst
- Institute for Regenerative Medicine (IREM), University of ZurichZurich, Switzerland
| | - Simone Eggert
- Division of Human Biology and Human Genetics, University of KaiserslauternKaiserslautern, Germany
| | - Gunter Stier
- Heidelberg University Biochemistry Center (BZH), University of HeidelbergHeidelberg, Germany
| | - Irmgard Sinning
- Heidelberg University Biochemistry Center (BZH), University of HeidelbergHeidelberg, Germany
| | - Uwe Konietzko
- Institute for Regenerative Medicine (IREM), University of ZurichZurich, Switzerland
| | - Stefan Kins
- Division of Human Biology and Human Genetics, University of KaiserslauternKaiserslautern, Germany
| | - Bernd Simon
- European Molecular Biology Laboratory (EMBL), Structural and Computational BiologyHeidelberg, Germany
| | - Klemens Wild
- Heidelberg University Biochemistry Center (BZH), University of HeidelbergHeidelberg, Germany
| |
Collapse
|
32
|
Montagna E, Dorostkar MM, Herms J. The Role of APP in Structural Spine Plasticity. Front Mol Neurosci 2017; 10:136. [PMID: 28539872 PMCID: PMC5423954 DOI: 10.3389/fnmol.2017.00136] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/21/2017] [Indexed: 11/15/2022] Open
Abstract
Amyloid precursor protein (APP) is a transmembrane protein highly expressed in neurons. The full-length protein has cell-adhesion and receptor-like properties, which play roles in synapse formation and stability. Furthermore, APP can be cleaved by several proteases into numerous fragments, many of which affect synaptic function and stability. This review article focuses on the mechanisms of APP in structural spine plasticity, which encompasses the morphological alterations at excitatory synapses. These occur as changes in the number and morphology of dendritic spines, which correspond to the postsynaptic compartment of excitatory synapses. Both overexpression and knockout (KO) of APP lead to impaired synaptic plasticity. Recent data also suggest a role of APP in the regulation of astrocytic D-serine homeostasis, which in turn regulates synaptic plasticity.
Collapse
Affiliation(s)
- Elena Montagna
- Department for Translational Brain Research, German Center for Neurodegenerative Diseases (DZNE), Ludwig-Maximilian-University MunichMunich, Germany
| | - Mario M Dorostkar
- Center for Neuropathology and Prion Research, Ludwig-Maximilian-University MunichMunich, Germany
| | - Jochen Herms
- Department for Translational Brain Research, German Center for Neurodegenerative Diseases (DZNE), Ludwig-Maximilian-University MunichMunich, Germany.,Center for Neuropathology and Prion Research, Ludwig-Maximilian-University MunichMunich, Germany.,Munich Cluster of Systems Neurology (SyNergy), Ludwig-Maximilian-University MunichMunich, Germany
| |
Collapse
|
33
|
Andrew RJ, Kellett KAB, Thinakaran G, Hooper NM. A Greek Tragedy: The Growing Complexity of Alzheimer Amyloid Precursor Protein Proteolysis. J Biol Chem 2016; 291:19235-44. [PMID: 27474742 DOI: 10.1074/jbc.r116.746032] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Proteolysis of the amyloid precursor protein (APP) liberates various fragments including the proposed initiator of Alzheimer disease-associated dysfunctions, amyloid-β. However, recent evidence suggests that the accepted view of APP proteolysis by the canonical α-, β-, and γ-secretases is simplistic, with the discovery of a number of novel APP secretases (including δ- and η-secretases, alternative β-secretases) and additional metabolites, some of which may also cause synaptic dysfunction. Furthermore, various proteins have been identified that interact with APP and modulate its cleavage by the secretases. Here, we give an overview of the increasingly complex picture of APP proteolysis.
Collapse
Affiliation(s)
- Robert J Andrew
- From the Departments of Neurobiology, Neurology, and Pathology, The University of Chicago, Chicago, Illinois 60637 and
| | - Katherine A B Kellett
- the Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| | - Gopal Thinakaran
- From the Departments of Neurobiology, Neurology, and Pathology, The University of Chicago, Chicago, Illinois 60637 and
| | - Nigel M Hooper
- the Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, United Kingdom
| |
Collapse
|
34
|
Fe65 Is Phosphorylated on Ser289 after UV-Induced DNA Damage. PLoS One 2016; 11:e0155056. [PMID: 27176072 PMCID: PMC4866770 DOI: 10.1371/journal.pone.0155056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 04/24/2016] [Indexed: 11/19/2022] Open
Abstract
Fe65 undergoes a phosphatase-sensitive gel mobility shift after DNA damage, consistent with protein phosphorylation. A recent study identified Ser228 as a specific site of phosphorylation, targeted by the ATM and ATR protein kinases, with phosphorylation inhibiting the Fe65-dependent transcriptional activity of the amyloid precursor protein (APP). The direct binding of Fe65 to APP not only regulates target gene expression, but also contributes to secretase-mediated processing of APP, producing cytoactive proteolytic fragments including the APP intracellular domain (AICD) and cytotoxic amyloid β (Aβ) peptides. Given that the accumulation of Aβ peptides in neural plaques is a pathological feature of Alzheimer’s disease (AD), it is essential to understand the mechanisms controlling Aβ production. This will aid in the development of potential therapeutic agents that act to limit the deleterious production of Aβ peptides. The Fe65-APP complex has transcriptional activity and the complex is regulated by multiple post-translational modifications and other protein binding partners. In the present study, we have identified Ser289 as a novel site of UV-induced phosphorylation. Interestingly, this phosphorylation was mediated by ATM, rather than ATR, and occurred independently of APP. Neither phosphorylation nor mutation of Ser289 affected the Fe65-APP interaction, though this was markedly decreased after UV treatment, with a concomitant decrease in the protein levels of APP in cells. Using mutagenesis, we demonstrated that Fe65 Ser289 phosphorylation did not affect the transcriptional activity of the Fe65-APP complex, in contrast to the previously described Ser228 site.
Collapse
|
35
|
Abstract
FE65 is a brain-enriched, developmentally regulated adaptor protein that was first identified as a binding partner of amyloid precursor protein (APP), an important molecule in Alzheimer's disease. FE65 possesses three protein interaction domains, including an N-terminal WW domain and two C-terminal phosphotyrosine-binding (PTB) domains. It is capable of mediating the assembly of multimolecular complexes. Although initial work reveals its roles in APP processing and gene transactivation, increasing evidence suggests that FE65 participates in more diverse biological processes than originally anticipated. This article discusses the role of FE65 in signal transduction during cell stress and protein turnover through the ubiquitin-proteasome system and in various neuronal processes, including neurogenesis, neuronal migration and positioning, neurite outgrowth, synapse formation and synaptic plasticity, learning, and memory.
Collapse
|
36
|
FE65 and FE65L1 share common synaptic functions and genetically interact with the APP family in neuromuscular junction formation. Sci Rep 2016; 6:25652. [PMID: 27734846 PMCID: PMC4899880 DOI: 10.1038/srep25652] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 04/20/2016] [Indexed: 01/12/2023] Open
Abstract
The FE65 adaptor proteins (FE65, FE65L1 and FE65L2) bind proteins that function in diverse cellular pathways and are essential for specific biological processes. Mice lacking both FE65 and FE65L1 exhibit ectopic neuronal positioning in the cortex and muscle weakness. p97FE65-KO mice, expressing a shorter FE65 isoform able to bind amyloid precursor protein family members (APP, APLP1, APLP2), develop defective long-term potentiation (LTP) and aged mice display spatial learning and memory deficits that are absent from young mice. Here, we examined the central and peripheral nervous systems of FE65-KO, FE65L1-KO and FE65/FE65L1-DKO mice. We find spatial learning and memory deficits in FE65-KO and FE65L1-KO mice. Severe motor impairments, anxiety, hippocampal LTP deficits and neuromuscular junction (NMJ) abnormalities, characterized by decreased size and reduced apposition of pre- and postsynaptic sites, are observed in FE65/FE65L1-DKO mice. As their NMJ deficits resemble those of mutant APP/APLP2-DKO mice lacking the FE65/FE65L1 binding site, the NMJs of APLP2/FE65-DKO and APLP2/FE65L1-DKO mice were analyzed. NMJ deficits are aggravated in these mice when compared to single FE65- and FE65L1-KO mice. Together, our data demonstrate a role for FE65 proteins at central and peripheral synapses possibly occurring downstream of cell surface-associated APP/APLPs.
Collapse
|
37
|
Hunter S, Martin S, Brayne C. The APP Proteolytic System and Its Interactions with Dynamic Networks in Alzheimer's Disease. Methods Mol Biol 2016; 1303:71-99. [PMID: 26235060 DOI: 10.1007/978-1-4939-2627-5_3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Diseases of aging are often complex and multifactorial, involving many genetic and life course modifiers. Systems biology is becoming an essential tool to investigate disease initiation and disease progression. Alzheimer's disease (AD) can be used as a case study to investigate the application of systems biology to complex disease. Here we describe approaches to capturing biological data, representing data in terms of networks and interpreting their meaning in relation to the human population. We highlight issues that remain to be addressed both in terms of modeling disease progression and in relating findings to the current understanding of human disease.
Collapse
Affiliation(s)
- Sally Hunter
- Department of Public Health and Primary Care, Institute of Public Health, University of Cambridge School of Clinical Medicine, Forvie Site, Cambridge Biomedical Campus, Box 113, Cambridge, CB2 0SP, UK,
| | | | | |
Collapse
|
38
|
Amyloid precursor-like protein 2 (APLP2) affects the actin cytoskeleton and increases pancreatic cancer growth and metastasis. Oncotarget 2015; 6:2064-75. [PMID: 25576918 PMCID: PMC4385836 DOI: 10.18632/oncotarget.2990] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 12/10/2014] [Indexed: 01/12/2023] Open
Abstract
Amyloid precursor-like protein 2 (APLP2) is aberrantly expressed in pancreatic cancer. Here we showed that APLP2 is increased in pancreatic cancer metastases, particularly in metastatic lesions found in the diaphragm and intestine. Examination of matched human primary tumor-liver metastasis pairs showed that 38.1% of the patients had positive APLP2 expression in both the primary tumor and the corresponding liver metastasis. Stable knock-down of APLP2 expression (with inducible shRNA) in pancreatic cancer cells reduced the ability of these cells to migrate and invade. Loss of APLP2 decreased cortical actin and increased intracellular actin filaments in pancreatic cancer cells. Down-regulation of APLP2 decreased the weight and metastasis of orthotopically transplanted pancreatic tumors in nude mice.
Collapse
|
39
|
Hohman TJ, Chibnik L, Bush WS, Jefferson AL, De Jaeger PL, Thornton-Wells TA, Bennett DA, Schneider JA. GSK3β Interactions with Amyloid Genes: An Autopsy Verification and Extension. Neurotox Res 2015; 28:232-8. [PMID: 26194614 PMCID: PMC4625986 DOI: 10.1007/s12640-015-9541-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 06/11/2015] [Accepted: 07/07/2015] [Indexed: 12/19/2022]
Abstract
Glyocogen synthase kinase 3 (GSK3) plays an important role in the pathophysiology of Alzheimer's disease (AD) through the phosphorylation of tau. Recent work has suggested that GSK3β also plays a role in the amyloid pathway of AD through genetic interactions with APP and APBB2 on in vivo measures of amyloid. This project extends the previously identified genotype interactions to an autopsy measure of amyloid, while also testing the same interactions leveraging gene expression data quantified in the prefrontal cortex. 797 participants (251 cognitively normal, 196 mild cognitive impairment, and 350 Alzheimer's disease) were drawn from the Religious Orders Study and Rush Memory and Aging Project. A mean score of amyloid load was calculated across eight brain regions, gene expression levels from frozen sections of the dorsolateral prefrontal cortex were quantified using RNA amplification, and expression signals were generated using Beadstudio. Three SNPs previously identified in genetic interactions were genotyped using the Illumina 1M genotyping chip. Covariates included age, sex, education, and diagnosis. We were able to evaluate 2 of the 3 previously identified interactions, of which the interaction between GSK3β (rs334543) and APBB2 (rs2585590) was found in this autopsy sample (p = 0.04). We observed a comparable interaction between GSK3β and APBB2 when comparing the highest tertile of gene expression to the lowest tertile, t(1) = -2.03, p = 0.043. These results provide additional evidence of a genetic interaction between GSK3β and APBB2 and further suggest that GSK3β is involved in the pathophysiology of both of the primary neuropathologies of Alzheimer's disease.
Collapse
Affiliation(s)
- Timothy J Hohman
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University School of Medicine, Vanderbilt University Medical Center, 2525 West End Ave, 12th Floor, Suite 1200, Nashville, TN, 37203, USA,
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Martinez-Rodriguez S, Bacarizo J, Luque I, Camara-Artigas A. Crystal structure of the first WW domain of human YAP2 isoform. J Struct Biol 2015; 191:381-7. [DOI: 10.1016/j.jsb.2015.08.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 08/06/2015] [Accepted: 08/06/2015] [Indexed: 11/16/2022]
|
41
|
Lee HJ, Yoon JH, Ahn JS, Jo EH, Kim MY, Lee YC, Kim JW, Ann EJ, Park HS. Fe65 negatively regulates Jagged1 signaling by decreasing Jagged1 protein stability through the E3 ligase Neuralized-like 1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:2918-28. [PMID: 26276215 DOI: 10.1016/j.bbamcr.2015.08.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 08/06/2015] [Accepted: 08/09/2015] [Indexed: 11/30/2022]
Abstract
Fe65 is a highly conserved adaptor protein that interacts with several binding partners. Fe65 binds proteins to mediate various cellular processes. But the interacting partner and the regulatory mechanisms controlled by Fe65 are largely unknown. In this study, we found that Fe65 interacts with the C-terminus of Jagged1. Furthermore, Fe65 negatively regulates AP1-mediated Jagged1 intercellular domain transactivation in a Tip60-independent manner. We found that Fe65 triggers the degradation of Jagged1, but not the Jagged1 intracellular domain (JICD), through both proteasome and lysosome pathways. We also showed that Fe65 promotes recruitment of the E3 ligase Neuralized-like 1 (Neurl1) to membrane-tethered Jagged1 and monoubiquitination of Jagged1. These three proteins form a stable trimeric complex, thereby decreasing Jagged1 targeting by ubiquitin-mediated degradation. Consequently, Jagged1 is a novel binding partner of Fe65, and Fe65 may act as a novel effector of Jagged1 signaling.
Collapse
Affiliation(s)
- Hye-Jin Lee
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Ji-Hye Yoon
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Ji-Seon Ahn
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Eun-Hye Jo
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Mi-Yeon Kim
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Young Chul Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Jin Woo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Eun-Jung Ann
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea.
| | - Hee-Sae Park
- Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea.
| |
Collapse
|
42
|
Phosphorylation of FE65 Ser610 by serum- and glucocorticoid-induced kinase 1 modulates Alzheimer's disease amyloid precursor protein processing. Biochem J 2015; 470:303-17. [PMID: 26188042 PMCID: PMC4613528 DOI: 10.1042/bj20141485] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 07/17/2015] [Indexed: 01/15/2023]
Abstract
Phosphorylation of FE65 Ser610 by serum- and glucocorticoid-induced kinase 1 (SGK1) attenuates amyloid precursor protein (APP) processing via regulation of FE65–APP interaction. Alzheimer's disease (AD) is a fatal neurodegenerative disease affecting 36 million people worldwide. Genetic and biochemical research indicate that the excessive generation of amyloid-β peptide (Aβ) from amyloid precursor protein (APP), is a major part of AD pathogenesis. FE65 is a brain-enriched adaptor protein that binds to APP. However, the role of FE65 in APP processing and the mechanisms that regulate binding of FE65 to APP are not fully understood. In the present study, we show that serum- and glucocorticoid-induced kinase 1 (SGK1) phosphorylates FE65 on Ser610 and that this phosphorylation attenuates FE65 binding to APP. We also show that FE65 promotes amyloidogenic processing of APP and that FE65 Ser610 phosphorylation inhibits this effect. Furthermore, we found that the effect of FE65 Ser610 phosphorylation on APP processing is linked to a role of FE65 in metabolic turnover of APP via the proteasome. Thus FE65 influences APP degradation via the proteasome and phosphorylation of FE65 Ser610 by SGK1 regulates binding of FE65 to APP, APP turnover and processing.
Collapse
|
43
|
Sun Y, Sun J, Lungchukiet P, Quarni W, Yang S, Zhang X, Bai W. Fe65 Suppresses Breast Cancer Cell Migration and Invasion through Tip60 Mediated Cortactin Acetylation. Sci Rep 2015; 5:11529. [PMID: 26166158 PMCID: PMC4499803 DOI: 10.1038/srep11529] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 05/20/2015] [Indexed: 11/30/2022] Open
Abstract
Fe65 is a brain-enriched adaptor protein known for its role in the action of the Aβ amyloid precursor protein in neuronal cells and Alzheimer’s disease, but little is known about its functions in cancer cells. The present study documents for the first time a role of Fe65 in suppressing breast cancer cell migration and invasion. Mechanistic studies suggest that the suppression is mediated through its phosphotyrosine binding domain 1 that mediates the recruitment of Tip60 to cortactin to stimulate its acetylation. The studies identify the Tip60 acetyltransferase as a cytoplasmic drug target for the therapeutic intervention of metastatic breast cancers.
Collapse
Affiliation(s)
- Yuefeng Sun
- Departments of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL 33612
| | - Jianwei Sun
- Comprehensive Melanoma Research Center and Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Panida Lungchukiet
- Departments of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL 33612
| | - Waise Quarni
- Departments of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL 33612
| | - Shengyu Yang
- Comprehensive Melanoma Research Center and Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Xiaohong Zhang
- 1] Departments of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL 33612 [2] Department of Oncological Sciences, University of South Florida Morsani College of Medicine, Tampa, FL 33612 [3] Program of Cancer Biology and Evolution, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Wenlong Bai
- 1] Departments of Pathology and Cell Biology, University of South Florida Morsani College of Medicine, Tampa, FL 33612 [2] Department of Oncological Sciences, University of South Florida Morsani College of Medicine, Tampa, FL 33612 [3] Program of Cancer Biology and Evolution, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| |
Collapse
|
44
|
Fe65 Ser228 is phosphorylated by ATM/ATR and inhibits Fe65-APP-mediated gene transcription. Biochem J 2015; 465:413-21. [PMID: 25397632 DOI: 10.1042/bj20140656] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Fe65 binds the amyloid precursor protein (APP) and regulates the secretase-mediated processing of APP into several proteolytic fragments, including amyloid β-peptides (Aβ) and APP intracellular domain (AICD). Aβ accumulation in neural plaques is a pathological feature of Alzheimer's disease (AD) and AICD has important roles in the regulation of gene transcription (in complex with Fe65). It is therefore important to understand how Fe65 is regulated and how this contributes to the function and/or processing of APP. Studies have also implicated Fe65 in the cellular DNA damage response with knockout mice showing increased DNA strand breaks and Fe65 demonstrating a gel mobility shift after DNA damage, consistent with protein phosphorylation. In the present study, we identified Fe65 Ser(228) as a novel target of the ATM (ataxia telangiectasia mutated) and ATR (ataxia-telangiectasia- and Rad3-related protein) protein kinases, in a reaction that occurred independently of APP. Neither phosphorylation nor mutation of Ser(228) affected the Fe65-APP complex, though this was markedly decreased after UV treatment, with a concomitant decrease in the protein levels of APP in cells. Finally, mutation of Ser(228) to alanine (thus blocking phosphorylation) caused a significant increase in Fe65-APP transcriptional activity, whereas phosphomimetic mutants (S(228)D and S(228)E) showed decreased transcriptional activity. These studies identify a novel phosphorylation site within Fe65 and a novel regulatory mechanism for the transcriptional activity of the Fe65-APP complex.
Collapse
|
45
|
Zheng C, Gu X, Zhong Z, Zhu R, Gao T, Wang F. Two memory associated genes regulated by amyloid precursor protein intracellular domain: Novel insights into the pathogenesis of learning and memory impairment in Alzheimer's disease. Neural Regen Res 2015; 7:341-6. [PMID: 25774172 PMCID: PMC4350115 DOI: 10.3969/j.issn.1673-5374.2012.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Accepted: 01/22/2012] [Indexed: 12/24/2022] Open
Abstract
In this study, we employed chromatin immunoprecipitation, a useful method for studying the locations of transcription factors bound to specific DNA regions in specific cells, to investigate amyloid precursor protein intracellular domain binding sites in chromatin DNA from hippocampal neurons of rats, and to screen out five putative genes associated with the learning and memory functions. The promoter regions of the calcium/calmodulin-dependent protein kinase II alpha and glutamate receptor-2 genes were amplified by PCR from DNA products immunoprecipitated by amyloid precursor protein intracellular domain. An electrophoretic mobility shift assay and western blot analysis suggested that the promoter regions of these two genes associated with learning and memory were bound by amyloid precursor protein intracellular domain (in complex form). Our experimental findings indicate that the amyloid precursor protein intracellular domain is involved in the transcriptional regulation of learning- and memory-associated genes in hippocampal neurons. These data may provide new insights into the molecular mechanism underlying the symptoms of progressive memory loss in Alzheimer's disease.
Collapse
Affiliation(s)
- Chuandong Zheng
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Xi Gu
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Zhimei Zhong
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Rui Zhu
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Tianming Gao
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| | - Fang Wang
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, Guangdong Province, China
| |
Collapse
|
46
|
Suh J, Moncaster JA, Wang L, Hafeez I, Herz J, Tanzi RE, Goldstein LE, Guénette SY. FE65 and FE65L1 amyloid precursor protein-binding protein compound null mice display adult-onset cataract and muscle weakness. FASEB J 2015; 29:2628-39. [PMID: 25757569 DOI: 10.1096/fj.14-261453] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 02/19/2015] [Indexed: 12/11/2022]
Abstract
FE65 and FE65L1 are cytoplasmic adaptor proteins that bind a variety of proteins, including the amyloid precursor protein, and that mediate the assembly of multimolecular complexes. We previously reported that FE65/FE65L1 double knockout (DKO) mice display disorganized laminin in meningeal fibroblasts and a cobblestone lissencephaly-like phenotype in the developing cortex. Here, we examined whether loss of FE65 and FE65L1 causes ocular and muscular deficits, 2 phenotypes that frequently accompany cobblestone lissencephaly. Eyes of FE65/FE65L1 DKO mice develop normally, but lens degeneration becomes apparent in young adult mice. Abnormal lens epithelial cell migration, widespread small vacuole formation, and increased laminin expression underneath lens capsules suggest impaired interaction between epithelial cells and capsular extracellular matrix in DKO lenses. Cortical cataracts develop in FE65L1 knockout (KO) mice aged 16 months or more but are absent in wild-type or FE65 KO mice. FE65 family KO mice show attenuated grip strength, and the nuclei of DKO muscle cells frequently locate in the middle of muscle fibers. These findings reveal that FE65 and FE65L1 are essential for the maintenance of lens transparency, and their loss produce phenotypes in brain, eye, and muscle that are comparable to the clinical features of congenital muscular dystrophies in humans.
Collapse
Affiliation(s)
- Jaehong Suh
- *Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, and Molecular Aging and Development Laboratory, Boston University School of Medicine, Boston, Massachusetts, USA; and Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Juliet A Moncaster
- *Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, and Molecular Aging and Development Laboratory, Boston University School of Medicine, Boston, Massachusetts, USA; and Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lirong Wang
- *Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, and Molecular Aging and Development Laboratory, Boston University School of Medicine, Boston, Massachusetts, USA; and Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Imran Hafeez
- *Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, and Molecular Aging and Development Laboratory, Boston University School of Medicine, Boston, Massachusetts, USA; and Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Joachim Herz
- *Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, and Molecular Aging and Development Laboratory, Boston University School of Medicine, Boston, Massachusetts, USA; and Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Rudolph E Tanzi
- *Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, and Molecular Aging and Development Laboratory, Boston University School of Medicine, Boston, Massachusetts, USA; and Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Lee E Goldstein
- *Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, and Molecular Aging and Development Laboratory, Boston University School of Medicine, Boston, Massachusetts, USA; and Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Suzanne Y Guénette
- *Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, and Molecular Aging and Development Laboratory, Boston University School of Medicine, Boston, Massachusetts, USA; and Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
47
|
Ryu S, Teles F, Minopoli G, Russo T, Rosenfeld MG, Suh Y. An epigenomic role of Fe65 in the cellular response to DNA damage. Mutat Res 2015; 776:40-7. [PMID: 26255939 DOI: 10.1016/j.mrfmmm.2015.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 01/13/2015] [Accepted: 01/17/2015] [Indexed: 01/02/2023]
Abstract
Previous findings describe Fe65 as a key protein in the cellular response to genotoxic stress. However, the precise molecular mechanism by which Fe65 contributes to DNA damage signaling remains unclear. In this study, we hypothesized that the transcriptional activity of Fe65 may contribute to DNA damage pathways by regulating gene expression patterns activated in response to genotoxic stress. To address this hypothesis, we mapped the global binding profile of Fe65 by chromatin immunoprecipitation (ChIP)-sequencing in the SK-N-SH cells exposed to genotoxic stress. Unexpectedly, the genome-wide location analysis showed a substantial enrichment of Fe65 in the promoter regions of coding genes linked to DNA damage signaling pathways. To further investigate the role of Fe65 in the transcriptional regulation of putative coding target genes identified by ChIP-seq, we performed microarray assays using wild-type (WT) or Fe65 deficient mouse embryonic fibroblasts (MEFs) exposed to oxidative stress with multiple recovery times. Gene ontology analysis of the Fe65-depedent transcriptome suggested that Fe65 modulates the expression of genes critical for DNA damage response. Motif enrichment analysis of regulatory regions occupied by Fe65 revealed a strong correlation with key transcription factors involved in DNA damage signaling pathways, including E2F1, p53, and Jun. Comparison of ChIP-sequencing results with microarray results ultimately identified 248 Fe65-depedent target genes, the majority of which were known regulators of cell cycle, cell death, and DNA replication and repair pathways. We validated the target genes identified by in silico analysis by qPCR experiments. Collectively, our results provide strong evidence that Fe65 plays a role in DNA damage response and cell viability by epigenomic regulation of specific transcriptional programs activated upon genotoxic stress.
Collapse
Affiliation(s)
- Seungjin Ryu
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Francesca Teles
- Howard Hughes Medical Institute and Graduate Program in Biomedical Sciences, University of California at San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Giuseppina Minopoli
- Dipartimento di Biochimica e Biotecnologie Mediche, Università di Napoli Federico II, Napoli, Italy
| | - Tommaso Russo
- Dipartimento di Biochimica e Biotecnologie Mediche, Università di Napoli Federico II, Napoli, Italy
| | - Michael G Rosenfeld
- Howard Hughes Medical Institute and Graduate Program in Biomedical Sciences, University of California at San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Yousin Suh
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute of Aging Research, Guangdong Medical College, Dongguan, China.
| |
Collapse
|
48
|
The multifaceted nature of amyloid precursor protein and its proteolytic fragments: friends and foes. Acta Neuropathol 2015; 129:1-19. [PMID: 25287911 DOI: 10.1007/s00401-014-1347-2] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Revised: 09/26/2014] [Accepted: 09/26/2014] [Indexed: 12/29/2022]
Abstract
The amyloid precursor protein (APP) has occupied a central position in Alzheimer's disease (AD) pathophysiology, in large part due to the seminal role of amyloid-β peptide (Aβ), a proteolytic fragment derived from APP. Although the contribution of Aβ to AD pathogenesis is accepted by many in the research community, recent studies have unveiled a more complicated picture of APP's involvement in neurodegeneration in that other APP-derived fragments have been shown to exert pathological influences on neuronal function. However, not all APP-derived peptides are neurotoxic, and some even harbor neuroprotective effects. In this review, we will explore this complex picture by first discussing the pleiotropic effects of the major APP-derived peptides cleaved by multiple proteases, including soluble APP peptides (sAPPα, sAPPβ), various C- and N-terminal fragments, p3, and APP intracellular domain fragments. In addition, we will highlight two interesting sequences within APP that likely contribute to this duality in APP function. First, it has been found that caspase-mediated cleavage of APP in the cytosolic region may release a cytotoxic peptide, C31, which plays a role in synapse loss and neuronal death. Second, recent studies have implicated the -YENPTY- motif in the cytoplasmic region as a domain that modulates several APP activities through phosphorylation and dephosphorylation of the first tyrosine residue. Thus, this review summarizes the current understanding of various APP proteolytic products and the interplay among them to gain deeper insights into the possible mechanisms underlying neurodegeneration and AD pathophysiology.
Collapse
|
49
|
Regulation of neuronal survival and morphology by the E3 ubiquitin ligase RNF157. Cell Death Differ 2014; 22:626-42. [PMID: 25342469 DOI: 10.1038/cdd.2014.163] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 08/19/2014] [Accepted: 08/29/2014] [Indexed: 01/03/2023] Open
Abstract
Neuronal health is essential for the long-term integrity of the brain. In this study, we characterized the novel E3 ubiquitin ligase ring finger protein 157 (RNF157), which displays a brain-dominant expression in mouse. RNF157 is a homolog of the E3 ligase mahogunin ring finger-1, which has been previously implicated in spongiform neurodegeneration. We identified RNF157 as a regulator of survival in cultured neurons and established that the ligase activity of RNF157 is crucial for this process. We also uncovered that independently of its ligase activity, RNF157 regulates dendrite growth and maintenance. We further identified the adaptor protein APBB1 (amyloid beta precursor protein-binding, family B, member 1 or Fe65) as an interactor and proteolytic substrate of RNF157 in the control of neuronal survival. Here, the nuclear localization of Fe65 together with its interaction partner RNA-binding protein SART3 (squamous cell carcinoma antigen recognized by T cells 3 or Tip110) is crucial to trigger apoptosis. In summary, we described that the E3 ligase RNF157 regulates important aspects of neuronal development.
Collapse
|
50
|
Tam JHK, Seah C, Pasternak SH. The Amyloid Precursor Protein is rapidly transported from the Golgi apparatus to the lysosome and where it is processed into beta-amyloid. Mol Brain 2014; 7:54. [PMID: 25085554 PMCID: PMC4237969 DOI: 10.1186/s13041-014-0054-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 07/23/2014] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by cerebral deposition of β-amyloid peptide (Aβ). Aβ is produced by sequential cleavage of the Amyloid Precursor Protein (APP) by β- and γ-secretases. Many studies have demonstrated that the internalization of APP from the cell surface can regulate Aβ production, although the exact organelle in which Aβ is produced remains contentious. A number of recent studies suggest that intracellular trafficking also plays a role in regulating Aβ production, but these pathways are relatively under-studied. The goal of this study was to elucidate the intracellular trafficking of APP, and to examine the site of intracellular APP processing. RESULTS We have tagged APP on its C-terminal cytoplasmic tail with photoactivatable Green Fluorescent Protein (paGFP). By photoactivating APP-paGFP in the Golgi, using the Golgi marker Galactosyltranferase fused to Cyan Fluorescent Protein (GalT-CFP) as a target, we are able to follow a population of nascent APP molecules from the Golgi to downstream compartments identified with compartment markers tagged with red fluorescent protein (mRFP or mCherry); including rab5 (early endosomes) rab9 (late endosomes) and LAMP1 (lysosomes). Because γ-cleavage of APP releases the cytoplasmic tail of APP including the photoactivated GFP, resulting in loss of fluorescence, we are able to visualize the cleavage of APP in these compartments. Using APP-paGFP, we show that APP is rapidly trafficked from the Golgi apparatus to the lysosome; where it is rapidly cleared. Chloroquine and the highly selective γ-secretase inhibitor, L685, 458, cause the accumulation of APP in lysosomes implying that APP is being cleaved by secretases in the lysosome. The Swedish mutation dramatically increases the rate of lysosomal APP processing, which is also inhibited by chloroquine and L685, 458. By knocking down adaptor protein 3 (AP-3; a heterotetrameric protein complex required for trafficking many proteins to the lysosome) using siRNA, we are able to reduce this lysosomal transport. Blocking lysosomal transport of APP reduces Aβ production by more than a third. CONCLUSION These data suggests that AP-3 mediates rapid delivery of APP to lysosomes, and that the lysosome is a likely site of Aβ production.
Collapse
Affiliation(s)
- Joshua HK Tam
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, 100 Perth Drive, London N6A 5K8, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, the University of Western Ontario, London N6A 5K8, Ontario, Canada
| | - Claudia Seah
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, 100 Perth Drive, London N6A 5K8, Ontario, Canada
| | - Stephen H Pasternak
- J. Allyn Taylor Centre for Cell Biology, Molecular Brain Research Group, Robarts Research Institute, 100 Perth Drive, London N6A 5K8, Ontario, Canada
- Department of Clinical Neurological Sciences, London N6A 5K8, Ontario, Canada
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, the University of Western Ontario, London N6A 5K8, Ontario, Canada
| |
Collapse
|