1
|
Dho SE, Othman K, Zhang Y, McGlade CJ. NUMB alternative splicing and isoform-specific functions in development and disease. J Biol Chem 2025; 301:108215. [PMID: 39863103 PMCID: PMC11889595 DOI: 10.1016/j.jbc.2025.108215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/06/2025] [Accepted: 01/11/2025] [Indexed: 01/27/2025] Open
Abstract
The NUMB gene encodes a conserved adaptor protein with roles in asymmetric cell division and cell fate determination. First described as an inhibitor of Notch signaling, multifunctional NUMB proteins regulate multiple cellular pathways through protein complexes with ubiquitin ligases, polarity proteins and the endocytic machinery. The vertebrate NUMB protein isoforms were identified over 2 decades ago, yet the majority of functional studies exploring NUMB function in endocytosis, cell migration and adhesion, development and disease have largely neglected the potential for distinct isoform activity in design and interpretation. In this review we consolidate the literature that has directly addressed individual NUMB isoform functions, as well as interpret other functional studies through the lens of the specific isoforms that were utilized. We also summarize the emerging literature on the mechanisms that regulate alternative splicing of NUMB, and how this is subverted in disease. Finally, the importance of relative NUMB isoform expression as a determinant of activity and considerations for future studies of NUMB isoforms as unique proteins with distinct functions are discussed.
Collapse
Affiliation(s)
- Sascha E Dho
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Kamal Othman
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Yangjing Zhang
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - C Jane McGlade
- The Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
2
|
Kim B, Dabin LC, Tate MD, Karahan H, Sharify AD, Acri DJ, Al-Amin MM, Philtjens S, Smith DC, Wijeratne HRS, Park JH, Jucker M, Kim J. Effects of SPI1-mediated transcriptome remodeling on Alzheimer's disease-related phenotypes in mouse models of Aβ amyloidosis. Nat Commun 2024; 15:3996. [PMID: 38734693 PMCID: PMC11088624 DOI: 10.1038/s41467-024-48484-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
SPI1 was recently reported as a genetic risk factor for Alzheimer's disease (AD) in large-scale genome-wide association studies. However, it is unknown whether SPI1 should be downregulated or increased to have therapeutic benefits. To investigate the effect of modulating SPI1 levels on AD pathogenesis, we performed extensive biochemical, histological, and transcriptomic analyses using both Spi1-knockdown and Spi1-overexpression mouse models. Here, we show that the knockdown of Spi1 expression significantly exacerbates insoluble amyloid-β (Aβ) levels, amyloid plaque deposition, and gliosis. Conversely, overexpression of Spi1 significantly ameliorates these phenotypes and dystrophic neurites. Further mechanistic studies using targeted and single-cell transcriptomics approaches demonstrate that altered Spi1 expression modulates several pathways, such as immune response pathways and complement system. Our data suggest that transcriptional reprogramming by targeting transcription factors, like Spi1, might hold promise as a therapeutic strategy. This approach could potentially expand the current landscape of druggable targets for AD.
Collapse
Affiliation(s)
- Byungwook Kim
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Luke Child Dabin
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mason Douglas Tate
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hande Karahan
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Ahmad Daniel Sharify
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Dominic J Acri
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Md Mamun Al-Amin
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Stéphanie Philtjens
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Daniel Curtis Smith
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - H R Sagara Wijeratne
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jung Hyun Park
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, IN, 47405, USA
- Program in Neuroscience, Indiana University, Bloomington, IN, 47405, USA
| | - Mathias Jucker
- German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Jungsu Kim
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Medical Neuroscience Graduate Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
3
|
Ortega-Campos SM, García-Heredia JM. The Multitasker Protein: A Look at the Multiple Capabilities of NUMB. Cells 2023; 12:333. [PMID: 36672267 PMCID: PMC9856935 DOI: 10.3390/cells12020333] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/08/2023] [Accepted: 01/13/2023] [Indexed: 01/18/2023] Open
Abstract
NUMB, a plasma membrane-associated protein originally described in Drosophila, is involved in determining cell function and fate during early stages of development. It is secreted asymmetrically in dividing cells, with one daughter cell inheriting NUMB and the other inheriting its antagonist, NOTCH. NUMB has been proposed as a polarizing agent and has multiple functions, including endocytosis and serving as an adaptor in various cellular pathways such as NOTCH, Hedgehog, and the P53-MDM2 axis. Due to its role in maintaining cellular homeostasis, it has been suggested that NUMB may be involved in various human pathologies such as cancer and Alzheimer's disease. Further research on NUMB could aid in understanding disease mechanisms and advancing the field of personalized medicine and the development of new therapies.
Collapse
Affiliation(s)
- Sara M. Ortega-Campos
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, 41013 Sevilla, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - José Manuel García-Heredia
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, 41013 Sevilla, Spain
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Departamento de Bioquímica Vegetal y Biología Molecular, Universidad de Sevilla, 41012 Sevilla, Spain
| |
Collapse
|
4
|
Sarnowski C, Ghanbari M, Bis JC, Logue M, Fornage M, Mishra A, Ahmad S, Beiser AS, Boerwinkle E, Bouteloup V, Chouraki V, Cupples LA, Damotte V, DeCarli CS, DeStefano AL, Djoussé L, Fohner AE, Franz CE, Kautz TF, Lambert JC, Lyons MJ, Mosley TH, Mukamal KJ, Pase MP, Portilla Fernandez EC, Rissman RA, Satizabal CL, Vasan RS, Yaqub A, Debette S, Dufouil C, Launer LJ, Kremen WS, Longstreth WT, Ikram MA, Seshadri S. Meta-analysis of genome-wide association studies identifies ancestry-specific associations underlying circulating total tau levels. Commun Biol 2022; 5:336. [PMID: 35396452 PMCID: PMC8993877 DOI: 10.1038/s42003-022-03287-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 03/17/2022] [Indexed: 01/27/2023] Open
Abstract
Circulating total-tau levels can be used as an endophenotype to identify genetic risk factors for tauopathies and related neurological disorders. Here, we confirmed and better characterized the association of the 17q21 MAPT locus with circulating total-tau in 14,721 European participants and identified three novel loci in 953 African American participants (4q31, 5p13, and 6q25) at P < 5 × 10-8. We additionally detected 14 novel loci at P < 5 × 10-7, specific to either Europeans or African Americans. Using whole-exome sequence data in 2,279 European participants, we identified ten genes associated with circulating total-tau when aggregating rare variants. Our genetic study sheds light on genes reported to be associated with neurological diseases including stroke, Alzheimer's, and Parkinson's (F5, MAP1B, and BCAS3), with Alzheimer's pathological hallmarks (ADAMTS12, IL15, and FHIT), or with an important function in the brain (PARD3, ELFN2, UBASH3B, SLIT3, and NSD3), and suggests that the genetic architecture of circulating total-tau may differ according to ancestry.
Collapse
Affiliation(s)
- Chloé Sarnowski
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mark Logue
- National Center for PTSD, Behavioral Sciences Division, VA Boston Healthcare System, Boston, MA, USA
- Department of Psychiatry and Biomedical Genetics, Boston University School of Medicine, Boston, MA, USA
| | - Myriam Fornage
- University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Aniket Mishra
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000, Bordeaux, France
| | - Shahzad Ahmad
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Alexa S Beiser
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
| | - Eric Boerwinkle
- University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Vincent Bouteloup
- Centre Inserm U1219 Bordeaux Population Health, CIC1401-EC, Institut de Santé Publique, d'Epidémiologie et de Développement, Université de Bordeaux, CHU de Bordeaux, Pôle Santé Publique, Bordeaux, France
| | - Vincent Chouraki
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE- LabEx DISTALZ - Risk factors and molecular determinants of aging diseases, F-59000, Lille, France
| | - L Adrienne Cupples
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
| | - Vincent Damotte
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE- LabEx DISTALZ - Risk factors and molecular determinants of aging diseases, F-59000, Lille, France
| | - Charles S DeCarli
- Department of Neurology and Center for Neuroscience, University of California at Davis, Davis, CA, USA
| | - Anita L DeStefano
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Luc Djoussé
- Department of Medicine, Division of Aging, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alison E Fohner
- Institute of Public Health Genetics and Department of Epidemiology and Cardiovascular Health Research Unit, University of Washington, Seattle, WA, USA
| | - Carol E Franz
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, USA
| | - Tiffany F Kautz
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Jean-Charles Lambert
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE- LabEx DISTALZ - Risk factors and molecular determinants of aging diseases, F-59000, Lille, France
| | - Michael J Lyons
- Department of Psychology and Brain Sciences, Boston University, Boston, MA, USA
| | | | - Kenneth J Mukamal
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Matthew P Pase
- Turner Institute for Brain and Mental Health, Monash University, Melbourne, VIC, Australia
- Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | | | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA
| | - Claudia L Satizabal
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Ramachandran S Vasan
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Preventive Medicine & Epidemiology, Boston University School of Medicine, Boston, MA, USA
| | - Amber Yaqub
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Stephanie Debette
- University of Bordeaux, Inserm, Bordeaux Population Health Research Center, team VINTAGE, UMR 1219, F-33000, Bordeaux, France
- Bordeaux University Hospital, Department of Neurology, Institute for Neurodegenerative Diseases, Bordeaux, France
| | - Carole Dufouil
- Bordeaux University Hospital, Department of Neurology, Institute for Neurodegenerative Diseases, Bordeaux, France
| | | | - William S Kremen
- Department of Psychiatry and Center for Behavior Genetics of Aging, University of California, San Diego, La Jolla, CA, USA
| | - William T Longstreth
- Departments of Neurology and Epidemiology, University of Washington, Seattle, WA, USA
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sudha Seshadri
- Boston University and the NHLBI's Framingham Heart Study, Boston, MA, USA
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| |
Collapse
|
5
|
Zou D, Li Q, Pan W, Chen P, Sun M, Bao X. A novel non‑selective atypical PKC agonist could protect neuronal cell line from Aβ‑oligomer induced toxicity by suppressing Aβ generation. Mol Med Rep 2022; 25:153. [PMID: 35244193 PMCID: PMC8941380 DOI: 10.3892/mmr.2022.12669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/15/2022] [Indexed: 12/04/2022] Open
Abstract
Atypical protein kinase C (aPKCs) serve key functions in embryonic development by regulating apical-basal polarity. Previous studies have shed light on their roles during adulthood, especially in the development of Alzheimer's disease (AD). Although the crystal structure of PKCι has been resolved, an agonist of aPKCs remains to be discovered. In the present study, by using the Discovery Studio program and LibDock methodology, a small molecule library (K66-X4436 KINA Set) of compounds were screened for potential binding to PKCι. Subsequently, the computational docking results were validated using affinity selection-mass spectrometry, before in vitro kinase activity was used to determine the function of the hit compounds. A cell-based model assay that can mimic the pathology of AD was then established and used to assess the function of these hit compounds. As a result, the aPKC agonist Z640 was identified, which could bind to PKCι in silico, in vitro and in this cell-based model. Z640 was further confirmed as a non-selective aPKC agonist that can activate the kinase activity of both PKCι and PKCζ. In the cell-based assay, Z640 was found to protect neuronal cell lines from amyloid-β (Aβ) oligomer-induced cell death by reducing reactive oxygen species production and restore mitochondrial function. In addition, Z640 could reduce Aβ40 generation in a dose-dependent manner and shift amyloid precursor protein processing towards the non-amyloid pathway. To conclude, the present study is the first, to the best of the authors' knowledge to identify an aPKC agonist by combining computer-assisted drug discovery and cell-based assays. The present study also revealed that aPKC agonists have therapeutic potential for the treatment of AD.
Collapse
Affiliation(s)
- Dongmei Zou
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Qian Li
- Department of Biology, College of Staten Island, Staten Island, NY 10314, USA
| | - Wenyang Pan
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Peng Chen
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Miao Sun
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiaofeng Bao
- School of Pharmacy, Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
6
|
Chen D, Yu W, Aitken L, Gunn-Moore F. Willin/FRMD6: A Multi-Functional Neuronal Protein Associated with Alzheimer's Disease. Cells 2021; 10:cells10113024. [PMID: 34831245 PMCID: PMC8616527 DOI: 10.3390/cells10113024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 12/18/2022] Open
Abstract
The FERM domain-containing protein 6 (FRMD6), also known as Willin, is an upstream regulator of Hippo signaling that has recently been shown to modulate actin cytoskeleton dynamics and mechanical phenotype of neuronal cells through ERK signaling. Physiological functions of Willin/FRMD6 in the nervous system include neuronal differentiation, myelination, nerve injury repair, and vesicle exocytosis. The newly established neuronal role of Willin/FRMD6 is of particular interest given the mounting evidence suggesting a role for Willin/FRMD6 in Alzheimer's disease (AD), including a series of genome wide association studies that position Willin/FRMD6 as a novel AD risk gene. Here we describe recent findings regarding the role of Willin/FRMD6 in the nervous system and its actions in cellular perturbations related to the pathogenesis of AD.
Collapse
|
7
|
Sun M, Bao W, Huang C, Xia Z, Zhang C, Wang G, Wang R, Li J, Roux S, Li Q, Zou D, Ma K, Bao X. A Novel Probiotic Formula, BIOCG, Protects Against Alzheimer's-Related Cognitive Deficits via Regulation of Dendritic Spine Dynamics. Curr Alzheimer Res 2021; 18:558-572. [PMID: 34674621 DOI: 10.2174/1567205018666211022091110] [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: 01/27/2021] [Revised: 06/27/2021] [Accepted: 08/23/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The brain-gut-microbiome axis has emerged as an important pathway through which perturbations in the gut and/or microbial microenvironment can impact neurological function. Such alterations have been implicated in a variety of neuropsychiatric disorders, includ- ing depression, anxiety, and Alzheimer's Disease (AD) and the use of probiotics as therapy for th- ese diseases remains promising. However, the mechanisms underlying the gut microenvironment's influence on disease pathogenesis and therapy remain unclear. OBJECTIVE The objective of this study is to investigate the effect of a novel probiotic formula, BIOCG, on cognitive function and pathobiological mechanisms, including amyloid processing and dendritic spine dynamics, in a mouse model of AD. METHODS BIOCG was administered for 3 months to 3xTg or 3xTg; Thy1-YFP AD mice and func- tional outcomes were assessed via behavioral testing and electrophysiology. Mechanisms relevant to AD pathogenesis including dendritic spine morphology and turnover, Amyloid Precursor Pro- tein (APP) processing and microglial phenotype were also evaluated. Finally, we sequenced fecal samples following probiotic treatment to assess the impact on gut microbial composition and corre- late the changes with the above described measures. RESULTS Mice treated with BIOCG demonstrated preserved cognitive abilities and stronger Long- Term Potentiation (LTP), spontaneous Excitatory Postsynaptic Currents (sEPSC), and glutamate-in- duced LTPs, indicative of functional and electrophysiological effects. Moreover, we observed atten- uated AD pathogenesis, including reduced Amyloid Beta (Aβ) burden, as well as more mature den- dritic spines in the BIOCG-treated. Our finding of changes in microglial number and phenotype in the treatment group suggests that this formulation may mediate its effects via attenuation of neu- roinflammation. Sequencing data confirmed that the gut microbiome in treated mice was more varied and harbored a greater proportion of "beneficial" bacteria. CONCLUSION Overall, our results indicate that treatment with BIOCG enhances microbial diversity and, through gut-brain axis interactions, attenuates neuroinflammation to produce histologic and functional improvement in AD pathogenesis.
Collapse
Affiliation(s)
- Miao Sun
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu. China
| | - Wenchenyang Bao
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu. China
| | - Chengyu Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu. China
| | - Ziyue Xia
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu. China
| | - Changliang Zhang
- Jiangsu Biodep Biotechnology, 6-C2 Dongsheng West Road, Jiangyin 214400, Jiangsu. China
| | - Guangxian Wang
- Jiangsu Biodep Biotechnology, 6-C2 Dongsheng West Road, Jiangyin 214400, Jiangsu. China
| | - Runxin Wang
- Jiangsu Biodep Biotechnology, 6-C2 Dongsheng West Road, Jiangyin 214400, Jiangsu. China
| | - Jiangyu Li
- Admera Health, South Plainfield, NJ07080. United States
| | - Shaun Roux
- Probiotics Australia, 24-30 Blanck Street, Ormeau, QLD, 4208. Australia
| | - Qian Li
- Department of biology, College of Staten Island, Staten Island, NY 10314 . United States
| | - Dongmei Zou
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu. China
| | - Kai Ma
- Jiangsu Biodep Biotechnology, 6-C2 Dongsheng West Road, Jiangyin 214400, Jiangsu. China
| | - Xiaofeng Bao
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu. China
| |
Collapse
|
8
|
Houssin E, Pinot M, Bellec K, Le Borgne R. Par3 cooperates with Sanpodo for the assembly of Notch clusters following asymmetric division of Drosophila sensory organ precursor cells. eLife 2021; 10:e66659. [PMID: 34596529 PMCID: PMC8516416 DOI: 10.7554/elife.66659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 09/25/2021] [Indexed: 12/15/2022] Open
Abstract
In multiple cell lineages, Delta-Notch signalling regulates cell fate decisions owing to unidirectional signalling between daughter cells. In Drosophila pupal sensory organ lineage, Notch regulates the intra-lineage pIIa/pIIb fate decision at cytokinesis. Notch and Delta that localise apically and basally at the pIIa-pIIb interface are expressed at low levels and their residence time at the plasma membrane is in the order of minutes. How Delta can effectively interact with Notch to trigger signalling from a large plasma membrane area remains poorly understood. Here, we report that the signalling interface possesses a unique apico-basal polarity with Par3/Bazooka localising in the form of nano-clusters at the apical and basal level. Notch is preferentially targeted to the pIIa-pIIb interface, where it co-clusters with Bazooka and its cofactor Sanpodo. Clusters whose assembly relies on Bazooka and Sanpodo activities are also positive for Neuralized, the E3 ligase required for Delta activity. We propose that the nano-clusters act as snap buttons at the new pIIa-pIIb interface to allow efficient intra-lineage signalling.
Collapse
Affiliation(s)
- Elise Houssin
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F- 35000RennesFrance
- Equipe Labellisée Ligue Nationale contre le cancerGlasgowUnited Kingdom
| | - Mathieu Pinot
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F- 35000RennesFrance
- Equipe Labellisée Ligue Nationale contre le cancerGlasgowUnited Kingdom
| | - Karen Bellec
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F- 35000RennesFrance
- Equipe Labellisée Ligue Nationale contre le cancerGlasgowUnited Kingdom
| | - Roland Le Borgne
- Univ Rennes, CNRS, IGDR (Institut de Génétique et Développement de Rennes) - UMR 6290, F- 35000RennesFrance
- Equipe Labellisée Ligue Nationale contre le cancerGlasgowUnited Kingdom
| |
Collapse
|
9
|
Gupta R, Sahu M, Srivastava D, Tiwari S, Ambasta RK, Kumar P. Post-translational modifications: Regulators of neurodegenerative proteinopathies. Ageing Res Rev 2021; 68:101336. [PMID: 33775891 DOI: 10.1016/j.arr.2021.101336] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/10/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022]
Abstract
One of the hallmark features in the neurodegenerative disorders (NDDs) is the accumulation of aggregated and/or non-functional protein in the cellular milieu. Post-translational modifications (PTMs) are an essential regulator of non-functional protein aggregation in the pathogenesis of NDDs. Any alteration in the post-translational mechanism and the protein quality control system, for instance, molecular chaperone, ubiquitin-proteasome system, autophagy-lysosomal degradation pathway, enhances the accumulation of misfolded protein, which causes neuronal dysfunction. Post-translational modification plays many roles in protein turnover rate, accumulation of aggregate and can also help in the degradation of disease-causing toxic metabolites. PTMs such as acetylation, glycosylation, phosphorylation, ubiquitination, palmitoylation, SUMOylation, nitration, oxidation, and many others regulate protein homeostasis, which includes protein structure, functions and aggregation propensity. Different studies demonstrated the involvement of PTMs in the regulation of signaling cascades such as PI3K/Akt/GSK3β, MAPK cascade, AMPK pathway, and Wnt signaling pathway in the pathogenesis of NDDs. Further, mounting evidence suggests that targeting different PTMs with small chemical molecules, which acts as an inhibitor or activator, reverse misfolded protein accumulation and thus enhances the neuroprotection. Herein, we briefly discuss the protein aggregation and various domain structures of different proteins involved in the NDDs, indicating critical amino acid residues where PTMs occur. We also describe the implementation and involvement of various PTMs on signaling cascade and cellular processes in NDDs. Lastly, we implement our current understanding of the therapeutic importance of PTMs in neurodegeneration, along with emerging techniques targeting various PTMs.
Collapse
|
10
|
Wu PF, Bhore N, Lee YL, Chou JY, Chen YW, Wu PY, Hsu WM, Lee H, Huang YS, Lu PJ, Liao YF. Phosphatidylinositol-4-phosphate 5-kinase type 1α attenuates Aβ production by promoting non-amyloidogenic processing of amyloid precursor protein. FASEB J 2020; 34:12127-12146. [PMID: 32686865 DOI: 10.1096/fj.202000113r] [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: 01/15/2020] [Revised: 06/17/2020] [Accepted: 06/29/2020] [Indexed: 11/11/2022]
Abstract
Alzheimer's disease (AD) is characterized by a chronic decline in cognitive function and is pathologically typified by cerebral deposition of amyloid-β peptide (Aβ). The production of Aβ is mediated by sequential proteolysis of amyloid precursor protein (APP) by β- and γ-secretases, and has been implicated as the essential determinant of AD pathology. Previous studies have demonstrated that the level of phosphatidylinositol-4,5-bisphosphate [PI(4,5)P2] in the membrane may potentially modulate Aβ production. Given that PI(4,5)P2 is produced by type 1 phosphatidylinositol-4-phosphate 5-kinases (PIP5Ks), we sought to determine whether the level of PIP5K type Iα (PIP5K1A) can affect production of Aβ by modulating the lipid composition of the membrane. Using a HEK-derived cell line that constitutively expresses yellow fluorescent protein-tagged APP (APP-YFP), we demonstrated that overexpression of PIP5K1A results in significant enhancement of non-amyloidogenic APP processing and a concomitant suppression of the amyloidogenic pathway, leading to a marked decrease in secreted Aβ. Consistently, cells overexpressing PIP5K1A exhibited a significant redistribution of APP-YFP from endosomal compartments to the cell surface. Our findings suggest that PIP5K1A may play a critical role in governing Aβ production by modulating membrane distribution of APP, and as such, the pathway may be a valuable therapeutic target for AD.
Collapse
Affiliation(s)
- Po-Fan Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.,TIGP in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei, Taiwan
| | - Noopur Bhore
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.,TIGP in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Yen-Lurk Lee
- TIGP in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ju-Yun Chou
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Yun-Wen Chen
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Pei-Yi Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Wen-Ming Hsu
- Department of Surgery, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Hsinyu Lee
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Yi-Shuian Huang
- TIGP in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei, Taiwan.,TIGP in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.,TIGP in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Pei-Jung Lu
- TIGP in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yung-Feng Liao
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.,TIGP in Interdisciplinary Neuroscience, National Cheng Kung University and Academia Sinica, Taipei, Taiwan.,TIGP in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| |
Collapse
|
11
|
Zhao Y, Kiss T, DelFavero J, Li L, Li X, Zheng L, Wang J, Jiang C, Shi J, Ungvari Z, Csiszar A, Zhang XA. CD82-TRPM7-Numb signaling mediates age-related cognitive impairment. GeroScience 2020; 42:595-611. [PMID: 32088828 PMCID: PMC7205934 DOI: 10.1007/s11357-020-00166-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/04/2019] [Indexed: 01/21/2023] Open
Abstract
Aging is a crucial cause of cognitive decline and a major risk factor for Alzheimer's disease (AD); however, AD's underlying molecular mechanisms remain unclear. Recently, tetraspanins have emerged as important modulators of synaptic function and memory. We demonstrate that the level of tetraspanin CD82 is upregulated in the brains of AD patients and middle-aged mice. In young adult mice, injection of AAV-CD82 to the hippocampus induced AD-like cognitive deficits and impairments in neuronal spine density. CD82 overexpression increased TRPM7 α-kinase cleavage via caspase-3 activation and induced Numb phosphorylation at Thr346 and Ser348 residues. CD82 overexpression promoted beta-amyloid peptide (Aβ) secretion which could be reversed by Numb T346S348 mutants. Importantly, hippocampus-related memory functions were improved in Cd82-/- mice. Taken together, our findings provide the evidence that links the elevated CD82-TRPM7-Numb signaling to age-related cognitive impairment.
Collapse
Affiliation(s)
- Yin Zhao
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatrics, University of Oklahoma Health Sciences Center, Oklahoma City, 73104, USA
| | - Jordan DelFavero
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatrics, University of Oklahoma Health Sciences Center, Oklahoma City, 73104, USA
| | - Lu Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xing Li
- Department of Neurobiology, Key Laboratory of Neurological Diseases, Ministry of Education, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lu Zheng
- Department of Neurobiology, Key Laboratory of Neurological Diseases, Ministry of Education, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jie Wang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, 73104, USA
| | - Chao Jiang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, 73104, USA
| | - Jing Shi
- Department of Neurobiology, Key Laboratory of Neurological Diseases, Ministry of Education, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatrics, University of Oklahoma Health Sciences Center, Oklahoma City, 73104, USA
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatrics, University of Oklahoma Health Sciences Center, Oklahoma City, 73104, USA
| | - Xin A Zhang
- Stephenson Cancer Center and Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, 73104, USA.
| |
Collapse
|
12
|
Enhancing α-secretase Processing for Alzheimer's Disease-A View on SFRP1. Brain Sci 2020; 10:brainsci10020122. [PMID: 32098349 PMCID: PMC7071437 DOI: 10.3390/brainsci10020122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 12/24/2022] Open
Abstract
Amyloid β (Aβ) peptides generated via sequential β- and γ-secretase processing of the amyloid precursor protein (APP) are major etiopathological agents of Alzheimer's disease (AD). However, an initial APP cleavage by an α-secretase, such as the a disintegrin and metalloproteinase domain-containing protein ADAM10, precludes β-secretase cleavage and leads to APP processing that does not produce Aβ. The latter appears to underlie the disease symptom-attenuating effects of a multitude of experimental therapeutics in AD animal models. Recent work has indicated that an endogenous inhibitor of ADAM10, secreted-frizzled-related protein 1 (SFRP1), is elevated in human AD brains and associated with amyloid plaques in mouse AD models. Importantly, genetic or functional attenuation of SFRP1 lowered Aβ accumulation and improved AD-related histopathological and neurological traits. Given SFRP1's well-known activity in attenuating Wnt signaling, which is also commonly impaired in AD, SFRP1 appears to be a promising therapeutic target for AD. This idea, however, needs to be addressed with care because of cancer enhancement potentials resulting from a systemic loss of SFRP1 activity, as well as an upregulation of ADAM10 activity. In this focused review, I shall discuss α-secretase-effected APP processing in AD with a focus on SFRP1, and explore the contrasting perspectives arising from the recent findings.
Collapse
|
13
|
The Endolysosomal System and Proteostasis: From Development to Degeneration. J Neurosci 2019; 38:9364-9374. [PMID: 30381428 DOI: 10.1523/jneurosci.1665-18.2018] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 12/12/2022] Open
Abstract
How do neurons adapt their endolysosomal system to address the particular challenge of membrane transport across their elaborate cellular landscape and to maintain proteostasis for the lifetime of the organism? Here we review recent findings that address this central question. We discuss the cellular and molecular mechanisms of endolysosomal trafficking and the autophagy pathway in neurons, as well as their role in neuronal development and degeneration. These studies highlight the importance of understanding the basic cell biology of endolysosomal trafficking and autophagy and their roles in the maintenance of proteostasis within the context of neurons, which will be critical for developing effective therapies for various neurodevelopmental and neurodegenerative disorders.
Collapse
|
14
|
The role of PTB domain containing adaptor proteins on PICALM-mediated APP endocytosis and localization. Biochem J 2019; 476:2093-2109. [DOI: 10.1042/bcj20180840] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 07/01/2019] [Accepted: 07/12/2019] [Indexed: 02/06/2023]
Abstract
AbstractOne hallmark of Alzheimer's disease (AD) is the presence of amyloid plaques, which mainly consist of the amyloid precursor protein (APP) cleavage product amyloid β (Aβ). For cleavage to occur, the APP must be endocytosed from the cell surface. The phosphatidylinositol binding clathrin assembly protein (PICALM) is involved in clathrin-mediated endocytosis and polymorphisms in and near the gene locus were identified as genetic risk factors for AD. PICALM overexpression enhances APP internalization and Aβ production. Furthermore, PICALM shuttles into the nucleus, but its function within the nucleus is still unknown. Using co-immunoprecipitation, we demonstrated an interaction between PICALM and APP, which is abrogated by mutation of the APP NPXY-motif. Since the NPXY-motif is an internalization signal that binds to phosphotryrosine-binding domain-containing adaptor proteins (PTB-APs), we hypothesized that PTB-APs can modulate the APP-PICALM interaction. We found that interaction between PICALM and the PTB-APs (Numb, JIP1b and GULP1) enhances the APP-PICALM interaction. Fluorescence activated cell sorting analysis and internalization assays revealed differentially altered APP cell surface levels and endocytosis rates that depended upon the presence of PICALM and co-expression of distinct PTB-APs. Additionally, we were able to show an impact of PICALM nuclear shuttling upon co-expression of PTB-APs and PICALM, with the magnitude of the effect depending on which PTB-AP was co-expressed. Taken together, our results indicate a modulating effect of PTB-APs on PICALM-mediated APP endocytosis and localization.
Collapse
|
15
|
Yuksel M, Tacal O. Trafficking and proteolytic processing of amyloid precursor protein and secretases in Alzheimer's disease development: An up-to-date review. Eur J Pharmacol 2019; 856:172415. [PMID: 31132354 DOI: 10.1016/j.ejphar.2019.172415] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/26/2019] [Accepted: 05/23/2019] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD), which is predicted to affect 1 in 85 persons worldwide by 2050, results in progressive loss of neuronal functions, leading to impairments in memory and cognitive abilities. As being one of the major neuropathological hallmarks of AD, senile plaques mainly consist of amyloid-β (Aβ) peptides, which are derived from amyloid precursor protein (APP) via the sequential cleavage by β- and γ-secretases. Although the overproduction and accumulation of Aβ peptides are at the center of AD research, the new discoveries point out to the complexity of the disease development. In this respect, it is crucial to understand the processing and the trafficking of APP, the enzymes involved in its processing, the cleavage products and their therapeutic potentials. This review summarizes the salient features of APP processing focusing on APP, the canonical secretases as well as the novel secretases and the cleavage products with an update of the recent developments. We also discussed the intracellular trafficking of APP and secretases in addition to their potential in AD therapy.
Collapse
Affiliation(s)
- Melike Yuksel
- Department of Biochemistry, School of Pharmacy, Hacettepe University, 06100, Ankara, Turkey.
| | - Ozden Tacal
- Department of Biochemistry, School of Pharmacy, Hacettepe University, 06100, Ankara, Turkey.
| |
Collapse
|
16
|
Sun M, Huang C, Wang H, Zhang H. Par3 regulates polarized convergence between APP and BACE1 in hippocampal neurons. Neurobiol Aging 2019; 77:87-93. [PMID: 30784815 DOI: 10.1016/j.neurobiolaging.2019.01.023] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 01/07/2019] [Accepted: 01/22/2019] [Indexed: 12/25/2022]
Abstract
The convergence between amyloid precursor protein (APP) and its β-secretase β-site APP cleaving enzyme 1 (BACE1) is a prerequisite for the generation of β-amyloid peptide, a key pathogenic agent for Alzheimer's disease. Yet the underlying molecular mechanisms regulating their convergence remain unclear. Here, we show that the polarity protein partitioning-defective 3 (Par3) regulates the polarized convergence between APP and BACE1 in hippocampal neurons. Par3 forms a complex with BACE1 through its first PDZ domain, which is important for regulating BACE1 endosome-to-TGN trafficking. In the absence of Par3, there is an increase in the convergence between internalized APP and BACE1. In hippocampal neurons, loss of Par3 leads to increased APP and BACE1 convergence in axons but not in dendrites. This polarized convergence mainly occurs in retrograde or stalled axonal late endocytic organelles and is likely due to compartment-specific regulation of APP trafficking by Par3. Together, our data show a novel function for Par3 in regulating polarized convergence between APP and BACE1 in hippocampal neurons.
Collapse
Affiliation(s)
- Miao Sun
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA.
| | - Chengyu Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Hui Wang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA; Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Huaye Zhang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA.
| |
Collapse
|
17
|
Huang Z, Liu Y, Huang X. Formononetin may protect aged hearts from ischemia/reperfusion damage by enhancing autophagic degradation. Mol Med Rep 2018; 18:4821-4830. [PMID: 30320398 PMCID: PMC6236296 DOI: 10.3892/mmr.2018.9544] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 05/01/2018] [Indexed: 01/18/2023] Open
Abstract
Myocardial infarction is a leading cause of mortality worldwide, and timely blood/oxygen reperfusion may substantially improve the outcome of infarction. However, ischemia/reperfusion (I/R) may cause severe side effects through excess reactive oxygen species generation. To develop novel methods to relieve I/R induced cell damage, the present study used a component of traditional Chinese medicine. In the present study, isolated heart tissue from aged mice and H9C2 cells with chemically‑induced aging were used as experimental subjects, and it was demonstrated that formononetin was able to alleviate I/R‑induced cell or tissue apoptosis. By applying formononetin to I/R‑damaged tissue or cells, it was demonstrated that formononetin was able to enhance autophagy and thus alleviate I/R‑induced cell damage. Furthermore, it was observed that I/R was able to inhibit lysosomal degradation processes in aged tissues or cells by impairing the lysosome acidification level, and formononetin was able to reverse this process via the re‑acidification of lysosomes. In conclusion, the present study demonstrated that formononetin was able to alleviate I/R‑induced cellular apoptosis in aged cells by facilitating autophagy.
Collapse
Affiliation(s)
- Zhengxin Huang
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Yingfeng Liu
- Department of Cardiology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510000, P.R. China
| | - Xianping Huang
- Laboratory of Biochemistry, Hunan University of Chinese Medicine, Changsha, Hunan 410000, P.R. China
| |
Collapse
|
18
|
Wang S, Zhong H, Lu M, Song G, Zhang X, Lin M, Yang S, Qian M. Higher Serum C Reactive Protein Determined C Reactive Protein Single-Nucleotide Polymorphisms Are Involved in Inherited Depression. Psychiatry Investig 2018; 15:824-828. [PMID: 30048584 PMCID: PMC6111223 DOI: 10.30773/pi.2018.04.03.2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 03/11/2018] [Accepted: 04/03/2018] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE The pathogenesis of depression is not fully understood yet, but studies have suggested higher circulating C reactive protein (CRP) level might relate to depression occurrence. However, due to high variability of patients' individual condition, the results to date are inconsistent. Considering CRP single-nucleotide polymorphisms (SNPs) could also regulate plasma CRP levels, in the present study, we hypothesized that inherited CRP allelic variations may co-vary with depressive symptomatology. METHODS We recruited 60 depression patients with family depression history and 60 healthy control volunteers into this project. We detected circulation CRP level as well as genome CRP SNPs from participants of this project. RESULTS We have found a significantly higher circulating CRP level in patients with a positive family history. Furthermore, we also identified some certain inherited CRP SNPs (A allele in rs1417938 and C allele in rs1205) could up regulate serum CRP level and distributed more in depression patients with family history. CONCLUSION Our finding may raise new evidence that genetically increased serum CRP level through SNPs variation is likely to induce family inherited depression.
Collapse
Affiliation(s)
| | - Hua Zhong
- Huzhou Third People’s Hospital, Zhejiang, China
| | - Meijuan Lu
- Huzhou Third People’s Hospital, Zhejiang, China
| | - Guohua Song
- Huzhou Third People’s Hospital, Zhejiang, China
| | | | - Min Lin
- Huzhou Third People’s Hospital, Zhejiang, China
| | | | - Mincai Qian
- Huzhou Third People’s Hospital, Zhejiang, China
| |
Collapse
|
19
|
Guimas Almeida C, Sadat Mirfakhar F, Perdigão C, Burrinha T. Impact of late-onset Alzheimer's genetic risk factors on beta-amyloid endocytic production. Cell Mol Life Sci 2018; 75:2577-2589. [PMID: 29704008 PMCID: PMC11105284 DOI: 10.1007/s00018-018-2825-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 04/04/2018] [Accepted: 04/23/2018] [Indexed: 12/21/2022]
Abstract
The increased production of the 42 aminoacids long beta-amyloid (Aβ42) peptide has been established as a causal mechanism of the familial early onset Alzheimer's disease (AD). In contrast, the causal mechanisms of the late-onset AD (LOAD), that affects most AD patients, remain to be established. Indeed, Aβ42 accumulation has been detected more than 30 years before diagnosis. Thus, the mechanisms that control Aβ accumulation in LOAD likely go awry long before pathogenesis becomes detectable. Early on, APOE4 was identified as the biggest genetic risk factor for LOAD. However, since APOE4 is not present in all LOAD patients, genome-wide association studies of thousands of LOAD patients were undertaken to identify other genetic variants that could explain the development of LOAD. PICALM, BIN1, CD2AP, SORL1, and PLD3 are now with APOE4 among the identified genes at highest risk in LOAD that have been implicated in Aβ42 production. Recent evidence indicates that the regulation of the endocytic trafficking of the amyloid precursor protein (APP) and/or its secretases to and from sorting endosomes is determinant for Aβ42 production. Thus, here, we will review the described mechanisms, whereby these genetic risk factors can contribute to the enhanced endocytic production of Aβ42. Dissecting causal LOAD mechanisms of Aβ42 accumulation, underlying the contribution of each genetic risk factor, will be required to identify therapeutic targets for novel personalized preventive strategies.
Collapse
Affiliation(s)
- Cláudia Guimas Almeida
- Neuronal Trafficking in Aging Lab, CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria, 130, 1169-056, Lisbon, Portugal.
| | - Farzaneh Sadat Mirfakhar
- Neuronal Trafficking in Aging Lab, CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria, 130, 1169-056, Lisbon, Portugal
| | - Catarina Perdigão
- Neuronal Trafficking in Aging Lab, CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria, 130, 1169-056, Lisbon, Portugal
| | - Tatiana Burrinha
- Neuronal Trafficking in Aging Lab, CEDOC, Chronic Diseases Research Centre, NOVA Medical School|Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo Mártires da Pátria, 130, 1169-056, Lisbon, Portugal
| |
Collapse
|
20
|
Yibulaiyin H, Sun H, Yang Y. Depression is Associated with CRP SNPs in Patients with Family History. Transl Neurosci 2017; 8:201-206. [PMID: 29340226 PMCID: PMC5765705 DOI: 10.1515/tnsci-2017-0027] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 10/17/2017] [Indexed: 02/02/2023] Open
Abstract
Objective The pathogenesis of depression is not fully understood, but studies have suggested that higher circulating levels of C reactive protein (CRP) might relate to depression occurrence. However, due to the highly variability of individual patients’ conditions, the results to date are inconsistent. Considering Single nucleotide polymorphisms (SNPs) of CRP gene have also been suggested to predict plasma CRP levels. In the present study, we hypothesize that inherited CRP allelic variations may co-vary with depressive symptomatology. Methods We recruited patients with a diagnosis of depression, with or without family depression history. We then detected serum CRP levels, as well as genome CRP SNPs from participants of this project. Results We found a significantly higher circulating CRP levels in patients with a positive family history. Furthermore, we also identified certain inherited CRP SNPs (A allele in rs1417938 and C allele in rs1205) which could up-regulate serum CRP levels and thus be associated with depression occurrence. Conclusion Our findings raise new evidence for the relationship between circulating CRP level and depression occurrence.
Collapse
Affiliation(s)
- Hasiyeti Yibulaiyin
- 2th affiliated hospital of Xinjiang Medical University, Department of neurology. Xinjiang Provence, Urumqi, China
- E-mail:
| | - Haixia Sun
- 421th hospital of PLA, Department of geriatrics. Urumqi, China
| | - Yue Yang
- 5th affiliated hospital of Xinjiang Medical University, Department of neurology, Urumqi, China
| |
Collapse
|
21
|
Sun M, Zhang H. Par3 and aPKC regulate BACE1 endosome-to-TGN trafficking through PACS1. Neurobiol Aging 2017; 60:129-140. [PMID: 28946017 PMCID: PMC5653456 DOI: 10.1016/j.neurobiolaging.2017.08.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 08/11/2017] [Accepted: 08/24/2017] [Indexed: 12/29/2022]
Abstract
The cleavage of amyloid precursor protein (APP) by β-site APP cleaving enzyme 1 (BACE1) is the rate-limiting step in beta amyloid generation during Alzheimer's disease (AD) pathogenesis. In AD brains, BACE1 is abnormally accumulated in endocytic compartments, where the acidic pH is optimal for its activity. However, mechanisms regulating the endosome-to-trans-Golgi network (TGN) retrieval of BACE1 remain unclear. Here, we show that partitioning defective 3 (Par3) facilitates BACE1 retrograde trafficking from endosomes to the TGN. Par3 functions through aPKC-mediated phosphorylation of BACE1 on Ser498, which in turn promotes the interaction between BACE1 and phosphofurin acidic cluster sorting protein 1 and facilitates the retrograde trafficking of BACE1 to the TGN. In human AD brains, there is a significant decrease in Ser498 phosphorylation of BACE1 suggesting that defective phosphorylation-dependent retrograde transport of BACE1 is important in AD pathogenesis. Together, our studies provide mechanistic insight into a novel role for Par3 and aPKC in regulating the retrograde endosome-to-TGN trafficking of BACE1 and shed light on the mechanisms of AD pathogenesis.
Collapse
Affiliation(s)
- Miao Sun
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Huaye Zhang
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ, USA.
| |
Collapse
|
22
|
Sun J, Roy S. The physical approximation of APP and BACE-1: A key event in alzheimer's disease pathogenesis. Dev Neurobiol 2017; 78:340-347. [PMID: 29106038 DOI: 10.1002/dneu.22556] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 10/17/2017] [Accepted: 11/01/2017] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is characterized by the accumulation of insoluble deposits of Amyloid β (Aβ) in brains. Aβ is derived by sequential cleavage of the amyloid precursor protein (APP) by β-site secretase enzyme (BACE-1) and γ-secretase. Proteolytic processing of APP by BACE-1 is the rate-limiting step in Aβ production, and this pathway is a prime target for AD drug development. Both APP and BACE-1 are membrane-spanning proteins, transported via secretory and endocytic pathways; and the physical interaction of APP and BACE-1 during trafficking is a key cell biological event initiating the amyloidogenic pathway. Here, we highlight recent research on intracellular trafficking/sorting of APP and BACE-1, and discuss how dysregulation of these pathways might lead to enhanced convergence of APP and BACE-1, and subsequent β-cleavage of APP. © 2018 Wiley Periodicals, Inc. Develop Neurobiol 78: 340-347, 2018.
Collapse
Affiliation(s)
- Jichao Sun
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin, 53705.,Department of Neuroscience, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin, 53705
| | - Subhojit Roy
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin, 53705.,Department of Neuroscience, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin, 53705
| |
Collapse
|
23
|
Ruch TR, Bryant DM, Mostov KE, Engel JN. Par3 integrates Tiam1 and phosphatidylinositol 3-kinase signaling to change apical membrane identity. Mol Biol Cell 2016; 28:252-260. [PMID: 27881661 PMCID: PMC5231894 DOI: 10.1091/mbc.e16-07-0541] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/16/2016] [Accepted: 11/18/2016] [Indexed: 01/31/2023] Open
Abstract
Pathogens can alter epithelial polarity by recruiting polarity proteins to the apical membrane, but how a change in protein localization is linked to polarity disruption is not clear. In this study, we used chemically induced dimerization to rapidly relocalize proteins from the cytosol to the apical surface. We demonstrate that forced apical localization of Par3, which is normally restricted to tight junctions, is sufficient to alter apical membrane identity through its interactions with phosphatidylinositol 3-kinase (PI3K) and the Rac1 guanine nucleotide exchange factor Tiam1. We further show that PI3K activity is required upstream of Rac1, and that simultaneously targeting PI3K and Tiam1 to the apical membrane has a synergistic effect on membrane remodeling. Thus, Par3 coordinates the action of PI3K and Tiam1 to define membrane identity, revealing a signaling mechanism that can be exploited by human mucosal pathogens.
Collapse
Affiliation(s)
- Travis R Ruch
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143
| | - David M Bryant
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158.,Cancer Research UK Beatson Institute, University of Glasgow, Glasgow G61 1BD, United Kingdom.,Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1BD, United Kingdom
| | - Keith E Mostov
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158.,Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158
| | - Joanne N Engel
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143
| |
Collapse
|