1
|
AmeliMojarad M, AmeliMojarad M, Wang J, Tavakolpour V, Shariati P. A pan-cancer study of ADAM9's immunological function and prognostic value particularly in liver cancer. Sci Rep 2024; 14:26862. [PMID: 39505907 PMCID: PMC11541887 DOI: 10.1038/s41598-024-76049-x] [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: 06/08/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024] Open
Abstract
A pan-cancer analysis summarizing the overall changes in mRNA and protein stability of ADM9, as well as its oncogenic function on immune cell line modulation and checkpoints within the tumor microenvironment (TME), is lacking, despite the fact that ADM9 up-regulation is correlated with the progression of many cancers. Therefore, in this study, we comprehensively analyzed the role of ADAM9 expression and its prognostic value in different cancers to fill this gap. Multiple bioinformatics databases such as Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), and Clinical Proteomic Tumor Analysis Consortium (CPTAC) were used to evaluate the ADAM9 genetic alternation, phosphorylation, and methylation, and indicated highly positive correlated genes that might play a critical interaction with ADAM9 and their molecular function with GO analysis. We also evaluate the effect of higher ADAM9 with prominent immune modulatory genes and immune infiltration especially in liver cancer pathogenesis stimulates lower NK cell effector functions based on its role in MICA shedding and increasing the Tregs infiltration. Immunohistochemistry (IHC) staining from 90 pathologically verified samples proved the positive correlation between ADAM9 and tumor stages and proved the higher expression of ADAM9 correlated genes (SNX9, APP, TNF, CDH1, ITGAV, MAD2L2) in HCC pathogenesis. In conclusion, this pan-cancer study provides a comprehensive understanding of the prognostic value of ADAM9 in various tumors emphasizing its importance to be considered as an innovative treatment approach, especially in tumor immunity shortly.
Collapse
Affiliation(s)
- Mandana AmeliMojarad
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran.
| | - Melika AmeliMojarad
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Jiang Wang
- Department of Bioinformatics, Nanjing Medical University, Nanjing, China
| | - Vahid Tavakolpour
- Department of Stem Cells and Regenerative Medicine, Faculty of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnolog, Tehran, Iran
- Stem cell Technology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Parvin Shariati
- Department of Bioprocess Engineering, Institute of Industrial and Environmental Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran.
| |
Collapse
|
2
|
Peralta Cuasolo YM, Dupraz S, Unsain N, Bisbal M, Quassollo G, Galiano MR, Grassi D, Quiroga S, Sosa LJ. The GTPase Rab21 is required for neuronal development and migration in the cerebral cortex. J Neurochem 2023; 166:790-808. [PMID: 37534523 DOI: 10.1111/jnc.15925] [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: 10/05/2022] [Revised: 07/04/2023] [Accepted: 07/12/2023] [Indexed: 08/04/2023]
Abstract
Development of the mammalian neocortex requires proper inside-out migration of developing cortical neurons from the germinal ventricular zone toward the cortical plate. The mechanics of this migration requires precise coordination of different cellular phenomena including cytoskeleton dynamics, membrane trafficking, and cell adhesion. The small GTPases play a central role in all these events. The small GTPase Rab21 regulates migration and neurite growth in developing neurons. Moreover, regulators and effectors of Rab21 have been implicated in brain pathologies with cortical malformations, suggesting a key function for the Rab21 signaling pathway in cortical development. Mechanistically, it has been posited that Rab21 influences cell migration by controlling the trafficking of endocytic vesicles containing adhesion molecules. However, direct evidence of the participation of Rab21 or its mechanism of action in the regulation of cortical migration is still incomplete. In this study, we demonstrate that Rab21 plays a critical role in the differentiation and migration of pyramidal neurons by regulating the levels of the amyloid precursor protein on the neuronal cell surface. Rab21 loss of function increased the levels of membrane-exposed APP, resulting in impaired cortical neuronal differentiation and migration. These findings further our understanding of the processes governing the development of the cerebral cortex and shed light onto the molecular mechanisms behind cortical development disorders derived from the malfunctioning of Rab21 signaling effectors.
Collapse
Affiliation(s)
- Yael Macarena Peralta Cuasolo
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Sebastián Dupraz
- Axonal Growth and Regeneration, German Center for Neurodegenarative Diseases, Bonn, Germany
| | - Nicolas Unsain
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Biología Celular y Molecular (CeBiCeM, FCEFyN-UNC), Facultad de Ciencias Exactas Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mariano Bisbal
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto Universitario Ciencias Biomédicas de Córdoba (IUCBC), Córdoba, Argentina
| | - Gonzalo Quassollo
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mauricio R Galiano
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Diego Grassi
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Santiago Quiroga
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Lucas Javier Sosa
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
3
|
Sajad M, Ahmed MM, Thakur SC. An integrated bioinformatics strategy to elucidate the function of hub genes linked to Alzheimer's disease. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101534] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
4
|
Eysert F, Coulon A, Boscher E, Vreulx AC, Flaig A, Mendes T, Hughes S, Grenier-Boley B, Hanoulle X, Demiautte F, Bauer C, Marttinen M, Takalo M, Amouyel P, Desai S, Pike I, Hiltunen M, Chécler F, Farinelli M, Delay C, Malmanche N, Hébert SS, Dumont J, Kilinc D, Lambert JC, Chapuis J. Alzheimer's genetic risk factor FERMT2 (Kindlin-2) controls axonal growth and synaptic plasticity in an APP-dependent manner. Mol Psychiatry 2021; 26:5592-5607. [PMID: 33144711 PMCID: PMC8758496 DOI: 10.1038/s41380-020-00926-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 10/02/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022]
Abstract
Although APP metabolism is being intensively investigated, a large fraction of its modulators is yet to be characterized. In this context, we combined two genome-wide high-content screenings to assess the functional impact of miRNAs and genes on APP metabolism and the signaling pathways involved. This approach highlighted the involvement of FERMT2 (or Kindlin-2), a genetic risk factor of Alzheimer's disease (AD), as a potential key modulator of axon guidance, a neuronal process that depends on the regulation of APP metabolism. We found that FERMT2 directly interacts with APP to modulate its metabolism, and that FERMT2 underexpression impacts axonal growth, synaptic connectivity, and long-term potentiation in an APP-dependent manner. Last, the rs7143400-T allele, which is associated with an increased AD risk and localized within the 3'UTR of FERMT2, induced a downregulation of FERMT2 expression through binding of miR-4504 among others. This miRNA is mainly expressed in neurons and significantly overexpressed in AD brains compared to controls. Altogether, our data provide strong evidence for a detrimental effect of FERMT2 underexpression in neurons and insight into how this may influence AD pathogenesis.
Collapse
Affiliation(s)
- Fanny Eysert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Audrey Coulon
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Emmanuelle Boscher
- Centre de Recherche du CHU de Québec-Université Laval, CHUL, Axe Neurosciences, Québec City, QC, Canada
- Faculté de Médecine, Département de Psychiatrie et de Neurosciences, Université Laval, Québec City, QC, Canada
| | - Anaїs-Camille Vreulx
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Amandine Flaig
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Tiago Mendes
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Sandrine Hughes
- E-Phy-Science, Bioparc de Sophia Antipolis, 2400 route des Colles, Biot, 06410, France
| | - Benjamin Grenier-Boley
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Xavier Hanoulle
- Université de Lille, CNRS, UMR8576-Labex DISTALZ, Villeneuve d'Ascq, 59655, France
| | - Florie Demiautte
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Charlotte Bauer
- Université Côte d'Azur, Inserm, CNRS, IPMC, DistAlz Laboratory of Excellence, Valbonne, France
| | - Mikael Marttinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Mari Takalo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Philippe Amouyel
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Shruti Desai
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Ian Pike
- Proteome Sciences plc, Hamilton House, London, WC1H 9BB, UK
| | - Mikko Hiltunen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Frédéric Chécler
- Université Côte d'Azur, Inserm, CNRS, IPMC, DistAlz Laboratory of Excellence, Valbonne, France
| | - Mélissa Farinelli
- E-Phy-Science, Bioparc de Sophia Antipolis, 2400 route des Colles, Biot, 06410, France
| | - Charlotte Delay
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Nicolas Malmanche
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Sébastien S Hébert
- Centre de Recherche du CHU de Québec-Université Laval, CHUL, Axe Neurosciences, Québec City, QC, Canada
- Faculté de Médecine, Département de Psychiatrie et de Neurosciences, Université Laval, Québec City, QC, Canada
| | - Julie Dumont
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Devrim Kilinc
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Jean-Charles Lambert
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France
| | - Julien Chapuis
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Lille, 59019, France.
| |
Collapse
|
5
|
Steubler V, Erdinger S, Back MK, Ludewig S, Fässler D, Richter M, Han K, Slomianka L, Amrein I, von Engelhardt J, Wolfer DP, Korte M, Müller UC. Loss of all three APP family members during development impairs synaptic function and plasticity, disrupts learning, and causes an autism-like phenotype. EMBO J 2021; 40:e107471. [PMID: 34008862 PMCID: PMC8204861 DOI: 10.15252/embj.2020107471] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 03/29/2021] [Accepted: 04/01/2021] [Indexed: 12/15/2022] Open
Abstract
The key role of APP for Alzheimer pathogenesis is well established. However, perinatal lethality of germline knockout mice lacking the entire APP family has so far precluded the analysis of its physiological functions for the developing and adult brain. Here, we generated conditional APP/APLP1/APLP2 triple KO (cTKO) mice lacking the APP family in excitatory forebrain neurons from embryonic day 11.5 onwards. NexCre cTKO mice showed altered brain morphology with agenesis of the corpus callosum and disrupted hippocampal lamination. Further, NexCre cTKOs revealed reduced basal synaptic transmission and drastically reduced long-term potentiation that was associated with reduced dendritic length and reduced spine density of pyramidal cells. With regard to behavior, lack of the APP family leads not only to severe impairments in a panel of tests for learning and memory, but also to an autism-like phenotype including repetitive rearing and climbing, impaired social communication, and deficits in social interaction. Together, our study identifies essential functions of the APP family during development, for normal hippocampal function and circuits important for learning and social behavior.
Collapse
Affiliation(s)
- Vicky Steubler
- Department of Functional GenomicsInstitute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityHeidelbergGermany
| | - Susanne Erdinger
- Department of Functional GenomicsInstitute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityHeidelbergGermany
| | - Michaela K Back
- Institute of PathophysiologyFocus Program Translational Neuroscience (FTN)University Medical Center of the Johannes Gutenberg University MainzMainzGermany
| | - Susann Ludewig
- Division of Cellular NeurobiologyZoological Institute, TU BraunschweigBraunschweigGermany
- Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration GroupBraunschweigGermany
| | - Dominique Fässler
- Department of Functional GenomicsInstitute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityHeidelbergGermany
| | - Max Richter
- Department of Functional GenomicsInstitute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityHeidelbergGermany
| | - Kang Han
- Department of Functional GenomicsInstitute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityHeidelbergGermany
| | - Lutz Slomianka
- Institute of Anatomy and Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| | - Irmgard Amrein
- Institute of Anatomy and Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| | - Jakob von Engelhardt
- Institute of PathophysiologyFocus Program Translational Neuroscience (FTN)University Medical Center of the Johannes Gutenberg University MainzMainzGermany
| | - David P Wolfer
- Institute of Anatomy and Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
- Institute of Human Movement SciencesETH ZurichZurichSwitzerland
| | - Martin Korte
- Division of Cellular NeurobiologyZoological Institute, TU BraunschweigBraunschweigGermany
- Helmholtz Centre for Infection Research, Neuroinflammation and Neurodegeneration GroupBraunschweigGermany
| | - Ulrike C Müller
- Department of Functional GenomicsInstitute of Pharmacy and Molecular BiotechnologyHeidelberg UniversityHeidelbergGermany
| |
Collapse
|
6
|
Plasma lipidome is dysregulated in Alzheimer's disease and is associated with disease risk genes. Transl Psychiatry 2021; 11:344. [PMID: 34092785 PMCID: PMC8180517 DOI: 10.1038/s41398-021-01362-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 03/10/2021] [Accepted: 04/06/2021] [Indexed: 01/11/2023] Open
Abstract
Lipidomics research could provide insights of pathobiological mechanisms in Alzheimer's disease. This study explores a battery of plasma lipids that can differentiate Alzheimer's disease (AD) patients from healthy controls and determines whether lipid profiles correlate with genetic risk for AD. AD plasma samples were collected from the Sydney Memory and Ageing Study (MAS) Sydney, Australia (aged range 75-97 years; 51.2% male). Untargeted lipidomics analysis was performed by liquid chromatography coupled-mass spectrometry (LC-MS/MS). We found that several lipid species from nine lipid classes, particularly sphingomyelins (SMs), cholesterol esters (ChEs), phosphatidylcholines (PCs), phosphatidylethanolamines (PIs), phosphatidylinositols (PIs), and triglycerides (TGs) are dysregulated in AD patients and may help discriminate them from healthy controls. However, when the lipid species were grouped together into lipid subgroups, only the DG group was significantly higher in AD. ChEs, SMs, and TGs resulted in good classification accuracy using the Glmnet algorithm (elastic net penalization for the generalized linear model [glm]) with more than 80% AUC. In general, group lipids and the lipid subclasses LPC and PE had less classification accuracy compared to the other subclasses. We also found significant increases in SMs, PIs, and the LPE/PE ratio in human U251 astroglioma cell lines exposed to pathophysiological concentrations of oligomeric Aβ42. This suggests that oligomeric Aβ42 plays a contributory, if not causal role, in mediating changes in lipid profiles in AD that can be detected in the periphery. In addition, we evaluated the association of plasma lipid profiles with AD-related single nucleotide polymorphisms (SNPs) and polygenic risk scores (PRS) of AD. We found that FERMT2 and MS4A6A showed a significantly differential association with lipids in all lipid classes across disease and control groups. ABCA7 had a differential association with more than half of the DG lipids (52.63%) and PI lipids (57.14%), respectively. Additionally, 43.4% of lipids in the SM class were differentially associated with CLU. More than 30% of lipids in ChE, PE, and TG classes had differential associations with separate genes (ChE-PICALM, SLC24A4, and SORL1; PE-CLU and CR1; TG-BINI) between AD and control group. These data may provide renewed insights into the pathobiology of AD and the feasibility of identifying individuals with greater AD risk.
Collapse
|
7
|
da Rocha JF, Bastos L, Domingues SC, Bento AR, Konietzko U, da Cruz E Silva OAB, Vieira SI. APP Binds to the EGFR Ligands HB-EGF and EGF, Acting Synergistically with EGF to Promote ERK Signaling and Neuritogenesis. Mol Neurobiol 2021; 58:668-688. [PMID: 33009641 DOI: 10.1007/s12035-020-02139-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 09/17/2020] [Indexed: 12/31/2022]
Abstract
The amyloid precursor protein (APP) is a transmembrane glycoprotein central to Alzheimer's disease (AD) with functions in brain development and plasticity, including in neurogenesis and neurite outgrowth. Epidermal growth factor (EGF) and heparin-binding EGF-like growth factor (HB-EGF) are well-described neurotrophic and neuromodulator EGFR ligands, both implicated in neurological disorders, including AD. Pro-HB-EGF arose as a putative novel APP interactor in a human brain cDNA library yeast two-hybrid screen. Based on their structural and functional similarities, we first aimed to verify if APP could bind to (HB-)EGF proforms. Here, we show that APP interacts with these two EGFR ligands, and further characterized the effects of APP-EGF interaction in ERK activation and neuritogenesis. Yeast co-transformation and co-immunoprecipitation assays confirmed APP interaction with HB-EGF. Co-immunoprecipitation also revealed that APP binds to cellular pro-EGF. Overexpression of HB-EGF in HeLa cells, or exposure of SH-SY5Y cells to EGF, both resulted in increased APP protein levels. EGF and APP were observed to synergistically activate the ERK pathway, crucial for neuronal differentiation. Immunofluorescence analysis of cellular neuritogenesis in APP overexpression and EGF exposure conditions confirmed a synergistic effect in promoting the number and the mean length of neurite-like processes. Synergistic ERK activation and neuritogenic effects were completely blocked by the EGFR inhibitor PD 168393, implying APP/EGF-induced activation of EGFR as part of the mechanism. This work shows novel APP protein interactors and provides a major insight into the APP/EGF-driven mechanisms underlying neurite outgrowth and neuronal differentiation, with potential relevance for AD and for adult neuroregeneration.
Collapse
Affiliation(s)
- Joana F da Rocha
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Agra do Crasto, 3810-193, Aveiro, Portugal
| | - Luísa Bastos
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Agra do Crasto, 3810-193, Aveiro, Portugal
- Roche Sistemas de Diagnósticos, Lda, 2720-413, Amadora, Portugal
| | - Sara C Domingues
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Agra do Crasto, 3810-193, Aveiro, Portugal
| | - Ana R Bento
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Agra do Crasto, 3810-193, Aveiro, Portugal
| | - Uwe Konietzko
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Odete A B da Cruz E Silva
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Agra do Crasto, 3810-193, Aveiro, Portugal
| | - Sandra I Vieira
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Agra do Crasto, 3810-193, Aveiro, Portugal.
| |
Collapse
|
8
|
Asparagine Endopeptidase (δ Secretase), an Enzyme Implicated in Alzheimer's Disease Pathology, Is an Inhibitor of Axon Regeneration in Peripheral Nerves. eNeuro 2021; 8:ENEURO.0155-20.2020. [PMID: 33323399 PMCID: PMC7814480 DOI: 10.1523/eneuro.0155-20.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 11/20/2020] [Accepted: 11/23/2020] [Indexed: 01/11/2023] Open
Abstract
Asparagine endopeptidase (AEP) is a lysosomal protease implicated in the pathology of Alzheimer’s disease (AD). It is known to cleave the axonal microtubule associated protein, Tau, and amyloid precursor protein (APP), both of which might impede axon regeneration following peripheral nerve injury (PNI). Active AEP, AEP-cleaved fragments of Tau (Tau N368), and APP (APP N585) were found in injured peripheral nerves. In AEP null mice, elongation of regenerating axons after sciatic nerve transection and repair was increased relative to wild-type (WT) controls. Compound muscle action potentials (M responses) were restored in reinnervated muscles twice as fast after injury in AEP knock-out (KO) mice as WT controls. Neurite elongation in cultures of adult dorsal root ganglion (DRG) neurons derived from AEP KO mice was increased significantly relative to cultures from WT controls. In AEP KO mice exposed to 1 h of 20-Hz electrical stimulation (ES) at the time of nerve injury, no further enhancement of axon regeneration was observed. These findings support inhibition of AEP as a therapeutic target to enhance axon regeneration after PNI.
Collapse
|
9
|
Halakos EG, Connell AJ, Glazewski L, Wei S, Mason RW. Bottom up proteomics identifies neuronal differentiation pathway networks activated by cathepsin inhibition treatment in neuroblastoma cells that are enhanced by concurrent 13-cis retinoic acid treatment. J Proteomics 2020; 232:104068. [PMID: 33278663 DOI: 10.1016/j.jprot.2020.104068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/16/2020] [Accepted: 11/29/2020] [Indexed: 12/19/2022]
Abstract
Neuroblastoma is the second most common pediatric cancer involving the peripheral nervous system in which stage IVS metastatic tumors regress due to spontaneous differentiation. 13-cis retinoic acid (13-cis RA) is currently used in the clinic for its differentiation effects and although it improves outcomes, relapse is seen in half of high-risk patients. Combinatorial therapies have been shown to be more effective in oncotherapy and since cathepsin inhibition reduces tumor growth, we explored the potential of coupling 13-cis RA with a cathepsin inhibitor (K777) to enhance therapeutic efficacy against neuroblastoma. Shotgun proteomics was used to identify proteins affected by K777 and dual (13-cis RA/K777) treatment in neuroblastoma SK-N-SH cells. Cathepsin inhibition was more effective in increasing proteins involved in neuronal differentiation and neurite outgrowth than 13-cis RA alone, but the combination of both treatments enhanced the neuronal differentiation effect. SIGNIFICANCE: As neuroblastoma can spontaneously differentiate, determining which proteins are involved in differentiation can guide development of more accurate diagnostic markers and more effective treatments. In this study, we established a differentiation proteomic map of SK-N-SH cells treated with a cathepsin inhibitor (K777) and K777/13-cis RA (dual). Bioinformatic analysis revealed these treatments enhanced neuronal differentiation and axonogenesis pathways. The most affected proteins in these pathways may become valuable biomarkers of efficacy of drugs designed to enhance differentiation of neuroblastoma [1].
Collapse
Affiliation(s)
- Effie G Halakos
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Andrew J Connell
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Lisa Glazewski
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA
| | - Shuo Wei
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Robert W Mason
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
10
|
Ristori E, Cicaloni V, Salvini L, Tinti L, Tinti C, Simons M, Corti F, Donnini S, Ziche M. Amyloid-β Precursor Protein APP Down-Regulation Alters Actin Cytoskeleton-Interacting Proteins in Endothelial Cells. Cells 2020; 9:cells9112506. [PMID: 33228083 PMCID: PMC7699411 DOI: 10.3390/cells9112506] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 11/16/2022] Open
Abstract
The amyloid-β precursor protein (APP) is a ubiquitous membrane protein often associated with Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA). Despite its role in the development of the pathogenesis, APP exerts several physiological roles that have been mainly investigated in neuronal tissue. To date, the role of APP in vasculature and endothelial cells has not been fully elucidated. In this study, we used molecular and proteomic approaches to identify and investigate major cellular targets of APP down-regulation in endothelial cells. We found that APP is necessary for endothelial cells proliferation, migration and adhesion. The loss of APP alters focal adhesion stability and cell-cell junctions' expression. Moreover, APP is necessary to mediate endothelial response to the VEGF-A growth factor. Finally, we document that APP propagates exogenous stimuli and mediates cellular response in endothelial cells by modulating the Scr/FAK signaling pathway. Thus, the intact expression and processing of APP is required for normal endothelial function. The identification of molecular mechanisms responsible for vasoprotective properties of endothelial APP may have an impact on clinical efforts to preserve and protect healthy vasculature in patients at risk of the development of cerebrovascular disease and dementia including AD and CAA.
Collapse
Affiliation(s)
- Emma Ristori
- Department of Life Science, University of Siena, 53100 Siena, Italy;
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Vittoria Cicaloni
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Laura Salvini
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Laura Tinti
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Cristina Tinti
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
| | - Michael Simons
- Yale Cardiovascular Research Center, 300 George Street, New Haven, CT 06511, USA; (M.S.); (F.C.)
- Departments of Medicine (Cardiology) and Cell Biology, Yale School of Medicine, New Haven, CT 06511, USA
| | - Federico Corti
- Yale Cardiovascular Research Center, 300 George Street, New Haven, CT 06511, USA; (M.S.); (F.C.)
| | - Sandra Donnini
- Department of Life Science, University of Siena, 53100 Siena, Italy;
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
- Correspondence: (S.D.); (M.Z.); Tel.: +39-0577-235382 (S.D.)
| | - Marina Ziche
- Toscana Life Sciences Foundation, 53100 Siena, Italy; (V.C.); (L.S.); (L.T.); (C.T.)
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
- Correspondence: (S.D.); (M.Z.); Tel.: +39-0577-235382 (S.D.)
| |
Collapse
|
11
|
The Interaction Between Contactin and Amyloid Precursor Protein and Its Role in Alzheimer’s Disease. Neuroscience 2020; 424:184-202. [DOI: 10.1016/j.neuroscience.2019.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 10/01/2019] [Accepted: 10/03/2019] [Indexed: 01/06/2023]
|
12
|
Network-based identification of genetic factors in ageing, lifestyle and type 2 diabetes that influence to the progression of Alzheimer's disease. INFORMATICS IN MEDICINE UNLOCKED 2020. [DOI: 10.1016/j.imu.2020.100309] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
13
|
Dourlen P, Kilinc D, Malmanche N, Chapuis J, Lambert JC. The new genetic landscape of Alzheimer's disease: from amyloid cascade to genetically driven synaptic failure hypothesis? Acta Neuropathol 2019; 138:221-236. [PMID: 30982098 PMCID: PMC6660578 DOI: 10.1007/s00401-019-02004-0] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 03/30/2019] [Accepted: 04/02/2019] [Indexed: 12/18/2022]
Abstract
A strong genetic predisposition (60–80% of attributable risk) is present in Alzheimer’s disease (AD). In view of this major genetic component, identification of the genetic risk factors has been a major objective in the AD field with the ultimate aim to better understand the pathological processes. In this review, we present how the genetic risk factors are involved in APP metabolism, β-amyloid peptide production, degradation, aggregation and toxicity, innate immunity, and Tau toxicity. In addition, on the basis of the new genetic landscape, resulting from the recent high-throughput genomic approaches and emerging neurobiological information, we propose an over-arching model in which the focal adhesion pathway and the related cell signalling are key elements in AD pathogenesis. The core of the focal adhesion pathway links the physiological functions of amyloid precursor protein and Tau with the pathophysiological processes they are involved in. This model includes several entry points, fitting with the different origins for the disease, and supports the notion that dysregulation of synaptic plasticity is a central node in AD. Notably, our interpretation of the latest data from genome wide association studies complements other hypotheses already developed in the AD field, i.e., amyloid cascade, cellular phase or propagation hypotheses. Genetically driven synaptic failure hypothesis will need to be further tested experimentally within the general AD framework.
Collapse
Affiliation(s)
- Pierre Dourlen
- Unité INSERM 1167, RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, Institut Pasteur de Lille, University of Lille, U1167-Excellence Laboratory LabEx DISTALZ, BP 245, 1, rue du professeur Calmette, 59019, Lille Cedex, France
| | - Devrim Kilinc
- Unité INSERM 1167, RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, Institut Pasteur de Lille, University of Lille, U1167-Excellence Laboratory LabEx DISTALZ, BP 245, 1, rue du professeur Calmette, 59019, Lille Cedex, France
| | - Nicolas Malmanche
- Unité INSERM 1167, RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, Institut Pasteur de Lille, University of Lille, U1167-Excellence Laboratory LabEx DISTALZ, BP 245, 1, rue du professeur Calmette, 59019, Lille Cedex, France
| | - Julien Chapuis
- Unité INSERM 1167, RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, Institut Pasteur de Lille, University of Lille, U1167-Excellence Laboratory LabEx DISTALZ, BP 245, 1, rue du professeur Calmette, 59019, Lille Cedex, France
| | - Jean-Charles Lambert
- Unité INSERM 1167, RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, Institut Pasteur de Lille, University of Lille, U1167-Excellence Laboratory LabEx DISTALZ, BP 245, 1, rue du professeur Calmette, 59019, Lille Cedex, France.
| |
Collapse
|
14
|
Differential Expression of mRNAs in the Brain Tissues of Patients with Alzheimer's Disease Based on GEO Expression Profile and Its Clinical Significance. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8179145. [PMID: 30918899 PMCID: PMC6413412 DOI: 10.1155/2019/8179145] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/28/2018] [Accepted: 02/04/2019] [Indexed: 12/18/2022]
Abstract
Background Early diagnosis of Alzheimer's disease (AD) is an urgent point for AD prevention and treatment. The biomarkers of AD still remain indefinite. Based on the bioinformatics analysis of mRNA differential expressions in the brain tissues and the peripheral blood samples of Alzheimer's disease (AD) patients, we investigated the target mRNAs that could be used as an AD biomarker and developed a new effective, practical clinical examination program. Methods We compared the AD peripheral blood mononuclear cells (PBMCs) expression dataset (GEO accession GSE4226 and GSE18309) with AD brain tissue expression datasets (GEO accessions GSE1297 and GSE5281) from GEO in the present study. The GEO gene database was used to download the appropriate gene expression profiles to analyze the differential mRNA expressions between brain tissue and blood of AD patients and normal elderly. The Venn diagram was used to screen out the differential expression of mRNAs between the brain tissue and blood. The protein-protein interaction network map (PPI) was used to view the correlation between the possible genes. GO (gene ontology) and KEGG (Kyoto Gene and Genomic Encyclopedia) were used for gene enrichment analysis to determine the major affected genes and the function or pathway. Results Bioinformatics analysis revealed that there were differentially expressed genes in peripheral blood and hippocampus of AD patients. There were 4958 differential mRNAs in GSE18309, 577 differential mRNAs in GSE4226 in AD PBMCs sample, 7464 differential mRNAs in GSE5281, and 317 differential mRNAs in GSE129 in AD brain tissues, when comparing between AD patients and healthy elderly. Two mRNAs of RAB7A and ITGB1 coexpressed in hippocampus and peripheral blood were screened. Furthermore, functions of differential genes were enriched by the PPI network map, GO, and KEGG analysis, and finally the chemotaxis, adhesion, and inflammatory reactions were found out, respectively. Conclusions ITGB1 and RAB7A mRNA expressions were both changed in hippocampus and PBMCs, highly suggested being used as an AD biomarker with AD. Also, according to the results of this analysis, it is indicated that we can test the blood routine of the elderly for 2-3 years at a frequency of 6 months or one year. When a patient continuously detects the inflammatory manifestations, it is indicated as a potentially high-risk AD patient for AD prevention.
Collapse
|
15
|
Coronel R, Palmer C, Bernabeu-Zornoza A, Monteagudo M, Rosca A, Zambrano A, Liste I. Physiological effects of amyloid precursor protein and its derivatives on neural stem cell biology and signaling pathways involved. Neural Regen Res 2019; 14:1661-1671. [PMID: 31169172 PMCID: PMC6585543 DOI: 10.4103/1673-5374.257511] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The pathological implication of amyloid precursor protein (APP) in Alzheimer's disease has been widely documented due to its involvement in the generation of amyloid-β peptide. However, the physiological functions of APP are still poorly understood. APP is considered a multimodal protein due to its role in a wide variety of processes, both in the embryo and in the adult brain. Specifically, APP seems to play a key role in the proliferation, differentiation and maturation of neural stem cells. In addition, APP can be processed through two canonical processing pathways, generating different functionally active fragments: soluble APP-α, soluble APP-β, amyloid-β peptide and the APP intracellular C-terminal domain. These fragments also appear to modulate various functions in neural stem cells, including the processes of proliferation, neurogenesis, gliogenesis or cell death. However, the molecular mechanisms involved in these effects are still unclear. In this review, we summarize the physiological functions of APP and its main proteolytic derivatives in neural stem cells, as well as the possible signaling pathways that could be implicated in these effects. The knowledge of these functions and signaling pathways involved in the onset or during the development of Alzheimer's disease is essential to advance the understanding of the pathogenesis of Alzheimer's disease, and in the search for potential therapeutic targets.
Collapse
Affiliation(s)
- Raquel Coronel
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Charlotte Palmer
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Adela Bernabeu-Zornoza
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - María Monteagudo
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Andreea Rosca
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Alberto Zambrano
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| | - Isabel Liste
- Unidad de Regeneración Neural, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain
| |
Collapse
|
16
|
Tsang JYS, Lee MA, Ni YB, Chan SK, Cheung SY, Chan WW, Lau KF, Tse GMK. Amyloid Precursor Protein Is Associated with Aggressive Behavior in Nonluminal Breast Cancers. Oncologist 2018; 23:1273-1281. [PMID: 30108157 PMCID: PMC6291326 DOI: 10.1634/theoncologist.2018-0012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/05/2018] [Accepted: 05/15/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND β-amyloid precursor protein (APP), a potential target for Alzheimer's disease treatment, has recently been shown to take part in carcinogenesis. Increased APP promotes migration, survival, and proliferation in breast cancer cell lines. We examined the clinical value of APP in breast cancers. A comprehensive examination of clinicopathological features related to APP expression in a large cohort of breast cancers and the corresponding metastatic lymph nodes was performed. APP expression and its prognostic impact in different breast cancer subtypes were examined. RESULTS APP was highly expressed in nonluminal breast cancers and correlated with features associated with nonluminal breast cancers (including higher grade, the presence of necrosis, and higher proliferative index, growth factor receptor, and basal marker expression). Multivariate Cox hazard analysis demonstrated that APP was an independent adverse prognostic factor of disease-free survival (DFS; hazard ratio [HR], 2.090; p = .013; 95% confidence interval [CI], 1.165-3.748) and breast cancer-specific survival (BCSS; HR, 2.631; p = .002; 95% CI, 1.408-4.915) in the nonluminal group. The independent prognostic impact was also seen in triple negative breast cancers. Interestingly, a higher expression of APP was found in nodal metastasis compared with primary tumor. Such APP upregulation was correlated with further distal metastasis and poorer outcome (DFS: log-rank, 12.848; p < .001; BCSS: log-rank, 13.947; p < .001). CONCLUSION Our findings provided evidence of oncogenic roles of APP in clinical breast cancers. Patients with positive APP expression, particularly those with APP upregulation in lymph node metastases, may require vigilant monitoring of their disease and more aggressive therapy. IMPLICATIONS FOR PRACTICE β-amyloid precursor protein (APP), a potential target for Alzheimer's disease, has recently been implicated in oncogenesis. Here, evidence of its roles in clinical breast cancers is provided. Positive APP expression was found to be an independent prognostic factor in nonluminal cancers, particularly triple negative breast cancers (TNBCs). Interestingly, a higher APP in nodal metastases was associated with distal metastases. TNBCs are heterogeneous and currently have no available target therapy. APP could have therapeutic potential and be used to define the more aggressive cases in TNBCs. Current prognostic analysis is based on primary tumor. The present data suggest that investigation of nodal metastases could provide additional prognostic value.
Collapse
Affiliation(s)
- Julia Y S Tsang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Michelle A Lee
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yun-Bi Ni
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Siu-Ki Chan
- Department of Pathology, Kwong Wah Hospital, Hong Kong
| | | | - Wai-Wa Chan
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kwok-Fai Lau
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Gary M K Tse
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
17
|
Distal Axonal Proteins and Their Related MiRNAs in Cultured Cortical Neurons. Mol Neurobiol 2018; 56:2703-2713. [PMID: 30054858 DOI: 10.1007/s12035-018-1266-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/18/2018] [Indexed: 12/15/2022]
Abstract
Proteins and microRNAs (miRNAs) within the axon locally regulate axonal development. However, protein profiles of distal axons of cortical neurons have not been fully investigated. In particular, networks of genes encoding axonal proteins and their related miRNAs in sub compartments of neurons such as axons remain unknown. Using embryonic cortical neurons cultured in a microfluidic device and proteomic approaches, we found that distal axons contain 883 proteins. Bioinformatics analysis revealed that 94 out of these 883 proteins are related to regulating axonal growth. Of the 94 genes encoding these proteins, there were 56 candidate genes that can be putatively targeted by axon-enriched 62 miRNAs with 8mer sites that exactly match these target genes. Among them, we validated 11 proteins and 11 miRNAs, by means of western blot and RT-PCR, respectively. Treatment of distal axons with chondroitin sulfate proteoglycans (CSPGs) that inhibit axonal growth elevated miR-133b, -203a, -29a, and -92a, which were associated with reduced protein level of AKT, MTOR, PI3K, DPYSL2, MAP1B, and PPP2CA. In contrast, reduction of miR-128, -15b, -195, -26b, -34b, -376b, and -381 by CSPGs was accompanied by increased EZR, KIF5A, DCX, GSK3B, and ROCK2 proteins. In silico pathway analysis revealed an interconnected network of these miRNAs and protein coding genes that is highly related to regulating axonal growth. Our data provide new insights into networks of miRNAs and their related proteins in distal axons in mediating axonal growth.
Collapse
|
18
|
Mazi AR, Arzuman AS, Gurel B, Sahin B, Tuzuner MB, Ozansoy M, Baykal AT. Neonatal Neurodegeneration in Alzheimer's Disease Transgenic Mouse Model. J Alzheimers Dis Rep 2018; 2:79-91. [PMID: 30480251 PMCID: PMC6159732 DOI: 10.3233/adr-170049] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive disorder characterized by a variety of molecular pathologies causing cortical dementia with a prominent memory deficit. Formation of the pathology, which begins decades before the diagnosis of the disease, is highly correlated with the clinical symptoms. Several proteomics studies were performed using animal models to monitor the alterations of the brain tissue proteome at different stages of AD. However, proteome changes in the brain regions of newborn transgenic mouse model have not been investigated yet. To this end, we analyzed protein expression alterations in cortex, hippocampus and cerebellum of transgenic mice carrying five familial AD mutations (5XFAD) at neonatal day-1. Our results indicate a remarkable difference in protein expression profile of newborn 5XFAD brain with region specific variations. Additionally, the proteins, which show similar expression alteration pattern in postmortem human AD brains, were determined. Bioinformatics analysis showed that the molecular alterations were mostly related to the cell morphology, cellular assembly and organization, and neuroinflammation. Moreover, morphological analysis revealed that there is an increase in neurite number of 5XFAD mouse neurons in vitro. We suggest that, molecular alterations in the AD brain exist even at birth, and perhaps the disease is silenced until older ages when the brain becomes vulnerable.
Collapse
Affiliation(s)
- Aise Rumeysa Mazi
- Regenerative and Restorative Medicine Research Center, REMER, Istanbul, Turkey.,Institute of Health Science, Istanbul Medipol University, Istanbul, Turkey.,Department of Medical Biochemistry, Acibadem Mehmet Ali Aydinlar University, School of Medicine, Istanbul, Turkey
| | - Aysegul Sumeyye Arzuman
- Regenerative and Restorative Medicine Research Center, REMER, Istanbul, Turkey.,Institute of Health Science, Istanbul Medipol University, Istanbul, Turkey.,Department of Medical Biochemistry, Acibadem Mehmet Ali Aydinlar University, School of Medicine, Istanbul, Turkey
| | - Busra Gurel
- Regenerative and Restorative Medicine Research Center, REMER, Istanbul, Turkey.,Institute of Health Science, Istanbul Medipol University, Istanbul, Turkey.,Department of Medical Biochemistry, Acibadem Mehmet Ali Aydinlar University, School of Medicine, Istanbul, Turkey
| | - Betul Sahin
- Acibadem Labmed R&D Laboratory, Istanbul, Turkey
| | | | - Mehmet Ozansoy
- Regenerative and Restorative Medicine Research Center, REMER, Istanbul, Turkey.,Department of Physiology, International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ahmet Tarik Baykal
- Acibadem Labmed R&D Laboratory, Istanbul, Turkey.,Department of Medical Biochemistry, Acibadem Mehmet Ali Aydinlar University, School of Medicine, Istanbul, Turkey
| |
Collapse
|
19
|
Leija-Salazar M, Piette C, Proukakis C. Review: Somatic mutations in neurodegeneration. Neuropathol Appl Neurobiol 2018; 44:267-285. [PMID: 29369391 DOI: 10.1111/nan.12465] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/13/2018] [Indexed: 12/22/2022]
Abstract
Somatic mutations are postzygotic mutations which may lead to mosaicism, the presence of cells with genetic differences in an organism. Their role in cancer is well established, but detailed investigation in health and other diseases has only been recently possible. This has been empowered by the improvements of sequencing techniques, including single-cell sequencing, which can still be error-prone but is rapidly improving. Mosaicism appears relatively common in the human body, including the normal brain, probably arising in early development, but also potentially during ageing. In this review, we first discuss theoretical considerations and current evidence relevant to somatic mutations in the brain. We present a framework to explain how they may be integrated with current views on neurodegeneration, focusing mainly on sporadic late-onset neurodegenerative diseases (Parkinson's disease, Alzheimer's disease and amyotrophic lateral sclerosis). We review the relevant studies so far, with the first evidence emerging in Alzheimer's in particular. We also discuss the role of mosaicism in inherited neurodegenerative disorders, particularly somatic instability of tandem repeats. We summarize existing views and data to present a model whereby the time of origin and spatial distribution of relevant somatic mutations, combined with any additional risk factors, may partly determine the development and onset age of sporadic neurodegenerative diseases.
Collapse
Affiliation(s)
- M Leija-Salazar
- Department of Clinical Neuroscience, University College London Institute of Neurology, London, UK
| | - C Piette
- Department of Clinical Neuroscience, University College London Institute of Neurology, London, UK
| | - C Proukakis
- Department of Clinical Neuroscience, University College London Institute of Neurology, London, UK
| |
Collapse
|
20
|
Sosa LJ, Cáceres A, Dupraz S, Oksdath M, Quiroga S, Lorenzo A. The physiological role of the amyloid precursor protein as an adhesion molecule in the developing nervous system. J Neurochem 2017; 143:11-29. [PMID: 28677143 DOI: 10.1111/jnc.14122] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 06/28/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022]
Abstract
The amyloid precursor protein (APP) is a type I transmembrane glycoprotein better known for its participation in the physiopathology of Alzheimer disease as the source of the beta amyloid fragment. However, the physiological functions of the full length protein and its proteolytic fragments have remained elusive. APP was first described as a cell-surface receptor; nevertheless, increasing evidence highlighted APP as a cell adhesion molecule. In this review, we will focus on the current knowledge of the physiological role of APP as a cell adhesion molecule and its involvement in key events of neuronal development, such as migration, neurite outgrowth, growth cone pathfinding, and synaptogenesis. Finally, since APP is over-expressed in Down syndrome individuals because of the extra copy of chromosome 21, in the last section of the review, we discuss the potential contribution of APP to the neuronal and synaptic defects described in this genetic condition. Read the Editorial Highlight for this article on page 9. Cover Image for this issue: doi. 10.1111/jnc.13817.
Collapse
Affiliation(s)
- Lucas J Sosa
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Alfredo Cáceres
- Laboratorio Neurobiología, Instituto Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina.,Instituto Universitario Ciencias Biomédicas Córdoba, Córdoba, Argentina
| | - Sebastián Dupraz
- Axonal Growth and Regeneration, German Center for Neurodegenarative Diseases, Bonn, Germany
| | - Mariana Oksdath
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Santiago Quiroga
- Departamento de Química Biológica Ranwell Caputto, Facultad de Ciencias Químicas, CIQUIBIC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Alfredo Lorenzo
- Laboratorio de Neuropatología Experimental, Instituto de Investigación Médica Mercedes y Martín Ferreyra, INIMEC-CONICET-Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
21
|
Genome-wide, high-content siRNA screening identifies the Alzheimer's genetic risk factor FERMT2 as a major modulator of APP metabolism. Acta Neuropathol 2017; 133:955-966. [PMID: 27933404 PMCID: PMC5427165 DOI: 10.1007/s00401-016-1652-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 11/29/2016] [Accepted: 11/29/2016] [Indexed: 10/30/2022]
Abstract
Genome-wide association studies (GWASs) have identified 19 susceptibility loci for Alzheimer's disease (AD). However, understanding how these genes are involved in the pathophysiology of AD is one of the main challenges of the "post-GWAS" era. At least 123 genes are located within the 19 susceptibility loci; hence, a conventional approach (studying the genes one by one) would not be time- and cost-effective. We therefore developed a genome-wide, high-content siRNA screening approach and used it to assess the functional impact of gene under-expression on APP metabolism. We found that 832 genes modulated APP metabolism. Eight of these genes were located within AD susceptibility loci. Only FERMT2 (a β3-integrin co-activator) was also significantly associated with a variation in cerebrospinal fluid Aβ peptide levels in 2886 AD cases. Lastly, we showed that the under-expression of FERMT2 increases Aβ peptide production by raising levels of mature APP at the cell surface and facilitating its recycling. Taken as a whole, our data suggest that FERMT2 modulates the AD risk by regulating APP metabolism and Aβ peptide production.
Collapse
|
22
|
Not just amyloid: physiological functions of the amyloid precursor protein family. Nat Rev Neurosci 2017; 18:281-298. [PMID: 28360418 DOI: 10.1038/nrn.2017.29] [Citation(s) in RCA: 411] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyloid precursor protein (APP) gives rise to the amyloid-β peptide and thus has a key role in the pathogenesis of Alzheimer disease. By contrast, the physiological functions of APP and the closely related APP-like proteins (APLPs) remain less well understood. Studying these physiological functions has been challenging and has required a careful long-term strategy, including the analysis of different App-knockout and Aplp-knockout mice. In this Review, we summarize these findings, focusing on the in vivo roles of APP family members and their processing products for CNS development, synapse formation and function, brain injury and neuroprotection, as well as ageing. In addition, we discuss the implications of APP physiology for therapeutic approaches.
Collapse
|
23
|
Copenhaver PF, Ramaker JM. Neuronal migration during development and the amyloid precursor protein. CURRENT OPINION IN INSECT SCIENCE 2016; 18:1-10. [PMID: 27939704 PMCID: PMC5157842 DOI: 10.1016/j.cois.2016.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/06/2016] [Indexed: 06/06/2023]
Abstract
The Amyloid Precursor Protein (APP) is the source of amyloid peptides that accumulate in Alzheimer's disease. However, members of the APP family are strongly expressed in the developing nervous systems of invertebrates and vertebrates, where they regulate neuronal guidance, synaptic remodeling, and injury responses. In contrast to mammals, insects express only one APP ortholog (APPL), simplifying investigations into its normal functions. Recent studies have shown that APPL regulates neuronal migration in the developing insect nervous system, analogous to the roles ascribed to APP family proteins in the mammalian cortex. The comparative simplicity of insect systems offers new opportunities for deciphering the signaling mechanisms by which this enigmatic class of proteins contributes to the formation and function of the nervous system.
Collapse
Affiliation(s)
- Philip F Copenhaver
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA.
| | - Jenna M Ramaker
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, OR 97239, USA; Department of Pathology, Oregon Health & Science University, Portland, OR 97239, USA
| |
Collapse
|
24
|
Ramaker JM, Cargill RS, Swanson TL, Quirindongo H, Cassar M, Kretzschmar D, Copenhaver PF. Amyloid Precursor Proteins Are Dynamically Trafficked and Processed during Neuronal Development. Front Mol Neurosci 2016; 9:130. [PMID: 27932950 PMCID: PMC5122739 DOI: 10.3389/fnmol.2016.00130] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/10/2016] [Indexed: 01/10/2023] Open
Abstract
Proteolytic processing of the Amyloid Precursor Protein (APP) produces beta-amyloid (Aβ) peptide fragments that accumulate in Alzheimer's Disease (AD), but APP may also regulate multiple aspects of neuronal development, albeit via mechanisms that are not well understood. APP is a member of a family of transmembrane glycoproteins expressed by all higher organisms, including two mammalian orthologs (APLP1 and APLP2) that have complicated investigations into the specific activities of APP. By comparison, insects express only a single APP-related protein (APP-Like, or APPL) that contains the same protein interaction domains identified in APP. However, unlike its mammalian orthologs, APPL is only expressed by neurons, greatly simplifying an analysis of its functions in vivo. Like APP, APPL is processed by secretases to generate a similar array of extracellular and intracellular cleavage fragments, as well as an Aβ-like fragment that can induce neurotoxic responses in the brain. Exploiting the complementary advantages of two insect models (Drosophila melanogaster and Manduca sexta), we have investigated the regulation of APPL trafficking and processing with respect to different aspects of neuronal development. By comparing the behavior of endogenously expressed APPL with fluorescently tagged versions of APPL and APP, we have shown that some full-length protein is consistently trafficked into the most motile regions of developing neurons both in vitro and in vivo. Concurrently, much of the holoprotein is rapidly processed into N- and C-terminal fragments that undergo bi-directional transport within distinct vesicle populations. Unexpectedly, we also discovered that APPL can be transiently sequestered into an amphisome-like compartment in developing neurons, while manipulations targeting APPL cleavage altered their motile behavior in cultured embryos. These data suggest that multiple mechanisms restrict the bioavailability of the holoprotein to regulate APPL-dependent responses within the nervous system. Lastly, targeted expression of our double-tagged constructs (combined with time-lapse imaging) revealed that APP family proteins are subject to complex patterns of trafficking and processing that vary dramatically between different neuronal subtypes. In combination, our results provide a new perspective on how the regulation of APP family proteins can be modulated to accommodate a variety of cell type-specific responses within the embryonic and adult nervous system.
Collapse
Affiliation(s)
- Jenna M Ramaker
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science UniversityPortland, OR, USA; Neuroscience Graduate Program, Oregon Health and Science UniversityPortland, OR, USA
| | - Robert S Cargill
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University Portland, OR, USA
| | - Tracy L Swanson
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University Portland, OR, USA
| | - Hanil Quirindongo
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University Portland, OR, USA
| | - Marlène Cassar
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University Portland, OR, USA
| | - Doris Kretzschmar
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University Portland, OR, USA
| | - Philip F Copenhaver
- Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University Portland, OR, USA
| |
Collapse
|
25
|
Sosa LJ, Malter JS, Hu J, Bustos Plonka F, Oksdath M, Nieto Guil AF, Quiroga S, Pfenninger KH. Protein interacting with NIMA (never in mitosis A)-1 regulates axonal growth cone adhesion and spreading through myristoylated alanine-rich C kinase substrate isomerization. J Neurochem 2016; 137:744-55. [DOI: 10.1111/jnc.13612] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/20/2016] [Accepted: 03/03/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Lucas J. Sosa
- Department of Pediatrics and Colorado Intellectual and Developmental Disabilities Research Center; University of Colorado School of Medicine; Aurora Colorado USA
| | - James S. Malter
- Department of Pathology; University of Texas Southwestern Medical Center; Dallas Texas USA
| | - Jie Hu
- Department of Pathology; University of Texas Southwestern Medical Center; Dallas Texas USA
| | - Florentyna Bustos Plonka
- Departamento de Química Biológica-CIQUIBIC; Facultad de Ciencias Químicas; Universidad Nacional de Córdoba-CONICET; Córdoba Argentina
| | - Mariana Oksdath
- Departamento de Química Biológica-CIQUIBIC; Facultad de Ciencias Químicas; Universidad Nacional de Córdoba-CONICET; Córdoba Argentina
| | - Alvaro F. Nieto Guil
- Departamento de Química Biológica-CIQUIBIC; Facultad de Ciencias Químicas; Universidad Nacional de Córdoba-CONICET; Córdoba Argentina
| | - Santiago Quiroga
- Departamento de Química Biológica-CIQUIBIC; Facultad de Ciencias Químicas; Universidad Nacional de Córdoba-CONICET; Córdoba Argentina
| | - Karl H. Pfenninger
- Department of Pediatrics and Colorado Intellectual and Developmental Disabilities Research Center; University of Colorado School of Medicine; Aurora Colorado USA
| |
Collapse
|
26
|
Yu Y, Li Y, Zhang Y. Yeast Two-Hybrid Screening for Proteins that Interact with the Extracellular Domain of Amyloid Precursor Protein. Neurosci Bull 2016; 32:171-6. [PMID: 26960425 DOI: 10.1007/s12264-016-0021-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/11/2015] [Indexed: 12/25/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder in which amyloid β plaques are a pathological characteristic. Little is known about the physiological functions of amyloid β precursor protein (APP). Based on its structure as a type I transmembrane protein, it has been proposed that APP might be a receptor, but so far, no ligand has been reported. In the present study, 9 proteins binding to the extracellular domain of APP were identified using a yeast two-hybrid system. After confirming the interactions in the mammalian system, mutated PLP1, members of the FLRT protein family, and KCTD16 were shown to interact with APP. These proteins have been reported to be involved in Pelizaeus-Merzbacher disease (PMD) and axon guidance. Therefore, our results shed light on the mechanisms of physiological function of APP in AD, PMD, and axon guidance.
Collapse
Affiliation(s)
- You Yu
- State Key Laboratory of Membrane Biology, College of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Yinan Li
- State Key Laboratory of Membrane Biology, College of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, College of Life Sciences, PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China.
| |
Collapse
|
27
|
APP Receptor? To Be or Not To Be. Trends Pharmacol Sci 2016; 37:390-411. [PMID: 26837733 DOI: 10.1016/j.tips.2016.01.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 11/22/2022]
Abstract
Amyloid precursor protein (APP) and its metabolites play a key role in Alzheimer's disease pathogenesis. The idea that APP may function as a receptor has gained momentum based on its structural similarities to type I transmembrane receptors and the identification of putative APP ligands. We review the recent experimental evidence in support of this notion and discuss how this concept is viewed in the field. Specifically, we focus on the structural and functional characteristics of APP as a cell surface receptor, and on its interaction with adaptors and signaling proteins. We also address the importance of APP function as a receptor in Alzheimer's disease etiology and discuss how this function might be potentially important for the development of novel therapeutic approaches.
Collapse
|
28
|
Sadallah M, Labat-Gest V, Tempia F. Propagation of Neuronal Damage to Embryonic Grafts Transplanted in the Hippocampus of Murine Models of Alzheimer's Disease. Rejuvenation Res 2015; 18:554-63. [PMID: 26540615 DOI: 10.1089/rej.2015.1672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by the presence of two principal hallmarks-amyloid plaques and neurofibrillary tangles. The primary cause of the majority of AD cases is not known. Likewise, the mechanisms underlying the propagation of the pathology from affected tissue to neighboring healthy neurons are largely unknown, but knowledge about them could be helpful to design strategies aimed at halting the progression of the disease. To throw light on the mechanisms of propagation of neuronal damage to healthy tissue, wild-type (WT) hippocampal solid tissue chunks derived from green fluorescent protein (GFP)-positive embryos were grafted into the hippocampus of 6-month-old WT and 3xTg-AD mice, a triple-transgenic mouse model that exhibits both amyloid-beta (Aβ) and tau protein pathology. The histological and morphological alterations of the grafted tissues were assessed 3 months post-transplantation. Tissues grafted in 3xTg-AD hosts, compared to those grafted in WT recipients, presented a significant decrease in neurite outgrowth (35.4%) and dendritic spine density (41.3%), mainly due to a reduction of stubby and thin-shaped spines. Moreover, some cells of the tissue transplanted in 3xTg-AD hosts accumulated intracellular amyloid peptide deposits similar to the cells of the host. Furthermore, the immunohistochemical examination of reactive astrocytes and microglia revealed the presence of more inflammation in the grafted tissues hosted in 3xTg-AD compared to WT recipients. These results show a propagation of neuronal damage to initially healthy embryonic grafts, validating this methodology for future studies on the mechanisms of the progression of AD pathology to surrounding regions.
Collapse
Affiliation(s)
- Mohcene Sadallah
- 1 Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino , Orbassano (Torino), Italy .,2 Department of Biology, Ecole Normale Supérieure de Kouba , Algiers, Algeria
| | - Vivien Labat-Gest
- 1 Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino , Orbassano (Torino), Italy
| | - Filippo Tempia
- 1 Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino , Orbassano (Torino), Italy .,3 Department of Neuroscience and National Institute of Neuroscience-Italy (INN), University of Torino , Torino, Italy
| |
Collapse
|
29
|
Kipkorir T, Colangelo CM, Manuelidis L. Proteomic analysis of host brain components that bind to infectious particles in Creutzfeldt-Jakob disease. Proteomics 2015; 15:2983-98. [PMID: 25930988 DOI: 10.1002/pmic.201500059] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 03/26/2015] [Accepted: 04/29/2015] [Indexed: 11/07/2022]
Abstract
Transmissible encephalopathies (TSEs), such as Creutzfeldt-Jakob disease (CJD) and scrapie, are caused by infectious agents that provoke strain-specific patterns of disease. Misfolded host prion protein (PrP-res amyloid) is believed to be the causal infectious agent. However, particles that are stripped of PrP retain both high infectivity and viral proteins not detectable in uninfected mouse controls. We here detail host proteins bound with FU-CJD agent infectious brain particles by proteomic analysis. More than 98 proteins were differentially regulated, and 56 FU-CJD exclusive proteins were revealed after PrP, GFAP, C1q, ApoE, and other late pathologic response proteins were removed. Stripped FU-CJD particles revealed HSC70 (144× the uninfected control), cyclophilin B, an FU-CJD exclusive protein required by many viruses, and early endosome-membrane pathways known to facilitate viral processing, replication, and spread. Synaptosomal elements including synapsin-2 (at 33×) and AP180 (a major FU-CJD exclusive protein) paralleled the known ultrastructural location of 25 nm virus-like TSE particles and infectivity in synapses. Proteins without apparent viral or neurodegenerative links (copine-3), and others involved in viral-induced protein misfolding and aggregation, were also identified. Human sCJD brain particles contained 146 exclusive proteins, and heat shock, synaptic, and viral pathways were again prominent, in addition to Alzheimer, Parkinson, and Huntington aggregation proteins. Host proteins that bind TSE infectious particles can prevent host immune recognition and contribute to prolonged cross-species transmissions (the species barrier). Our infectious particle strategy, which reduces background sequences by >99%, emphasizes host targets for new therapeutic initiatives. Such therapies can simultaneously subvert common pathways of neurodegeneration.
Collapse
|
30
|
Lim S, Yoo BK, Kim HS, Gilmore HL, Lee Y, Lee HP, Kim SJ, Letterio J, Lee HG. Amyloid-β precursor protein promotes cell proliferation and motility of advanced breast cancer. BMC Cancer 2014; 14:928. [PMID: 25491510 PMCID: PMC4295427 DOI: 10.1186/1471-2407-14-928] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 12/05/2014] [Indexed: 12/21/2022] Open
Abstract
Background Amyloid-β precursor protein (APP) is a highly conserved single transmembrane protein that has been linked to Alzheimer disease. Recently, the increased expression of APP in multiple types of cancers has been reported where it has significant correlation with the cancer cell proliferation. However, the function of APP in the pathogenesis of breast cancer has not previously been determined. In this study, we studied the pathological role of APP in breast cancer and revealed its potential mechanism. Methods The expression level of APP in multiple breast cancer cell lines was measured by Western blot analysis and the breast cancer tissue microarray was utilized to analyze the expression pattern of APP in human patient specimens. To interrogate the functional role of APP in cell growth and apoptosis, the effect of APP knockdown in MDA-MB-231 cells were analyzed. Specifically, multiple signal transduction pathways and functional alterations linked to cell survival and motility were examined in in vivo animal model as well as in vitro cell culture with the manipulation of APP expression. Results We found that the expression of APP is increased in mouse and human breast cancer cell lines, especially in the cell line possessing higher metastatic potential. Moreover, the analysis of human breast cancer tissues revealed a significant correlation between the level of APP and tumor development. Knockdown of APP (APP-kd) in breast cancer cells caused the retardation of cell growth in vitro and in vivo, with both the induction of p27kip1 and caspase-3-mediated apoptosis. APP-kd cells also had higher sensitivity to treatment of chemotherapeutic agents, TRAIL and 5-FU. Such anti-tumorigenic effects shown in the APP-kd cells partially came from reduced pro-survival AKT activation in response to IGF-1, leading to activation of key signaling regulators for cell growth, survival, and pro-apoptotic events such as GSK3-β and FOXO1. Notably, knock-down of APP in metastatic breast cancer cells limited cell migration and invasion ability upon stimulation of IGF-1. Conclusion The present data strongly suggest that the increase of APP expression is causally linked to tumorigenicity as well as invasion of aggressive breast cancer and, therefore, the targeting of APP may be an effective therapy for breast cancer.
Collapse
Affiliation(s)
- Seunghwan Lim
- Department of Pediatrics, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, 2103 Cornell Road, Cleveland, OH 44106, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Plácido A, Pereira C, Duarte A, Candeias E, Correia S, Santos R, Carvalho C, Cardoso S, Oliveira C, Moreira P. The role of endoplasmic reticulum in amyloid precursor protein processing and trafficking: Implications for Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1444-53. [DOI: 10.1016/j.bbadis.2014.05.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 04/29/2014] [Accepted: 05/06/2014] [Indexed: 12/21/2022]
|
32
|
Zaręba-Kozioł M, Szwajda A, Dadlez M, Wysłouch-Cieszyńska A, Lalowski M. Global analysis of S-nitrosylation sites in the wild type (APP) transgenic mouse brain-clues for synaptic pathology. Mol Cell Proteomics 2014; 13:2288-305. [PMID: 24895380 DOI: 10.1074/mcp.m113.036079] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Alzheimer's disease (AD) is characterized by an early synaptic loss, which strongly correlates with the severity of dementia. The pathogenesis and causes of characteristic AD symptoms are not fully understood. Defects in various cellular cascades were suggested, including the imbalance in production of reactive oxygen and nitrogen species. Alterations in S-nitrosylation of several proteins were previously demonstrated in various AD animal models and patients. In this work, using combined biotin-switch affinity/nano-LC-MS/MS and bioinformatic approaches we profiled endogenous S-nitrosylation of brain synaptosomal proteins from wild type and transgenic mice overexpressing mutated human Amyloid Precursor Protein (hAPP). Our data suggest involvement of S-nitrosylation in the regulation of 138 synaptic proteins, including MAGUK, CamkII, or synaptotagmins. Thirty-eight proteins were differentially S-nitrosylated in hAPP mice only. Ninety-five S-nitrosylated peptides were identified for the first time (40% of total, including 33 peptides exclusively in hAPP synaptosomes). We verified differential S-nitrosylation of 10 (26% of all identified) synaptosomal proteins from hAPP mice, by Western blotting with specific antibodies. Functional enrichment analysis linked S-nitrosylated proteins to various cellular pathways, including: glycolysis, gluconeogenesis, calcium homeostasis, ion, and vesicle transport, suggesting a basic role of this post-translational modification in the regulation of synapses. The linkage of SNO-proteins to axonal guidance and other processes related to APP metabolism exclusively in the hAPP brain, implicates S-nitrosylation in the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Monika Zaręba-Kozioł
- From the ‡Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | - Michał Dadlez
- From the ‡Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | - Maciej Lalowski
- ¶Biomedicum Helsinki, Institute of Biomedicine, Biochemistry/Developmental Biology, Meilahti Clinical Proteomics Core Unit, University of Helsinki, Finland; ‖Folkhälsan Institute of Genetics, Helsinki, Finland
| |
Collapse
|
33
|
Soldano A, Hassan BA. Beyond pathology: APP, brain development and Alzheimer's disease. Curr Opin Neurobiol 2014; 27:61-7. [PMID: 24632309 DOI: 10.1016/j.conb.2014.02.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 01/15/2014] [Accepted: 02/06/2014] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia among the elderly. Research in the AD field has been mostly focused on the biology of the Aβ peptide but increasing evidence is shifting attention toward the physiological role of APP as key to understanding AD pathology. It is becoming apparent that APP plays a central role in the mechanisms that guarantee the accuracy and the robustness of brain wiring. In the present review we explore APP functions with focus on some of the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Alessia Soldano
- VIB Center for the Biology of Disease, VIB, 3000 Leuven, Belgium; Center for Human Genetics, University of Leuven School of Medicine, 3000 Leuven, Belgium
| | - Bassem A Hassan
- VIB Center for the Biology of Disease, VIB, 3000 Leuven, Belgium; Center for Human Genetics, University of Leuven School of Medicine, 3000 Leuven, Belgium.
| |
Collapse
|
34
|
Sosa LJ, Postma NL, Estrada-Bernal A, Hanna M, Guo R, Busciglio J, Pfenninger KH. Dosage of amyloid precursor protein affects axonal contact guidance in Down syndrome. FASEB J 2013; 28:195-205. [PMID: 24036883 DOI: 10.1096/fj.13-232686] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Amyloid precursor protein (APP), encoded on Hsa21, functions as a cell adhesion molecule (CAM) in axonal growth cones (GCs) of the developing brain. We show here that axonal GCs of human fetal Down syndrome (DS) neurons (and of a DS mouse model) overexpress APP protein relative to euploid controls. We investigated whether DS neurons generate an abnormal, APP-dependent GC phenotype in vitro. On laminin, which binds APP and β1 integrins (Itgb1), DS neurons formed enlarged and faster-advancing GCs compared to controls. On peptide matrices that bind APP only, but not on those binding exclusively Itgb1 or L1CAM, DS GCs were significantly enlarged (2.0-fold), formed increased close adhesions (1.8-fold), and advanced faster (1.4-fold). In assays involving alternating stripes of monospecific matrices, human control GCs exhibited no preference for any of the substrates, whereas DS GCs preferred the APP-binding matrix (cross-over decreased significantly from 48.2 to 27.2%). Reducing APP expression in DS GCs with siRNA normalized most measures of the phenotype, including substrate choice. These experiments show that human DS neurons exhibit an APP-dependent, abnormal GC phenotype characterized by increased adhesion and altered contact guidance. The results suggest that APP overexpression may perturb axonal pathfinding and circuit formation in developing DS brain.
Collapse
Affiliation(s)
- Lucas J Sosa
- 3Department of Pediatrics, University of Colorado, Mailbox 8313, 12800 E. 19th Ave, Aurora, CO 80045, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Busciglio J, Capone G, O'Byran J, Gardiner K. Down Syndrome: Genes, Model Systems, and Progress towards Pharmacotherapies and Clinical Trials for Cognitive Deficits. Cytogenet Genome Res 2013; 141:260-71. [DOI: 10.1159/000354306] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2013] [Indexed: 11/19/2022] Open
|