1
|
Zimmerman AJ, Serrano-Rodriguez A, Wilson SJ, Linsenbardt DN, Brigman JL, Weick J. Knockout of AMPA receptor binding protein Neuron-Specific Gene 2 (NSG2) enhances associative learning and cognitive flexibility. RESEARCH SQUARE 2024:rs.3.rs-4790348. [PMID: 39257983 PMCID: PMC11384823 DOI: 10.21203/rs.3.rs-4790348/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
The vast majority of gene mutations and/or gene knockouts result in either no observable changes, or significant deficits in molecular, cellular, or organismal function. However, in a small number of cases, mutant animal models display enhancements in specific behaviors such as learning and memory. To date, most gene deletions shown to enhance cognitive ability generally affect a limited number of pathways such as NMDA receptor- and translation-dependent plasticity, or GABA receptor- and potassium channel-mediated inhibition. While endolysosomal trafficking of AMPA receptors is a critical mediator of synaptic plasticity, mutations in genes that affect AMPAR trafficking either have no effect or are deleterious for synaptic plasticity, learning and memory. NSG2 is one of the three-member family of Neuron-specific genes (NSG1-3), which have been shown to regulate endolysosomal trafficking of a number of proteins critical for neuronal function, including AMPAR subunits (GluA1-2). Based on these findings and the largely universal expression throughout mammalian brain, we predicted that genetic knockout of NSG2 would result in significant impairments across multiple behavioral modalities including motor, affective, and learning/memory paradigms. However, in the current study we show that loss of NSG2 had highly selective effects on associative learning and memory, leaving motor and affective behaviors intact. For instance, NSG2 KO animals performed equivalent to wild-type C57Bl/6n mice on rotarod and Catwalk motor tasks, and did not display alterations in anxiety-like behavior on open field and elevated zero maze tasks. However, NSG2 KO animals demonstrated enhanced recall in the Morris water maze, accelerated reversal learning in a touch-screen task, and accelerated acquisition and enhanced recall on a Trace Fear Conditioning task. Together, these data point to a specific involvement of NSG2 on multiple types of associative learning, and expand the repertoire of pathways that can be targeted for cognitive enhancement.
Collapse
|
2
|
Trovò L, Kouvaros S, Schwenk J, Fernandez-Fernandez D, Fritzius T, Rem PD, Früh S, Gassmann M, Fakler B, Bischofberger J, Bettler B. Synaptotagmin-11 facilitates assembly of a presynaptic signaling complex in post-Golgi cargo vesicles. EMBO Rep 2024; 25:2610-2634. [PMID: 38698221 PMCID: PMC11169412 DOI: 10.1038/s44319-024-00147-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 05/05/2024] Open
Abstract
GABAB receptors (GBRs), the G protein-coupled receptors for GABA, regulate synaptic transmission throughout the brain. A main synaptic function of GBRs is the gating of Cav2.2-type Ca2+ channels. However, the cellular compartment where stable GBR/Cav2.2 signaling complexes form remains unknown. In this study, we demonstrate that the vesicular protein synaptotagmin-11 (Syt11) binds to both the auxiliary GBR subunit KCTD16 and Cav2.2 channels. Through these dual interactions, Syt11 recruits GBRs and Cav2.2 channels to post-Golgi vesicles, thus facilitating assembly of GBR/Cav2.2 signaling complexes. In addition, Syt11 stabilizes GBRs and Cav2.2 channels at the neuronal plasma membrane by inhibiting constitutive internalization. Neurons of Syt11 knockout mice exhibit deficits in presynaptic GBRs and Cav2.2 channels, reduced neurotransmitter release, and decreased GBR-mediated presynaptic inhibition, highlighting the critical role of Syt11 in the assembly and stable expression of GBR/Cav2.2 complexes. These findings support that Syt11 acts as a vesicular scaffold protein, aiding in the assembly of signaling complexes from low-abundance components within transport vesicles. This mechanism enables insertion of pre-assembled functional signaling units into the synaptic membrane.
Collapse
Affiliation(s)
- Luca Trovò
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Jochen Schwenk
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | | | | | - Simon Früh
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Martin Gassmann
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Bernd Fakler
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- CIBSS Center for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation, Freiburg, Germany
| | | | - Bernhard Bettler
- Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
3
|
Overby M, Serrano-Rodriguez A, Dadras S, Christiansen AK, Ozcelik G, Lichtenthaler SF, Weick JP, Müller HK. Neuron-specific gene NSG1 binds to and positively regulates sortilin ectodomain shedding via a metalloproteinase-dependent mechanism. J Biol Chem 2023; 299:105446. [PMID: 37949230 PMCID: PMC10704435 DOI: 10.1016/j.jbc.2023.105446] [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: 05/30/2023] [Revised: 10/15/2023] [Accepted: 11/03/2023] [Indexed: 11/12/2023] Open
Abstract
Increasing evidence suggests that aberrant regulation of sortilin ectodomain shedding can contribute to amyloid-β pathology and frontotemporal dementia, although the mechanism by which this occurs has not been elucidated. Here, we probed for novel binding partners of sortilin using multiple and complementary approaches and identified two proteins of the neuron-specific gene (NSG) family, NSG1 and NSG2, that physically interact and colocalize with sortilin. We show both NSG1 and NSG2 induce subcellular redistribution of sortilin to NSG1- and NSG2-enriched compartments. However, using cell surface biotinylation, we found only NSG1 reduced sortilin cell surface expression, which caused significant reductions in uptake of progranulin, a molecular determinant for frontotemporal dementia. In contrast, we demonstrate NSG2 has no effect on sortilin cell surface abundance or progranulin uptake, suggesting specificity for NSG1 in the regulation of sortilin cell surface expression. Using metalloproteinase inhibitors and A disintegrin and metalloproteinase 10 KO cells, we further show that NSG1-dependent reduction of cell surface sortilin occurred via proteolytic processing by A disintegrin and metalloproteinase 10 with a concomitant increase in shedding of sortilin ectodomain to the extracellular space. This represents a novel regulatory mechanism for sortilin ectodomain shedding that is regulated in a neuron-specific manner. Furthermore, this finding has implications for the development of strategies for brain-specific regulation of sortilin and possibly sortilin-driven pathologies.
Collapse
Affiliation(s)
- Malene Overby
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Antonio Serrano-Rodriguez
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Somayeh Dadras
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Ann Kathrine Christiansen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Gözde Ozcelik
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum Rechts der lsar, Technical University of Munich, Munich, Germany
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Neuroproteomics, School of Medicine, Klinikum Rechts der lsar, Technical University of Munich, Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Jason Porter Weick
- Department of Neurosciences, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Heidi Kaastrup Müller
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
4
|
Hampel H, Caruso G, Nisticò R, Piccioni G, Mercuri NB, Giorgi FS, Ferrarelli F, Lemercier P, Caraci F, Lista S, Vergallo A. Biological Mechanism-based Neurology and Psychiatry: A BACE1/2 and Downstream Pathway Model. Curr Neuropharmacol 2023; 21:31-53. [PMID: 34852743 PMCID: PMC10193755 DOI: 10.2174/1570159x19666211201095701] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 02/04/2023] Open
Abstract
In oncology, comprehensive omics and functional enrichment studies have led to an extensive profiling of (epi)genetic and neurobiological alterations that can be mapped onto a single tumor's clinical phenotype and divergent clinical phenotypes expressing common pathophysiological pathways. Consequently, molecular pathway-based therapeutic interventions for different cancer typologies, namely tumor type- and site-agnostic treatments, have been developed, encouraging the real-world implementation of a paradigm shift in medicine. Given the breakthrough nature of the new-generation translational research and drug development in oncology, there is an increasing rationale to transfertilize this blueprint to other medical fields, including psychiatry and neurology. In order to illustrate the emerging paradigm shift in neuroscience, we provide a state-of-the-art review of translational studies on the β-site amyloid precursor protein cleaving enzyme (BACE) and its most studied downstream effector, neuregulin, which are molecular orchestrators of distinct biological pathways involved in several neurological and psychiatric diseases. This body of data aligns with the evidence of a shared genetic/biological architecture among Alzheimer's disease, schizoaffective disorder, and autism spectrum disorders. To facilitate a forward-looking discussion about a potential first step towards the adoption of biological pathway-based, clinical symptom-agnostic, categorization models in clinical neurology and psychiatry for precision medicine solutions, we engage in a speculative intellectual exercise gravitating around BACE-related science, which is used as a paradigmatic case here. We draw a perspective whereby pathway-based therapeutic strategies could be catalyzed by highthroughput techniques embedded in systems-scaled biology, neuroscience, and pharmacology approaches that will help overcome the constraints of traditional descriptive clinical symptom and syndrome-focused constructs in neurology and psychiatry.
Collapse
Affiliation(s)
- Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | | | - Robert Nisticò
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
- School of Pharmacy, University of Rome “Tor Vergata”, Rome, Italy
| | - Gaia Piccioni
- Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
- Department of Physiology and Pharmacology “V.Erspamer”, Sapienza University of Rome, Rome, Italy
| | - Nicola B. Mercuri
- Department of Systems Medicine, University of Rome “Tor Vergata”, Rome, Italy
- IRCCS Santa Lucia Foundation, Rome, Italy
| | - Filippo Sean Giorgi
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Fabio Ferrarelli
- Department of Psychiatry, University of Pittsburgh, School of Medicine, Pittsburgh, PA, USA
| | - Pablo Lemercier
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | - Filippo Caraci
- Oasi Research Institute-IRCCS, Troina, Italy
- Department of Drug Sciences, University of Catania, Catania, Italy
| | - Simone Lista
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
- Memory Resources and Research Center (CMRR), Neurology Department, Gui de Chauliac University Hospital, Montpellier, France
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| |
Collapse
|
5
|
Qi L, Sun C, Sun S, Li A, Hu Q, Liu Y, Zhang Y. Phosphatidylinositol (3,5)-bisphosphate machinery regulates neurite thickness through neuron-specific endosomal protein NSG1/NEEP21. J Biol Chem 2022; 299:102775. [PMID: 36493904 PMCID: PMC9823133 DOI: 10.1016/j.jbc.2022.102775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 10/31/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Phosphatidylinositol (3,5)-bisphosphate [PtdIns(3,5)P2] is a critical signaling phospholipid involved in endolysosome homeostasis. It is synthesized by a protein complex composed of PIKfyve, Vac14, and Fig4. Defects in PtdIns(3,5)P2 synthesis underlie a number of human neurological disorders, including Charcot-Marie-Tooth disease, child onset progressive dystonia, and others. However, neuron-specific functions of PtdIns(3,5)P2 remain less understood. Here, we show that PtdIns(3,5)P2 pathway is required to maintain neurite thickness. Suppression of PIKfyve activities using either pharmacological inhibitors or RNA silencing resulted in decreased neurite thickness. We further find that the regulation of neurite thickness by PtdIns(3,5)P2 is mediated by NSG1/NEEP21, a neuron-specific endosomal protein. Knockdown of NSG1 expression also led to thinner neurites. mCherry-tagged NSG1 colocalized and interacted with proteins in the PtdIns(3,5)P2 machinery. Perturbation of PtdIns(3,5)P2 dynamics by overexpressing Fig4 or a PtdIns(3,5)P2-binding domain resulted in mislocalization of NSG1 to nonendosomal locations, and suppressing PtdIns(3,5)P2 synthesis resulted in an accumulation of NSG1 in EEA1-positive early endosomes. Importantly, overexpression of NSG1 rescued neurite thinning in PtdIns(3,5)P2-deficient CAD neurons and primary cortical neurons. Our study uncovered the role of PtdIns(3,5)P2 in the morphogenesis of neurons, which revealed a novel aspect of the pathogenesis of PtdIns(3,5)P2-related neuropathies. We also identified NSG1 as an important downstream protein of PtdIns(3,5)P2, which may provide a novel therapeutic target in neurological diseases.
Collapse
Affiliation(s)
- Lijuan Qi
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, China,National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chen Sun
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
| | - Shenqing Sun
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Aiqing Li
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Qiuming Hu
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, China
| | - Yaobo Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Institute of Neuroscience, Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, Jiangsu, China
| | - Yanling Zhang
- Department of Biochemistry and Molecular Biology, Soochow University Medical College, Suzhou, Jiangsu, China,For correspondence: Yanling Zhang
| |
Collapse
|
6
|
Yesiltepe M, Yin T, Tambini MD, Breuillaud L, Zehntner SP, D’Adamio L. Late-long-term potentiation magnitude, but not Aβ levels and amyloid pathology, is associated with behavioral performance in a rat knock-in model of Alzheimer disease. Front Aging Neurosci 2022; 14:1040576. [PMID: 36438008 PMCID: PMC9691854 DOI: 10.3389/fnagi.2022.1040576] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/31/2022] [Indexed: 09/23/2023] Open
Abstract
Cleavage of Amyloid precursor protein by β- and γ-secretases lead to Aβ formation. The widely accepted pathogenic model states that these mutations cause AD via an increase in Aβ formation and accumulation of Aβ in Amyloid plaques. APP mutations cause early onset familial forms of Alzheimer's disease (FAD) in humans. We generated App-Swedish (Apps ) knock-in rats, which carry a pathogenic APP mutation in the endogenous rat App gene. This mutation increases β-secretase processing of APP leading to both augmented Aβ production and facilitation of glutamate release in Apps/s rats, via a β-secretase and APP-dependent glutamate release mechanism. Here, we studied 11 to 14-month-old male and female Apps/s rats. To determine whether the Swedish App mutation leads to behavioral deficits, Apps/s knock-in rats were subjected to behavioral analysis using the IntelliCage platform, an automated behavioral testing system. This system allows behavioral assessment in socially housed animals reflecting a more natural, less stress-inducing environment and eliminates experimenter error and bias while increasing precision of measurements. Surprisingly, a spatial discrimination and flexibility task that can reveal deficits in higher order brain function showed that Apps/s females, but not Apps/s male rats, performed significantly worse than same sex controls. Moreover, female control rats performed significantly better than control and Apps/s male rats. The Swedish mutation causes a significant increase in Aβ production in 14-month-old animals of both sexes. Yet, male and female Apps/s rats showed no evidence of AD-related amyloid pathology. Finally, Apps/s rats did not show signs of significant neuroinflammation. Given that the APP Swedish mutation causes alterations in glutamate release, we analyzed Long-term potentiation (LTP), a long-lasting form of synaptic plasticity that is a cellular basis for learning and memory. Strikingly, LTP was significantly increased in Apps/s control females compared to both Apps/s sexes and control males. In conclusion, this study shows that behavioral performances are sex and App-genotype dependent. In addition, they are associated with LTP values and not Aβ or AD-related pathology. These data, and the failures of anti-Aβ therapies in humans, suggest that alternative pathways, such as those leading to LTP dysfunction, should be targeted for disease-modifying AD therapy.
Collapse
Affiliation(s)
- Metin Yesiltepe
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer’s Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Tao Yin
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer’s Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Marc D. Tambini
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer’s Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | | | | | - Luciano D’Adamio
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer’s Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
7
|
Pfundstein G, Nikonenko AG, Sytnyk V. Amyloid precursor protein (APP) and amyloid β (Aβ) interact with cell adhesion molecules: Implications in Alzheimer’s disease and normal physiology. Front Cell Dev Biol 2022; 10:969547. [PMID: 35959488 PMCID: PMC9360506 DOI: 10.3389/fcell.2022.969547] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/07/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is an incurable neurodegenerative disorder in which dysfunction and loss of synapses and neurons lead to cognitive impairment and death. Accumulation and aggregation of neurotoxic amyloid-β (Aβ) peptides generated via amyloidogenic processing of amyloid precursor protein (APP) is considered to play a central role in the disease etiology. APP interacts with cell adhesion molecules, which influence the normal physiological functions of APP, its amyloidogenic and non-amyloidogenic processing, and formation of Aβ aggregates. These cell surface glycoproteins also mediate attachment of Aβ to the neuronal cell surface and induce intracellular signaling contributing to Aβ toxicity. In this review, we discuss the current knowledge surrounding the interactions of cell adhesion molecules with APP and Aβ and analyze the evidence of the critical role these proteins play in regulating the processing and physiological function of APP as well as Aβ toxicity. This is a necessary piece of the complex AD puzzle, which we should understand in order to develop safe and effective therapeutic interventions for AD.
Collapse
Affiliation(s)
- Grant Pfundstein
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
| | | | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, Australia
- *Correspondence: Vladimir Sytnyk,
| |
Collapse
|
8
|
Yap CC, Winckler B. Spatial regulation of endosomes in growing dendrites. Dev Biol 2022; 486:5-14. [PMID: 35306006 PMCID: PMC10646839 DOI: 10.1016/j.ydbio.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 02/21/2022] [Accepted: 03/13/2022] [Indexed: 01/19/2023]
Abstract
Many membrane proteins are highly enriched in either dendrites or axons. This non-uniform distribution is a critical feature of neuronal polarity and underlies neuronal function. The molecular mechanisms responsible for polarized distribution of membrane proteins has been studied for some time and many answers have emerged. A less well studied feature of neurons is that organelles are also frequently non-uniformly distributed. For instance, EEA1-positive early endosomes are somatodendritic whereas synaptic vesicles are axonal. In addition, some organelles are present in both axons and dendrites, but not distributed uniformly along the processes. One well known example are lysosomes which are abundant in the soma and proximal dendrite, but sparse in the distal dendrite and the distal axon. The mechanisms that determine the spatial distribution of organelles along dendrites are only starting to be studied. In this review, we will discuss the cell biological mechanisms of how the distribution of diverse sets of endosomes along the proximal-distal axis of dendrites might be regulated. In particular, we will focus on the regulation of bulk homeostatic mechanisms as opposed to local regulation. We posit that immature dendrites regulate organelle motility differently from mature dendrites in order to spatially organize dendrite growth, branching and sculpting.
Collapse
|
9
|
Austin R, Chander P, Zimmerman AJ, Overby M, Digilio L, Yap CC, Linsenbardt DN, Müller HK, Weick JP. Global loss of Neuron-specific gene 1 causes alterations in motor coordination, increased anxiety, and diurnal hyperactivity in male mice. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12816. [PMID: 35577358 PMCID: PMC9262855 DOI: 10.1111/gbb.12816] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/03/2022] [Accepted: 05/04/2022] [Indexed: 11/26/2022]
Abstract
The Neuron-specific gene family (NSG1-3) consists of small endolysosomal proteins that are critical for trafficking multiple receptors and signaling molecules in neurons. NSG1 has been shown to play a critical role in AMPAR recycling from endosomes to plasma membrane during synaptic plasticity. However, to date nothing is known about whether NSG1 is required for normal behavior at an organismal level. Here we performed a battery of behavioral tests to determine whether loss of NSG1 would affect motor, cognitive, and/or affective behaviors, as well as circadian-related activity. Consistent with unique cerebellar expression of NSG1 among family members, we found that NSG1 was obligatory for motor coordination but not for gross motor function or learning. NSG1 knockout (KO) also altered performance across other behavioral modalities including anxiety-related and diurnal activity paradigms. Surprisingly, NSG1 KO did not cause significant impairments across all tasks within a given modality, but had specific effects within each modality. For instance, we found increases in anxiety-related behaviors in tasks with multiple stressors (e.g., elevation and exposure), but not those with a single main stressor (e.g., exposure). Interestingly, NSG1 KO animals displayed a significant increase in locomotor activity during subjective daytime, suggesting a possible impact on diurnal activity rhythms or vigilance. Surprisingly, loss of NSG1 had no effect on hippocampal-dependent learning despite previous studies showing deficits in CA1 long-term potentiation. Together, these findings do not support a role of NSG1 in hippocampal-dependent learning, but support a role in mediating proper neuronal function across amygdalar and cerebellar circuits.
Collapse
Affiliation(s)
- Roman Austin
- Department of NeurosciencesUniversity of New Mexico School of MedicineAlbuquerqueNew MexicoUSA
| | - Praveen Chander
- Department of NeurosciencesUniversity of New Mexico School of MedicineAlbuquerqueNew MexicoUSA
| | - Amber J. Zimmerman
- Department of NeurosciencesUniversity of New Mexico School of MedicineAlbuquerqueNew MexicoUSA
| | - Malene Overby
- Department of Clinical Medicine, Translational Neuropsychiatry UnitAarhus UniversityAarhus CDenmark
| | - Laura Digilio
- Department of Cell BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Chan Choo Yap
- Department of Cell BiologyUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - David N. Linsenbardt
- Department of NeurosciencesUniversity of New Mexico School of MedicineAlbuquerqueNew MexicoUSA
| | - Heidi Kaastrup Müller
- Department of Clinical Medicine, Translational Neuropsychiatry UnitAarhus UniversityAarhus CDenmark
| | - Jason P. Weick
- Department of NeurosciencesUniversity of New Mexico School of MedicineAlbuquerqueNew MexicoUSA
| |
Collapse
|
10
|
Xia X, Li R, Zhou P, Xing Z, Lu C, Long Z, Wang F, Wang R. Decreased NSG3 enhances PD-L1 expression by Erk1/2 pathway to promote pancreatic cancer progress. Am J Cancer Res 2021; 11:916-929. [PMID: 33791163 PMCID: PMC7994162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 11/03/2020] [Indexed: 06/12/2023] Open
Abstract
Inhibiting the functioning of PD-1/PD-L1 to activate human immune system and improve the prognosis of pancreatic cancer (PC) would provide a significant boost to handling the disease. One research found the expression level of NSG3 was reduced in pediatric pilocytic astrocytoma, so is PC and we found NSG3 could regulate the expression of PD-L1. So NSG3 could become a new target for enhancing the immune response to PC. The GEPIA website was employed to analyze the prognoses in PC patients with different NSG3 levels. Immunohistochemistry (IHC) analysis was applied to detect different levels of NSG3 in para-PC and PC tissues. Cell biological function tests (in vitro) were performed and a subcutaneous nude mice tumor model (in vivo) was established to verify the effect of NSG3 on PC. Immunoblotting and RT-qPCR were utilized to demonstrate the inhibiting effect of NSG3 on PD-L1 through regulating Erk1/2 phosphorylation. A subcutaneous C57BL/6 tumor mice model was established to assess the possibility of a synergistic effect of NSG3 expression and the use of an anti-PD-L1 antibody on PC. PC tissues had decreased NSG3 expression levels, which led to poor prognosis. Overexpressing NSG3 suppressed proliferation, invasion and migration capacities of PC cells. On the contrary, knocking-down NSG3 prompted PC malignancy whether in vivo or in vitro. Importantly, NSG3 prevented Erk1/2 phosphorylation to inhibit PD-L1 expression. Additionally, NSG3 and an immune checkpoint inhibitor anti-PD-1 antibody acted synergistically, which enhanced the efficacy of the inhibitor. NSG3 inhibited PD-L1 expression by suppressing Erk1/2 phosphorylation to improve the immune response to PC. NSG3 is, therefore, a potential new diagnostic and prognostic marker, particularly useful in immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Xigang Xia
- The Second Clinical Medical College, Jingzhou Central Hospital, Yangtze UniversityJingzhou 434020, China
- Pancreatic Surgery, Zhongnan Hospital of Wuhan UniversityWuhan 430022, China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
| | - Ran Li
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
- Guizhou Medical UniversityUniversity Town, Guian New District, Guizhou 550025, China
| | - Peng Zhou
- The Second Clinical Medical College, Jingzhou Central Hospital, Yangtze UniversityJingzhou 434020, China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
| | - Zhixiang Xing
- The Second Clinical Medical College, Jingzhou Central Hospital, Yangtze UniversityJingzhou 434020, China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
| | - Chao Lu
- The Second Clinical Medical College, Jingzhou Central Hospital, Yangtze UniversityJingzhou 434020, China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
| | - Zhida Long
- The Second Clinical Medical College, Jingzhou Central Hospital, Yangtze UniversityJingzhou 434020, China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
| | - Feiyang Wang
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Rui Wang
- The Second Clinical Medical College, Jingzhou Central Hospital, Yangtze UniversityJingzhou 434020, China
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan 430022, China
| |
Collapse
|
11
|
Marques-Coelho D, Iohan LDCC, Melo de Farias AR, Flaig A, Lambert JC, Costa MR. Differential transcript usage unravels gene expression alterations in Alzheimer's disease human brains. NPJ Aging Mech Dis 2021; 7:2. [PMID: 33398016 PMCID: PMC7782705 DOI: 10.1038/s41514-020-00052-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in aging individuals. Yet, the pathophysiological processes involved in AD onset and progression are still poorly understood. Among numerous strategies, a comprehensive overview of gene expression alterations in the diseased brain could contribute for a better understanding of the AD pathology. In this work, we probed the differential expression of genes in different brain regions of healthy and AD adult subjects using data from three large transcriptomic studies: Mayo Clinic, Mount Sinai Brain Bank (MSBB), and ROSMAP. Using a combination of differential expression of gene and isoform switch analyses, we provide a detailed landscape of gene expression alterations in the temporal and frontal lobes, harboring brain areas affected at early and late stages of the AD pathology, respectively. Next, we took advantage of an indirect approach to assign the complex gene expression changes revealed in bulk RNAseq to individual cell types/subtypes of the adult brain. This strategy allowed us to identify previously overlooked gene expression changes in the brain of AD patients. Among these alterations, we show isoform switches in the AD causal gene amyloid-beta precursor protein (APP) and the risk gene bridging integrator 1 (BIN1), which could have important functional consequences in neuronal cells. Altogether, our work proposes a novel integrative strategy to analyze RNAseq data in AD and other neurodegenerative diseases based on both gene/transcript expression and regional/cell-type specificities.
Collapse
Affiliation(s)
- Diego Marques-Coelho
- Brain Institute, Federal University of Rio Grande do Norte, Av. Nascimento de Castro, 2155, Natal, Brazil
- Bioinformatics Multidisciplinary Environment (BioME), Federal University of Rio Grande do Norte, Natal, Brazil
| | - Lukas da Cruz Carvalho Iohan
- Brain Institute, Federal University of Rio Grande do Norte, Av. Nascimento de Castro, 2155, Natal, Brazil
- Bioinformatics Multidisciplinary Environment (BioME), Federal University of Rio Grande do Norte, Natal, Brazil
| | - Ana Raquel Melo de Farias
- Brain Institute, Federal University of Rio Grande do Norte, Av. Nascimento de Castro, 2155, Natal, Brazil
- Unité INSERM 1167, RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, Institut Pasteur de Lille, University of Lille, Lille Cedex, France
| | - Amandine Flaig
- Unité INSERM 1167, RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, Institut Pasteur de Lille, University of Lille, 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, Lille Cedex, France
| | - Marcos Romualdo Costa
- Brain Institute, Federal University of Rio Grande do Norte, Av. Nascimento de Castro, 2155, Natal, Brazil.
- Unité INSERM 1167, RID-AGE-Risk Factors and Molecular Determinants of Aging-Related Diseases, Institut Pasteur de Lille, University of Lille, Lille Cedex, France.
| |
Collapse
|
12
|
Ren S, Breuillaud L, Yao W, Yin T, Norris KA, Zehntner SP, D'Adamio L. TNF-α-mediated reduction in inhibitory neurotransmission precedes sporadic Alzheimer's disease pathology in young Trem2 R47H rats. J Biol Chem 2021; 296:100089. [PMID: 33434745 PMCID: PMC7949092 DOI: 10.1074/jbc.ra120.016395] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/14/2020] [Accepted: 11/16/2020] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative dementia associated with deposition of amyloid plaques and neurofibrillary tangles, formed by amyloid β (Aβ) peptides and phosphor-tau, respectively, in the central nervous system. Approximately 2% of AD cases are due to familial AD (FAD); ∼98% of cases are sporadic AD (SAD). Animal models with FAD are commonly used to study SAD pathogenesis. Because mechanisms leading to FAD and SAD may be distinct, to study SAD pathogenesis, we generated Trem2R47H knock-in rats, which carry the SAD risk factor p.R47H variant of the microglia gene triggering receptor expressed on myeloid cells 2 (TREM2). Trem2R47H rats produce human-Aβ from a humanized-App rat allele because human-Aβ is more toxic than rodent-Aβ and the pathogenic role of the p.R47H TREM2 variant has been linked to human-Aβ-clearing deficits. Using periadolescent Trem2R47H rats, we previously demonstrated that supraphysiological tumor necrosis factor-α (TNF-α) boosts glutamatergic transmission, which is excitatory, and suppresses long-term potentiation, a surrogate of learning and memory. Here, we tested the effect of the p.R47H variant on the inhibitory neurotransmitter γ-aminobutyric acid. We report that GABAergic transmission is decreased in Trem2R47H/R47H rats. This decrease is due to acute and reversible action of TNF-α and is not associated with increased human-Aβ levels and AD pathology. Thus, the p.R47H variant changes the excitatory/inhibitory balance, favoring excitation. This imbalance could potentiate glutamate excitotoxicity and contribute to neuronal dysfunction, enhanced neuronal death, and neurodegeneration. Future studies will determine whether this imbalance represents an early, Aβ-independent pathway leading to dementia and may reveal the AD-modifying therapeutic potential of TNF-α inhibition in the central nervous system.
Collapse
Affiliation(s)
- Siqiang Ren
- Department of Pharmacology, Physiology & Neuroscience New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | | | - Wen Yao
- Department of Pharmacology, Physiology & Neuroscience New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Tao Yin
- Department of Pharmacology, Physiology & Neuroscience New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | - Kelly A Norris
- Department of Pharmacology, Physiology & Neuroscience New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, New Jersey, USA
| | | | - Luciano D'Adamio
- Department of Pharmacology, Physiology & Neuroscience New Jersey Medical School, Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, New Jersey, USA.
| |
Collapse
|
13
|
Ren S, Yao W, Tambini MD, Yin T, Norris KA, D'Adamio L. Microglia TREM2R47H Alzheimer-linked variant enhances excitatory transmission and reduces LTP via increased TNF-α levels. eLife 2020; 9:57513. [PMID: 32579116 PMCID: PMC7338048 DOI: 10.7554/elife.57513] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/23/2020] [Indexed: 01/09/2023] Open
Abstract
To study the mechanisms by which the p.R47H variant of the microglia gene and Alzheimer’s disease (AD) risk factor TREM2 increases dementia risk, we created Trem2R47H KI rats. Trem2R47H rats were engineered to produce human Aβ to define human-Aβ-dependent and -independent pathogenic mechanisms triggered by this variant. Interestingly, pre- and peri-adolescent Trem2R47H rats present increased brain concentrations of TNF-α, augmented glutamatergic transmission, suppression of Long-term-Potentiation (LTP), an electrophysiological surrogate of learning and memory, but normal Aβ levels. Acute reduction of TNF-α activity with a neutralizing anti-TNF-α antibody occludes the boost in amplitude of glutamatergic transmission and LTP suppression observed in young Trem2R47H/R47H rats. Thus, the microglia-specific pathogenic Trem2 variant boosts glutamatergic neuronal transmission and suppresses LTP by increasing brain TNF-α concentrations, directly linking microglia to neuronal dysfunction. Future studies will determine whether this phenomenon represents an early, Aβ-independent pathway that facilitates dementia pathogenesis in humans.
Collapse
Affiliation(s)
- Siqiang Ren
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Newark, United States.,Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, United States
| | - Wen Yao
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Newark, United States.,Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, United States
| | - Marc D Tambini
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Newark, United States.,Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, United States
| | - Tao Yin
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Newark, United States.,Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, United States
| | - Kelly A Norris
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Newark, United States.,Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, United States
| | - Luciano D'Adamio
- Department of Pharmacology, Physiology and Neuroscience, New Jersey Medical School, Newark, United States.,Brain Health Institute, Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research, Rutgers, The State University of New Jersey, Newark, United States
| |
Collapse
|
14
|
Tambini MD, Yao W, D'Adamio L. Facilitation of glutamate, but not GABA, release in Familial Alzheimer's APP mutant Knock-in rats with increased β-cleavage of APP. Aging Cell 2019; 18:e13033. [PMID: 31496118 PMCID: PMC6826143 DOI: 10.1111/acel.13033] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/11/2019] [Indexed: 12/01/2022] Open
Abstract
Amyloid precursor protein (APP) modulates glutamate release via cytoplasmic and intravesicular interactions with the synaptic vesicle release machinery. The intravesicular domain, called ISVAID, contains the BACE1 cleavage site of APP. We have tested the functional significance of BACE1 processing of APP using App‐Swedish (Apps) knock‐in rats, which carry an App mutation that causes familial Alzheimer's disease (FAD) in humans. We show that in Apps rats, β‐cleavage of APP is favored over α‐cleavage. Apps rats show facilitated glutamate, but not GABA, release. Our data support the notion that APP tunes glutamate release, and that BACE1 cleavage of the ISVAID segment of APP facilitates this function. We define this phenomenon as BACE1 on APP‐dependent glutamate release (BAD‐Glu). Unsurprisingly, Apps rats show no evidence of AD‐related pathology at 15 days and 3 months of age, indicating that alterations in BAD‐Glu are not caused by pathological lesions. The evidence that a pathogenic APP mutation causes an early enhancement of BAD‐Glu suggests that alterations of BACE1 processing of APP in glutamatergic synaptic vesicles could contribute to dementia.
Collapse
Affiliation(s)
- Marc D. Tambini
- Department of Pharmacology, Physiology & Neuroscience New Jersey Medical School Brain Health Institute Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research Rutgers, The State University of New Jersey Newark NJ USA
| | - Wen Yao
- Department of Pharmacology, Physiology & Neuroscience New Jersey Medical School Brain Health Institute Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research Rutgers, The State University of New Jersey Newark NJ USA
| | - Luciano D'Adamio
- Department of Pharmacology, Physiology & Neuroscience New Jersey Medical School Brain Health Institute Jacqueline Krieger Klein Center in Alzheimer's Disease and Neurodegeneration Research Rutgers, The State University of New Jersey Newark NJ USA
| |
Collapse
|
15
|
Tuning of Glutamate, But Not GABA, Release by an Intrasynaptic Vesicle APP Domain Whose Function Can Be Modulated by β- or α-Secretase Cleavage. J Neurosci 2019; 39:6992-7005. [PMID: 31235642 DOI: 10.1523/jneurosci.0207-19.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 11/21/2022] Open
Abstract
APP, whose mutations cause familial Alzheimer's disease (FAD), modulates neurotransmission via interaction of its cytoplasmic tail with the synaptic release machinery. Here we identified an intravesicular domain of APP, called intraluminal SV-APP interacting domain (ISVAID), which interacts with glutamatergic, but not GABAergic, synaptic vesicle proteins. ISVAID contains the β- and α-secretase cleavage sites of APP: proteomic analysis of the interactome of ISVAID suggests that β- and α-secretase cleavage of APP cuts inside the interaction domain of ISVAID and destabilizes protein-protein interactions. We have tested the functional significance of the ISVAID and of β-/α-secretase-processing of APP using various ISVAID-derived peptides in competition experiments on both female and male mouse and rats hippocampal slices. A peptide encompassing the entire ISVAID facilitated glutamate, but not GABA, release acting as dominant negative inhibitor of the functions of this APP domain in acute hippocampal slices. In contrast, peptides representing the product of β-/α-secretase-processing of ISVAID did not alter excitatory neurotransmitter release. These findings suggest that cleavage of APP by either β- or α-secretase may inactivate the ISVAID, thereby enhancing glutamate release. Our present data support the notion that APP tunes glutamate release, likely through intravesicular and extravesicular interactions with synaptic vesicle proteins and the neurotransmitter release machinery, and that β-/α cleavage of APP facilitates the release of excitatory neurotransmitter.SIGNIFICANCE STATEMENT Alzheimer's disease has been linked to mutations in APP. However, the biological function of APP is poorly understood. Here we show that an intravesicular APP domain interacts with the proteins that control the release of glutamate, but not GABA. Interfering with the function of this domain promotes glutamate release. This APP domain contains the sites cleaved by β- and α-secretases: our data suggest that β-/α cleavage of APP inactivates this functional APP domain promoting excitatory neurotransmitter release.
Collapse
|
16
|
Dinamarca MC, Raveh A, Schneider A, Fritzius T, Früh S, Rem PD, Stawarski M, Lalanne T, Turecek R, Choo M, Besseyrias V, Bildl W, Bentrop D, Staufenbiel M, Gassmann M, Fakler B, Schwenk J, Bettler B. Complex formation of APP with GABA B receptors links axonal trafficking to amyloidogenic processing. Nat Commun 2019; 10:1331. [PMID: 30902970 PMCID: PMC6430795 DOI: 10.1038/s41467-019-09164-3] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 02/21/2019] [Indexed: 12/30/2022] Open
Abstract
GABAB receptors (GBRs) are key regulators of synaptic release but little is known about trafficking mechanisms that control their presynaptic abundance. We now show that sequence-related epitopes in APP, AJAP-1 and PIANP bind with nanomolar affinities to the N-terminal sushi-domain of presynaptic GBRs. Of the three interacting proteins, selectively the genetic loss of APP impaired GBR-mediated presynaptic inhibition and axonal GBR expression. Proteomic and functional analyses revealed that APP associates with JIP and calsyntenin proteins that link the APP/GBR complex in cargo vesicles to the axonal trafficking motor. Complex formation with GBRs stabilizes APP at the cell surface and reduces proteolysis of APP to Aβ, a component of senile plaques in Alzheimer's disease patients. Thus, APP/GBR complex formation links presynaptic GBR trafficking to Aβ formation. Our findings support that dysfunctional axonal trafficking and reduced GBR expression in Alzheimer's disease increases Aβ formation.
Collapse
Affiliation(s)
- Margarita C Dinamarca
- Department of Biomedicine, Institute of Physiology, University of Basel, Klingelbergstr. 50/70, 4056, Basel, Switzerland
| | - Adi Raveh
- Department of Biomedicine, Institute of Physiology, University of Basel, Klingelbergstr. 50/70, 4056, Basel, Switzerland
| | - Andy Schneider
- Faculty of Medicine, Institute of Physiology, University of Freiburg, Hermann-Herder-Str. 7, 79104, Freiburg, Germany
| | - Thorsten Fritzius
- Department of Biomedicine, Institute of Physiology, University of Basel, Klingelbergstr. 50/70, 4056, Basel, Switzerland
| | - Simon Früh
- Department of Biomedicine, Institute of Physiology, University of Basel, Klingelbergstr. 50/70, 4056, Basel, Switzerland
| | - Pascal D Rem
- Department of Biomedicine, Institute of Physiology, University of Basel, Klingelbergstr. 50/70, 4056, Basel, Switzerland
| | - Michal Stawarski
- Department of Biomedicine, Institute of Physiology, University of Basel, Klingelbergstr. 50/70, 4056, Basel, Switzerland
| | - Txomin Lalanne
- Department of Biomedicine, Institute of Physiology, University of Basel, Klingelbergstr. 50/70, 4056, Basel, Switzerland
| | - Rostislav Turecek
- Department of Biomedicine, Institute of Physiology, University of Basel, Klingelbergstr. 50/70, 4056, Basel, Switzerland
- Institute of Experimental Medicine, ASCR, Vı´denska´ 1083, 14220, Prague 4-Krc, Czech Republic
| | - Myeongjeong Choo
- Department of Biomedicine, Institute of Physiology, University of Basel, Klingelbergstr. 50/70, 4056, Basel, Switzerland
| | - Valérie Besseyrias
- Department of Biomedicine, Institute of Physiology, University of Basel, Klingelbergstr. 50/70, 4056, Basel, Switzerland
| | - Wolfgang Bildl
- Faculty of Medicine, Institute of Physiology, University of Freiburg, Hermann-Herder-Str. 7, 79104, Freiburg, Germany
| | - Detlef Bentrop
- Faculty of Medicine, Institute of Physiology, University of Freiburg, Hermann-Herder-Str. 7, 79104, Freiburg, Germany
| | - Matthias Staufenbiel
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Otfried-Müller-Strasse 27, 72076, Tübingen, Germany
| | - Martin Gassmann
- Department of Biomedicine, Institute of Physiology, University of Basel, Klingelbergstr. 50/70, 4056, Basel, Switzerland
| | - Bernd Fakler
- Faculty of Medicine, Institute of Physiology, University of Freiburg, Hermann-Herder-Str. 7, 79104, Freiburg, Germany
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany
| | - Jochen Schwenk
- Faculty of Medicine, Institute of Physiology, University of Freiburg, Hermann-Herder-Str. 7, 79104, Freiburg, Germany.
- Signalling Research Centers BIOSS and CIBSS, University of Freiburg, Schänzlestr. 18, 79104, Freiburg, Germany.
| | - Bernhard Bettler
- Department of Biomedicine, Institute of Physiology, University of Basel, Klingelbergstr. 50/70, 4056, Basel, Switzerland.
| |
Collapse
|
17
|
Neuron-Specific Gene 2 (NSG2) Encodes an AMPA Receptor Interacting Protein That Modulates Excitatory Neurotransmission. eNeuro 2019; 6:eN-NWR-0292-18. [PMID: 30680309 PMCID: PMC6345199 DOI: 10.1523/eneuro.0292-18.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 12/13/2018] [Accepted: 12/17/2018] [Indexed: 01/10/2023] Open
Abstract
Neurons have evolved a number of unique protein-coding genes that regulate trafficking of protein complexes within small organelles throughout dendrites and axons. Neuron-specific gene 2 (NSG2) encodes for one of the most abundant proteins in the nervous system during perinatal development. NSG2 belongs to a family of small neuronal endosomal proteins but its function has remained uncharacterized to date. Here, we show that NSG2 is found in discrete punctae restricted to the somatodendritic arbors of developing mouse and human neurons, and a significant proportion of NSG2 punctae colocalize with postsynaptic HOMER1 and surface-expressed AMPA-type glutamate receptors (AMPARs) at excitatory synapses. Immunoprecipitation revealed that NSG2 physically interacts with both the GluA1 and GluA2 AMPAR subunits in mouse brain. Knock-out of NSG2 in mouse hippocampal neurons selectively impaired the frequency of miniature EPSCs (mEPSCs) and caused alterations in PSD95 expression at postsynaptic densities (PSDs). In contrast, NSG2 overexpression caused a significant increase in the amplitude of mEPSCs as well as GluA2 surface expression. Thus, NSG2 functions as an AMPAR-binding protein that is required for normal synapse formation and/or maintenance, and has unique functions compared with other NSG family members.
Collapse
|
18
|
Wang Z, Xu Q, Cai F, Liu X, Wu Y, Song W. BACE2, a conditional β-secretase, contributes to Alzheimer's disease pathogenesis. JCI Insight 2019; 4:123431. [PMID: 30626751 DOI: 10.1172/jci.insight.123431] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 12/05/2018] [Indexed: 12/21/2022] Open
Abstract
Deposition of amyloid-β protein (Aβ) to form neuritic plaques is the characteristic neuropathology of Alzheimer's disease (AD). Aβ is generated from amyloid precursor protein (APP) by β- and γ-secretase cleavages. BACE1 is the β-secretase and its inhibition induces severe side effects, whereas its homolog BACE2 normally suppresses Aβ by cleaving APP/Aβ at the θ-site (Phe20) within the Aβ domain. Here, we report that BACE2 also processes APP at the β site, and the juxtamembrane helix (JH) of APP inhibits its β-secretase activity, enabling BACE2 to cleave nascent APP and aggravate AD symptoms. JH-disrupting mutations and clusterin binding to JH triggered BACE2-mediated β-cleavage. Both BACE2 and clusterin were elevated in aged mouse brains, and enhanced β-cleavage during aging. Therefore, BACE2 contributes to AD pathogenesis as a conditional β-secretase and could be a preventive and therapeutic target for AD without the side effects of BACE1 inhibition.
Collapse
Affiliation(s)
- Zhe Wang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China.,Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, British Columbia, Canada.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qin Xu
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Fang Cai
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Xi Liu
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Yili Wu
- Department of Psychiatry, Jining Medical University, Jining, Shandong, China
| | - Weihong Song
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, China.,Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, Vancouver, British Columbia, Canada.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Yap CC, Digilio L, McMahon LP, Garcia ADR, Winckler B. Degradation of dendritic cargos requires Rab7-dependent transport to somatic lysosomes. J Cell Biol 2018; 217:3141-3159. [PMID: 29907658 PMCID: PMC6122995 DOI: 10.1083/jcb.201711039] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 03/30/2018] [Accepted: 05/31/2018] [Indexed: 12/13/2022] Open
Abstract
Neurons are large and long lived, creating high needs for regulating protein turnover. Disturbances in proteostasis lead to aggregates and cellular stress. We characterized the behavior of the short-lived dendritic membrane proteins Nsg1 and Nsg2 to determine whether these proteins are degraded locally in dendrites or centrally in the soma. We discovered a spatial heterogeneity of endolysosomal compartments in dendrites. Early EEA1-positive and late Rab7-positive endosomes are found throughout dendrites, whereas the density of degradative LAMP1- and cathepsin (Cat) B/D-positive lysosomes decreases steeply past the proximal segment. Unlike in fibroblasts, we found that the majority of dendritic Rab7 late endosomes (LEs) do not contain LAMP1 and that a large proportion of LAMP1 compartments do not contain CatB/D. Second, Rab7 activity is required to mobilize distal predegradative LEs for transport to the soma and terminal degradation. We conclude that the majority of dendritic LAMP1 endosomes are not degradative lysosomes and that terminal degradation of dendritic cargos such as Nsg1, Nsg2, and DNER requires Rab7-dependent transport in LEs to somatic lysosomes.
Collapse
Affiliation(s)
- Chan Choo Yap
- Department of Cell Biology, University of Virginia, Charlottesville, VA
| | - Laura Digilio
- Department of Cell Biology, University of Virginia, Charlottesville, VA
| | - Lloyd P McMahon
- Department of Cell Biology, University of Virginia, Charlottesville, VA
| | | | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, VA
| |
Collapse
|
20
|
Maiorano AM, Lourenco DL, Tsuruta S, Ospina AMT, Stafuzza NB, Masuda Y, Filho AEV, Cyrillo JNDSG, Curi RA, Silva JAIIDV. Assessing genetic architecture and signatures of selection of dual purpose Gir cattle populations using genomic information. PLoS One 2018; 13:e0200694. [PMID: 30071036 PMCID: PMC6071998 DOI: 10.1371/journal.pone.0200694] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 07/02/2018] [Indexed: 12/31/2022] Open
Abstract
Gir is one of the main cattle breeds raised in tropical South American countries. Strong artificial selection through its domestication resulted in increased genetic differentiation among the countries in recent years. Over the years, genomic studies in Gir have become more common. However, studies of population structure and signatures of selection in divergent Gir populations are scarce and need more attention to better understand genetic differentiation, gene flow, and genetic distance. Genotypes of 173 animals selected for growth traits and 273 animals selected for milk production were used in this study. Clear genetic differentiation between beef and dairy populations was observed. Different criteria led to genetic divergence and genetic differences in allele frequencies between the two populations. Gene segregation in each population was forced by artificial selection, promoting isolation, and increasing genetic variation between them. Results showed evidence of selective forces in different regions of the genome. A total of 282 genes were detected under selection in the test population based on the fixation index (Fst), integrated haplotype score (iHS), and cross-population extend haplotype homozygosity (XP-EHH) approaches. The QTL mapping identified 35 genes associated with reproduction, milk composition, growth, meat and carcass, health, or body conformation traits. The investigation of genes and pathways showed that quantitative traits associated to fertility, milk production, beef quality, and growth were involved in the process of differentiation of these populations. These results would support further investigations of population structure and differentiation in the Gir breed.
Collapse
Affiliation(s)
- Amanda Marchi Maiorano
- Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista “Júlio de Mesquita Filho”, Jaboticabal, Sao Paulo, Brazil
- * E-mail:
| | - Daniela Lino Lourenco
- Animal and Dairy Science, Animal Breeding and Genetics, University of Georgia, Athens, Georgia, United States of America
| | - Shogo Tsuruta
- Animal and Dairy Science, Animal Breeding and Genetics, University of Georgia, Athens, Georgia, United States of America
| | - Alejandra Maria Toro Ospina
- Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista “Júlio de Mesquita Filho”, Jaboticabal, Sao Paulo, Brazil
| | - Nedenia Bonvino Stafuzza
- Faculdade de Ciências Agrárias e Veterinárias, Universidade Estadual Paulista “Júlio de Mesquita Filho”, Jaboticabal, Sao Paulo, Brazil
| | - Yutaka Masuda
- Animal and Dairy Science, Animal Breeding and Genetics, University of Georgia, Athens, Georgia, United States of America
| | | | | | - Rogério Abdallah Curi
- Faculdade de Medicina Veterinária e Zootecnia, Universidade Estadual Paulista “Júlio de Mesquita Filho”, Botucatu, Sao Paulo, Brazil
| | | |
Collapse
|
21
|
Yap CC, Digilio L, McMahon L, Winckler B. The endosomal neuronal proteins Nsg1/NEEP21 and Nsg2/P19 are itinerant, not resident proteins of dendritic endosomes. Sci Rep 2017; 7:10481. [PMID: 28874679 PMCID: PMC5585371 DOI: 10.1038/s41598-017-07667-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 06/29/2017] [Indexed: 12/28/2022] Open
Abstract
Membrane traffic critically regulates most aspects of neuronal function. Neurons express many neuronal-specific proteins that regulate membrane traffic, including the poorly understood small transmembrane proteins neural-specific gene 1 and 2 (Nsg1/NEEP21 and Nsg2/P19). Nsg1 has been implicated in regulating endosomal recycling and sorting of several important neuronal receptors. Nsg2 is largely unstudied. At steady-state, Nsg1 and Nsg2 only partially co-localize with known endosomal compartments, and it was suggested that they mark a neuronal-specific endosome. Since Nsg1 localizes to and functions in the dendritic endosome, we set out to discover how Nsg1 and Nsg2 localization to endosomes is regulated in primary rat hippocampal neurons, using quadruple immunolocalization against endogenous proteins, live imaging of dendritic endosomes, and interference approaches against the endosomal regulators Rab5 and Rab7. In contrast to previous conclusions, we now show that Nsg1 and Nsg2 are not resident endosomal proteins, but traffic rapidly from the cell surface to lysosomes and have a half-life of less than two hours. Their partial co-localization with canonical endosomal markers thus reflects their rapid flux towards degradation rather than specific targeting to a singular compartment. These findings will require rethinking of how this class of endosomal proteins regulates trafficking of much longer-lived receptors.
Collapse
Affiliation(s)
- Chan Choo Yap
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Laura Digilio
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Lloyd McMahon
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
22
|
Shi L, Muthusamy N, Smith D, Bergson C. Dynein binds and stimulates axonal motility of the endosome adaptor and NEEP21 family member, calcyon. Int J Biochem Cell Biol 2017; 90:93-102. [PMID: 28734834 DOI: 10.1016/j.biocel.2017.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 07/11/2017] [Accepted: 07/12/2017] [Indexed: 01/31/2023]
Abstract
The neuron-enriched, endosomal protein Calcyon (Caly) regulates endocytosis and vesicle sorting, and is important for synaptic plasticity and brain development. In the current investigation of Caly interacting proteins in brain, the microtubule retrograde motor subunit, cytoplasmic dynein 1 heavy chain (DYNC1H), and microtubule structural proteins, α and β tubulin, were identified as Caly associated proteins by MALDI-ToF/ToF. Direct interaction of the Caly-C terminus with dynein and tubulin was further confirmed in in vitro studies. In Cos-7 cells, mCherry-Caly moved along the microtubule network in organelles largely labeled by the late endosome marker Rab7. Expression of the dynein inhibitor CC1, produced striking alterations in Caly distribution, consistent with retrograde motors playing a prominent role in Caly localization and movement. In axons of cultured adult rat sensory neurons, Caly-positive organelles co-localized with dynein intermediate chain (DYNC1I1-isoform IC-1B) and the dynein regulator, lissencephaly 1 (LIS1), both of which co-precipitated from brain with the Caly C-terminus. Manipulation of dynein function in axons altered the motile properties of Caly indicating that Caly vesicles utilize the retrograde motor. Altogether, the current evidence for association with dynein motors raises the possibility that the endocytic and cargo sorting functions of Caly in neurons could be regulated by interaction with the microtubule transport system.
Collapse
Affiliation(s)
- Liang Shi
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Nagendran Muthusamy
- Department of Neuroscience, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Deanna Smith
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
| | - Clare Bergson
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
23
|
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: 398] [Impact Index Per Article: 56.9] [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
|
24
|
Barford K, Yap CC, Dwyer ND, Winckler B. The related neuronal endosomal proteins NEEP21 (Nsg1) and P19 (Nsg2) have divergent expression profiles in vivo. J Comp Neurol 2017; 525:1861-1878. [PMID: 28299779 DOI: 10.1002/cne.24168] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 10/28/2016] [Accepted: 11/18/2016] [Indexed: 01/23/2023]
Abstract
Endosomal maturation and transport constitutes a complex trafficking system present in all cell types. Neurons have adapted their endosomal system to meet their unique and complex needs. These adaptations include repurposing existing proteins to diversify endocytosis and trafficking, as well as preferential expression of certain regulators more highly in neurons than other cell types. These neuronal regulators include the family of Neuron-Specific Gene family members (Nsg), NEEP21 (Nsg1), and P19 (Nsg2). NEEP21/Nsg1 plays a role in the trafficking of multiple receptors, including the cell adhesion molecule L1/NgCAM, the neurotransmitter receptor GluA2, and β-APP. Recently, we showed that NEEP2/Nsg1 and P19/Nsg2 are not expressed in all neuronal cell types in vitro. However, it is not known where and when NEEP21/Nsg1 and P19/Nsg2 are expressed in vivo, and whether both proteins are always coexpressed. Here, we show that NEEP21/Nsg1 and P19/Nsg2 are present in both overlapping and distinct cell populations in the hippocampus, neocortex, and cerebellum during development. NEEP21/Nsg1 and P19/Nsg2 levels are highest during embryonic development, and expression persists in the juvenile mouse brain. In particular, a subset of layer V cortical neurons retains relatively high expression of both NEEP21/Nsg1 and P19/Nsg2 at postnatal day 16 as well as in the CA1-3 regions of the hippocampus. In the cerebellum, NEEP21/Nsg1 expression becomes largely restricted to Purkinje neurons in adulthood whereas P19/Nsg2 expression strikingly disappears from the cerebellum with age. This divergent and restricted expression likely reflects differential needs for this class of trafficking regulators in different neurons during different stages of maturation.
Collapse
Affiliation(s)
- Kelly Barford
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Chan Choo Yap
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Noelle D Dwyer
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| | - Bettina Winckler
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
25
|
Ludewig S, Korte M. Novel Insights into the Physiological Function of the APP (Gene) Family and Its Proteolytic Fragments in Synaptic Plasticity. Front Mol Neurosci 2017; 9:161. [PMID: 28163673 PMCID: PMC5247455 DOI: 10.3389/fnmol.2016.00161] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 12/14/2016] [Indexed: 12/31/2022] Open
Abstract
The amyloid precursor protein (APP) is well known to be involved in the pathophysiology of Alzheimer's disease (AD) via its cleavage product amyloid ß (Aß). However, the physiological role of APP, its various proteolytic products and the amyloid precursor-like proteins 1 and 2 (APLP1/2) are still not fully clarified. Interestingly, it has been shown that learning and memory processes represented by functional and structural changes at synapses are altered in different APP and APLP1/2 mouse mutants. In addition, APP and its fragments are implicated in regulating synaptic strength further reinforcing their modulatory role at the synapse. While APLP2 and APP are functionally redundant, the exclusively CNS expressed APLP1, might have individual roles within the synaptic network. The proteolytic product of non-amyloidogenic APP processing, APPsα, emerged as a neurotrophic peptide that facilitates long-term potentiation (LTP) and restores impairments occurring with age. Interestingly, the newly discovered η-secretase cleavage product, An-α acts in the opposite direction, namely decreasing LTP. In this review we summarize recent findings with emphasis on the physiological role of the APP gene family and its proteolytic products on synaptic function and plasticity, especially during processes of hippocampal LTP. Therefore, we focus on literature that provide electrophysiological data by using different mutant mouse strains either lacking full-length or parts of the APP proteins or that utilized secretase inhibitors as well as secreted APP fragments.
Collapse
Affiliation(s)
- Susann Ludewig
- Division of Cellular Neurobiology, Zoological Institute, TU Braunschweig Braunschweig, Germany
| | - Martin Korte
- Division of Cellular Neurobiology, Zoological Institute, TU BraunschweigBraunschweig, Germany; Helmholtz Centre for Infection Research, AG NINDBraunschweig, Germany
| |
Collapse
|
26
|
Miller AJ, Roman B, Norstrom E. A method for easily customizable gradient gel electrophoresis. Anal Biochem 2016; 509:12-14. [DOI: 10.1016/j.ab.2016.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 07/01/2016] [Accepted: 07/04/2016] [Indexed: 10/21/2022]
|
27
|
Del Prete D, Rice RC, Rajadhyaksha AM, D'Adamio L. Amyloid Precursor Protein (APP) May Act as a Substrate and a Recognition Unit for CRL4CRBN and Stub1 E3 Ligases Facilitating Ubiquitination of Proteins Involved in Presynaptic Functions and Neurodegeneration. J Biol Chem 2016; 291:17209-27. [PMID: 27325702 DOI: 10.1074/jbc.m116.733626] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Indexed: 12/23/2022] Open
Abstract
The amyloid precursor protein (APP), whose mutations cause Alzheimer disease, plays an important in vivo role and facilitates transmitter release. Because the APP cytosolic region (ACR) is essential for these functions, we have characterized its brain interactome. We found that the ACR interacts with proteins that regulate the ubiquitin-proteasome system, predominantly with the E3 ubiquitin-protein ligases Stub1, which binds the NH2 terminus of the ACR, and CRL4(CRBN), which is formed by Cul4a/b, Ddb1, and Crbn, and interacts with the COOH terminus of the ACR via Crbn. APP shares essential functions with APP-like protein-2 (APLP2) but not APP-like protein-1 (APLP1). Noteworthy, APLP2, but not APLP1, interacts with Stub1 and CRL4(CRBN), pointing to a functional pathway shared only by APP and APLP2. In vitro ubiquitination/ubiquitome analysis indicates that these E3 ligases are enzymatically active and ubiquitinate the ACR residues Lys(649/650/651/676/688) Deletion of Crbn reduces ubiquitination of Lys(676) suggesting that Lys(676) is physiologically ubiquitinated by CRL4(CRBN) The ACR facilitated in vitro ubiquitination of presynaptic proteins that regulate exocytosis, suggesting a mechanism by which APP tunes transmitter release. Other dementia-related proteins, namely Tau and apoE, interact with and are ubiquitinated via the ACR in vitro This, and the evidence that CRBN and CUL4B are linked to intellectual disability, prompts us to hypothesize a pathogenic mechanism, in which APP acts as a modulator of E3 ubiquitin-protein ligase(s), shared by distinct neuronal disorders. The well described accumulation of ubiquitinated protein inclusions in neurodegenerative diseases and the link between the ubiquitin-proteasome system and neurodegeneration make this concept plausible.
Collapse
Affiliation(s)
- Dolores Del Prete
- From the Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461 and
| | - Richard C Rice
- the Division of Pediatric Neurology, Department of Pediatrics, and
| | - Anjali M Rajadhyaksha
- the Division of Pediatric Neurology, Department of Pediatrics, and Feil Family Brain and Mind Research Institute, Weill Cornell Autism Research Program, Weill Cornell Medical College, New York, New York 10065
| | - Luciano D'Adamio
- From the Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York 10461 and
| |
Collapse
|
28
|
Laßek M, Weingarten J, Wegner M, Mueller BF, Rohmer M, Baeumlisberger D, Arrey TN, Hick M, Ackermann J, Acker-Palmer A, Koch I, Müller U, Karas M, Volknandt W. APP Is a Context-Sensitive Regulator of the Hippocampal Presynaptic Active Zone. PLoS Comput Biol 2016; 12:e1004832. [PMID: 27092780 PMCID: PMC4836664 DOI: 10.1371/journal.pcbi.1004832] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/25/2016] [Indexed: 01/18/2023] Open
Abstract
The hallmarks of Alzheimer's disease (AD) are characterized by cognitive decline and behavioral changes. The most prominent brain region affected by the progression of AD is the hippocampal formation. The pathogenesis involves a successive loss of hippocampal neurons accompanied by a decline in learning and memory consolidation mainly attributed to an accumulation of senile plaques. The amyloid precursor protein (APP) has been identified as precursor of Aβ-peptides, the main constituents of senile plaques. Until now, little is known about the physiological function of APP within the central nervous system. The allocation of APP to the proteome of the highly dynamic presynaptic active zone (PAZ) highlights APP as a yet unknown player in neuronal communication and signaling. In this study, we analyze the impact of APP deletion on the hippocampal PAZ proteome. The native hippocampal PAZ derived from APP mouse mutants (APP-KOs and NexCreAPP/APLP2-cDKOs) was isolated by subcellular fractionation and immunopurification. Subsequently, an isobaric labeling was performed using TMT6 for protein identification and quantification by high-resolution mass spectrometry. We combine bioinformatics tools and biochemical approaches to address the proteomics dataset and to understand the role of individual proteins. The impact of APP deletion on the hippocampal PAZ proteome was visualized by creating protein-protein interaction (PPI) networks that incorporated APP into the synaptic vesicle cycle, cytoskeletal organization, and calcium-homeostasis. The combination of subcellular fractionation, immunopurification, proteomic analysis, and bioinformatics allowed us to identify APP as structural and functional regulator in a context-sensitive manner within the hippocampal active zone network.
Collapse
Affiliation(s)
- Melanie Laßek
- Institute for Cell Biology and Neuroscience, Biologicum, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Jens Weingarten
- Institute for Cell Biology and Neuroscience, Biologicum, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Martin Wegner
- Institute for Molecular Bioinformatics, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Benjamin F. Mueller
- Institute of Pharmaceutical Chemistry, Cluster of Excellence “Macromolecular Complexes”, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Marion Rohmer
- Institute of Pharmaceutical Chemistry, Cluster of Excellence “Macromolecular Complexes”, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | | | | | - Meike Hick
- Department of Pharmacy and Molecular Biotechnology, University Heidelberg, Heidelberg Germany
| | - Jörg Ackermann
- Institute for Molecular Bioinformatics, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Amparo Acker-Palmer
- Institute for Cell Biology and Neuroscience, Biologicum, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Ina Koch
- Institute for Molecular Bioinformatics, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Ulrike Müller
- Department of Pharmacy and Molecular Biotechnology, University Heidelberg, Heidelberg Germany
| | - Michael Karas
- Institute of Pharmaceutical Chemistry, Cluster of Excellence “Macromolecular Complexes”, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
| | - Walter Volknandt
- Institute for Cell Biology and Neuroscience, Biologicum, Johann Wolfgang Goethe-University, Frankfurt am Main, Germany
- * E-mail:
| |
Collapse
|
29
|
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: 10.1] [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
|
30
|
Fanutza T, Del Prete D, Ford MJ, Castillo PE, D’Adamio L. APP and APLP2 interact with the synaptic release machinery and facilitate transmitter release at hippocampal synapses. eLife 2015; 4:e09743. [PMID: 26551565 PMCID: PMC4755753 DOI: 10.7554/elife.09743] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 11/08/2015] [Indexed: 12/16/2022] Open
Abstract
The amyloid precursor protein (APP), whose mutations cause familial Alzheimer's disease, interacts with the synaptic release machinery, suggesting a role in neurotransmission. Here we mapped this interaction to the NH2-terminal region of the APP intracellular domain. A peptide encompassing this binding domain -named JCasp- is naturally produced by a γ-secretase/caspase double-cut of APP. JCasp interferes with the APP-presynaptic proteins interaction and, if linked to a cell-penetrating peptide, reduces glutamate release in acute hippocampal slices from wild-type but not APP deficient mice, indicating that JCasp inhibits APP function.The APP-like protein-2 (APLP2) also binds the synaptic release machinery. Deletion of APP and APLP2 produces synaptic deficits similar to those caused by JCasp. Our data support the notion that APP and APLP2 facilitate transmitter release, likely through the interaction with the neurotransmitter release machinery. Given the link of APP to Alzheimer's disease, alterations of this synaptic role of APP could contribute to dementia.
Collapse
Affiliation(s)
- Tomas Fanutza
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, United States
| | - Dolores Del Prete
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, United States
| | | | - Pablo E Castillo
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, New York, United States
| | - Luciano D’Adamio
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, United States
| |
Collapse
|
31
|
Ctip2-, Satb2-, Prox1-, and GAD65-Expressing Neurons in Rat Cultures: Preponderance of Single- and Double-Positive Cells, and Cell Type-Specific Expression of Neuron-Specific Gene Family Members, Nsg-1 (NEEP21) and Nsg-2 (P19). PLoS One 2015; 10:e0140010. [PMID: 26465886 PMCID: PMC4605768 DOI: 10.1371/journal.pone.0140010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/31/2015] [Indexed: 11/19/2022] Open
Abstract
The brain consists of many distinct neuronal cell types, but which cell types are present in widely used primary cultures of embryonic rodent brain is often not known. We characterized how abundantly four cell type markers (Ctip2, Satb2, Prox1, GAD65) were represented in cultured rat neurons, how easily neurons expressing different markers can be transfected with commonly used plasmids, and whether neuronal-enriched endosomal proteins Nsg-1 (NEEP21) and Nsg-2 (P19) are ubiquitously expressed in all types of cultured neurons. We found that cultured neurons stably maintain cell type identities that are reflective of cell types in vivo. This includes neurons maintaining simultaneous expression of two transcription factors, such as Ctip2+/Satb2+ or Prox1+/Ctip2+ double-positive cells, which have also been described in vivo. Secondly, we established the superior efficiency of CAG promoters for both Lipofectamine-mediated transfection as well as for electroporation. Thirdly, we discovered that Nsg-1 and Nsg-2 were not expressed equally in all neurons: whereas high levels of both Nsg-1 and Nsg-2 were found in Satb2-, Ctip2-, and GAD65-positive neurons, Prox1-positive neurons in hippocampal cultures expressed low levels of both. Our findings thus highlight the importance of identifying neuronal cell types for doing cell biology in cultured neurons: Keeping track of neuronal cell type might uncover effects in assays that might otherwise be masked by the mixture of responsive and non-responsive neurons in the dish.
Collapse
|
32
|
Calcyon stimulates neuregulin 1 maturation and signaling. Mol Psychiatry 2015; 20:1251-60. [PMID: 25349163 DOI: 10.1038/mp.2014.131] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 07/27/2014] [Accepted: 08/21/2014] [Indexed: 01/02/2023]
Abstract
Neuregulin1 (NRG1) is a single transmembrane protein that plays a critical role in neural development and synaptic plasticity. Both NRG1 and its receptor, ErbB4, are well-established risk genes of schizophrenia. The NRG1 ecto-domain (ED) binds and activates ErbB4 following proteolytic cleavage of pro-NRG1 precursor protein. Although several studies have addressed the function of NRG1 in brain, very little is known about the cleavage and shedding mechanism. Here we show that the neuronal vesicular protein calcyon is a potent activator and key determinant of NRG1 ED cleavage and shedding. Calcyon stimulates clathrin-mediated endocytosis and endosomal targeting; and its levels are elevated in postmortem brains of schizophrenics. Overexpression of calcyon stimulates NRG1 cleavage and signaling in vivo, and as a result, GABA transmission is enhanced in calcyon overexpressing mice. Conversely, NRG1 cleavage, ErbB4 activity and GABA transmission are decreased in calcyon null mice. Moreover, stimulation of NRG1 cleavage by calcyon was recapitulated in HEK 293 cells suggesting the mechanism involved is cell-autonomous. Finally, studies with site-specific mutants in calcyon and inhibitors for the major sheddases indicate that the stimulatory effects of calcyon on NRG1 cleavage and shedding depend on clathrin-mediated endocytosis, β-secretase 1, and interaction with clathrin adaptor proteins. Together these results identify a novel mechanism for NRG1 cleavage and shedding.
Collapse
|
33
|
Gautam V, D'Avanzo C, Berezovska O, Tanzi RE, Kovacs DM. Synaptotagmins interact with APP and promote Aβ generation. Mol Neurodegener 2015. [PMID: 26202512 PMCID: PMC4511450 DOI: 10.1186/s13024-015-0028-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Background Accumulation of the β-amyloid peptide (Aβ) is a major pathological hallmark of Alzheimer’s disease (AD). Recent studies have shown that synaptic Aβ toxicity may directly impair synaptic function. However, proteins regulating Aβ generation at the synapse have not been characterized. Here, we sought to identify synaptic proteins that interact with the extracellular domain of APP and regulate Aβ generation. Results Affinity purification-coupled mass spectrometry identified members of the Synaptotagmin (Syt) family as novel interacting proteins with the APP ectodomain in mouse brains. Syt-1, −2 and −9 interacted with APP in cells and in mouse brains in vivo. Using a GST pull-down approach, we have further demonstrated that the Syt interaction site lies in the 108 amino acids linker region between the E1 and KPI domains of APP. Stable overexpression of Syt-1 or Syt-9 with APP in CHO and rat pheochromocytoma cells (PC12) significantly increased APP-CTF and sAPP levels, with a 2 to 3 fold increase in secreted Aβ levels in PC12 cells. Moreover, using a stable knockdown approach to reduce the expression of endogenous Syt-1 in PC12 cells, we have observed a ~ 50 % reduction in secreted Aβ generation. APP processing also decreased in these cells, shown by lower CTF levels. Lentiviral-mediated knock down of endogenous Syt-1 in mouse primary neurons also led to a significant reduction in both Aβ40 and Aβ42 generation. As secreted sAPPβ levels were significantly reduced in PC12 cells lacking Syt-1 expression, our results suggest that Syt-1 regulates Aβ generation by modulating BACE1-mediated cleavage of APP. Conclusion Altogether, our data identify the synaptic vesicle proteins Syt-1 and 9 as novel APP-interacting proteins that promote Aβ generation and thus may play an important role in the pathogenesis of AD. Electronic supplementary material The online version of this article (doi:10.1186/s13024-015-0028-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Vivek Gautam
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| | - Carla D'Avanzo
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| | - Oksana Berezovska
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| | - Dora M Kovacs
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| |
Collapse
|
34
|
Muthusamy N, Chen YJ, Yin DM, Mei L, Bergson C. Complementary roles of the neuron-enriched endosomal proteins NEEP21 and calcyon in neuronal vesicle trafficking. J Neurochem 2015; 132:20-31. [PMID: 25376768 DOI: 10.1111/jnc.12989] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 10/17/2014] [Accepted: 10/23/2014] [Indexed: 01/18/2023]
Abstract
Understanding mechanisms governing the trafficking of transmembrane (TM) cargoes to synapses and other specialized membranes in neurons represents a long-standing challenge in cell biology. Investigation of the neuron-enriched endosomal protein of 21 kDa (NEEP21, or NSG1or P21) and Calcyon (Caly, or NSG3) indicates that the emergence of the NEEP21/Caly/P19 gene family could play a vital role in the success of these mechanisms in vertebrates. The upshot of a sizeable body of work is that the NEEP21 and Caly perform distinct endocytic and recycling functions, which impact (i) α amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid-type glutamate receptor trafficking at excitatory synapses; (ii) transport to/in neuronal axons; as well as (iii) proteolytic processing of amyloid precursor protein and neuregulin 1, suggesting roles in neuron development, synaptic function, and neurodegeneration. We argue that their distinct effects on cargo endocytosis and recycling depend on interactions with vesicle trafficking and synaptic scaffolding proteins. As they play complementary, but opposing roles in cargo endocytosis, recycling, and degradation, balancing NEEP21 and Caly expression levels or activity could be important for homeostasis in a variety of signaling pathways, and also lead to a novel therapeutic strategy for disorders like Alzheimer's disease and schizophrenia. This review focuses on two closely related, neuron-enriched endosomal proteins: NEEP21 and Calcyon which perform distinct roles in regulating receptor endocytosis, recycling, and degradation. Based on an in-depth examination of the literature, we argue that these two proteins carry out complementary yet sometimes opposing vesicle trafficking functions that impact excitatory transmission, transcytosis, axonal transport, and also proteolytic processing by beta-secretase I (BACE1). Finally, we propose that balancing NEEP21 and Calcyon expression and/or activity could be important for homeostasis in a variety of signaling pathways, and also lead to a novel therapeutic strategy for disorders like Alzheimer's disease and schizophrenia. AMPA = α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; NMDA = N-Methyl-D-aspartate.
Collapse
Affiliation(s)
- Nagendran Muthusamy
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC
| | | | | | | | | |
Collapse
|
35
|
Maturational conversion of dendritic early endosomes and their roles in L1-mediated axon growth. J Neurosci 2015; 34:14633-43. [PMID: 25355216 DOI: 10.1523/jneurosci.1837-14.2014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The function of endosomes is intricately linked to cellular function in all cell types, including neurons. Intriguingly, neurons express cell type-specific proteins that localize to endosomes, but little is known about how these neuronal proteins interface with canonical endosomes and ubiquitously expressed endosomal components, such as EEA1 (Early Endosomal Antigen 1). NEEP21 (Neuronal Early Endosomal Protein 21 kDa) localizes to somatodendritic endosomes, and downregulation of NEEP21 perturbs the correct trafficking of multiple receptors, including glutamate receptors (GluA2) during LTP and amyloidogenic processing of βAPP. Our own work implicated NEEP21 in correct trafficking of the axonal cell adhesion molecule L1/neuron-glia cell adhesion molecule (NgCAM). NEEP21 dynamically localizes with EEA1-positive early endosomes but is also found in EEA1-negative endosomes. Live imaging reveals that NEEP21-positive, EEA1-negative endosomes arise as a consequence of maturational conversion of EEA1/NEEP21 double-positive endosomes. Interfering with EEA1 function causes missorting of L1/NgCAM, axon outgrowth defects on the L1 substrate, and disturbance of NEEP21 localization. Last, we uncover evidence that functional interference with NEEP21 reduces axon and dendrite growth of primary rat hippocampal neurons on L1 substrate but not on N-cadherin substrate, thus implicating endosomal trafficking through somatodendritic early endosomes in L1-mediated axon growth.
Collapse
|
36
|
Del Prete D, Lombino F, Liu X, D'Adamio L. APP is cleaved by Bace1 in pre-synaptic vesicles and establishes a pre-synaptic interactome, via its intracellular domain, with molecular complexes that regulate pre-synaptic vesicles functions. PLoS One 2014; 9:e108576. [PMID: 25247712 PMCID: PMC4172690 DOI: 10.1371/journal.pone.0108576] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 08/31/2014] [Indexed: 12/21/2022] Open
Abstract
Amyloid Precursor Protein (APP) is a type I membrane protein that undergoes extensive processing by secretases, including BACE1. Although mutations in APP and genes that regulate processing of APP, such as PSENs and BRI2/ITM2B, cause dementias, the normal function of APP in synaptic transmission, synaptic plasticity and memory formation is poorly understood. To grasp the biochemical mechanisms underlying the function of APP in the central nervous system, it is important to first define the sub-cellular localization of APP in synapses and the synaptic interactome of APP. Using biochemical and electron microscopy approaches, we have found that APP is localized in pre-synaptic vesicles, where it is processed by Bace1. By means of a proteomic approach, we have characterized the synaptic interactome of the APP intracellular domain. We focused on this region of APP because in vivo data underline the central functional and pathological role of the intracellular domain of APP. Consistent with the expression of APP in pre-synaptic vesicles, the synaptic APP intracellular domain interactome is predominantly constituted by pre-synaptic, rather than post-synaptic, proteins. This pre-synaptic interactome of the APP intracellular domain includes proteins expressed on pre-synaptic vesicles such as the vesicular SNARE Vamp2/Vamp1 and the Ca2+ sensors Synaptotagmin-1/Synaptotagmin-2, and non-vesicular pre-synaptic proteins that regulate exocytosis, endocytosis and recycling of pre-synaptic vesicles, such as target-membrane-SNAREs (Syntaxin-1b, Syntaxin-1a, Snap25 and Snap47), Munc-18, Nsf, α/β/γ-Snaps and complexin. These data are consistent with a functional role for APP, via its carboxyl-terminal domain, in exocytosis, endocytosis and/or recycling of pre-synaptic vesicles.
Collapse
Affiliation(s)
- Dolores Del Prete
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Franco Lombino
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Xinran Liu
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Luciano D'Adamio
- Department of Microbiology & Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
37
|
Laßek M, Weingarten J, Volknandt W. The synaptic proteome. Cell Tissue Res 2014; 359:255-65. [PMID: 25038742 DOI: 10.1007/s00441-014-1943-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 06/04/2014] [Indexed: 11/29/2022]
Abstract
Synapses are focal hot spots for signal transduction and plasticity in the brain. A synapse comprises an axon terminus, the presynapse, the synaptic cleft containing extracellular matrix proteins as well as adhesion molecules, and the postsynaptic density as target structure for chemical signaling. The proteomes of the presynaptic and postsynaptic active zones control neurotransmitter release and perception. These tasks demand short- and long-term structural and functional dynamics of the synapse mediated by its proteinaceous inventory. This review addresses subcellular fractionation protocols and the related proteomic approaches to the various synaptic subcompartments with an emphasis on the presynaptic active zone (PAZ). Furthermore, it discusses major constituents of the PAZ including the amyloid precursor protein family members. Numerous proteins regulating the rearrangement of the cytoskeleton are indicative of the functional and structural dynamics of the pre- and postsynapse. The identification of protein candidates of the synapse provides the basis for further analyzing the interaction of synaptic proteins with their targets, and the effect of their deletion opens novel insights into the functional role of these proteins in neuronal communication. The knowledge of the molecular interactome is also a prerequisite for understanding numerous neurodegenerative diseases.
Collapse
Affiliation(s)
- Melanie Laßek
- Molecular and Cellular Neurobiology, Goethe University, Frankfurt, Germany
| | | | | |
Collapse
|
38
|
Klevanski M, Saar M, Baumkötter F, Weyer SW, Kins S, Müller UC. Differential role of APP and APLPs for neuromuscular synaptic morphology and function. Mol Cell Neurosci 2014; 61:201-10. [PMID: 24998676 DOI: 10.1016/j.mcn.2014.06.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Revised: 06/02/2014] [Accepted: 06/05/2014] [Indexed: 11/26/2022] Open
Abstract
The analysis of mouse models indicated that APP and the related APLPs are important for synapse formation and function. The synaptic role of APP is, however, complex due to partially overlapping functions within the gene family. APP/APLPs are proteolytically cleaved and have both adhesive and signaling properties. Mice lacking individual APP family members are viable, whereas APP/APLP2 and APLP1/APLP2 double knockout (DKO) mice die shortly after birth. Here, we analyzed the morphology of the neuromuscular junction (NMJ) of lethal APLP1/APLP2-DKO mice in comparison to lethal APP/APLP2-DKO mutants and viable single KO mice. We report that, surprisingly, the NMJ phenotype of APLP1/APLP2-DKO mice shows striking differences as compared to APP/APLP2-DKO mice. Unexpectedly, APLP1/APLP2-DKO mice exhibit normal endplate patterning and lack presynaptic nerve terminal sprouting. However, at the level of individual synapses we show that APLP1/APLP2-DKO mice exhibit reduced size of pre- and postsynaptic compartments and reduced colocalization. As APP/APLP2-DKO and APLP1/APLP2-DKO mice show similar penetrance of early postnatal lethality, this suggests that deficits at the level of individual synapses due to impaired synaptic apposition and/or deficits in transmitter release may cause lethality. Using an in vitro cell-adhesion assay, we observed that APP trans-dimerization is considerably less efficient than APLP2 trans-interaction. Thus, differences between APP/APLP2 and APP/APLP1 NMJ formation may be in part explained by differences in APP/APLP2 trans-dimerization properties. Collectively, our study further highlights the distinct and essential role of APLP2 at NMJ synapses that cannot be compensated by APP.
Collapse
Affiliation(s)
- Maja Klevanski
- Department of Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Martina Saar
- Department of Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Frederik Baumkötter
- Department of Human Biology and Human Genetics, Technical University of Kaiserslautern, Erwin-Schrödinger-Strasse 13, 67663 Kaiserslautern, Germany
| | - Sascha W Weyer
- Department of Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Stefan Kins
- Department of Human Biology and Human Genetics, Technical University of Kaiserslautern, Erwin-Schrödinger-Strasse 13, 67663 Kaiserslautern, Germany
| | - Ulrike C Müller
- Department of Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany.
| |
Collapse
|
39
|
Abstract
Mislocalization of axonal proteins can result in misassembly and/or miswiring of neural circuits, causing disease. To date, only a handful of genes that control polarized localization of axonal membrane proteins have been identified. Here we report that Drosophila X11/Mint proteins are required for targeting several proteins, including human amyloid precursor protein (APP) and Drosophila APP-like protein (APPL), to axonal membranes and for their exclusion from dendrites of the mushroom body in Drosophila, a brain structure involved in learning and memory. Axonal localization of APP is mediated by an endocytic motif, and loss of X11/Mint results in a dramatic increase in cell-surface levels of APPL, especially on dendrites. Mutations in genes required for endocytosis show similar mislocalization of these proteins to dendrites, and strongly enhance defects seen in X11/Mint mutants. These results suggest that X11/Mint-dependent endocytosis in dendrites may serve to promote the axonal localization of membrane proteins. Since X11/Mint binds to APP, and abnormal trafficking of APP contributes to Alzheimer's disease, deregulation of X11/Mint may be important for Alzheimer's disease pathogenesis.
Collapse
|
40
|
Laßek M, Weingarten J, Einsfelder U, Brendel P, Müller U, Volknandt W. Amyloid precursor proteins are constituents of the presynaptic active zone. J Neurochem 2013; 127:48-56. [PMID: 23815291 DOI: 10.1111/jnc.12358] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Revised: 06/21/2013] [Accepted: 06/26/2013] [Indexed: 11/27/2022]
Abstract
The amyloid precursor protein (APP) and its mammalian homologs, APLP1, APLP2, have been allocated to an organellar pool residing in the Golgi apparatus and in endosomal compartments, and in its mature form to a cell surface-localized pool. In the brain, all APPs are restricted to neurons; however, their precise localization at the plasma membrane remained enigmatic. Employing a variety of subcellular fractionation steps, we isolated two synaptic vesicle (SV) pools from rat and mouse brain, a pool consisting of synaptic vesicles only and a pool comprising SV docked to the presynaptic plasma membrane. Immunopurification of these two pools using a monoclonal antibody directed against the 12 membrane span synaptic vesicle protein2 (SV2) demonstrated unambiguously that APP, APLP1 and APLP2 are constituents of the active zone of murine brain but essentially absent from free synaptic vesicles. The specificity of immunodetection was confirmed by analyzing the respective knock-out animals. The fractionation experiments further revealed that APP is accumulated in the fraction containing docked synaptic vesicles. These data present novel insights into the subsynaptic localization of APPs and are a prerequisite for unraveling the physiological role of all mature APP proteins in synaptic physiology.
Collapse
Affiliation(s)
- Melanie Laßek
- Department of Molecular and Cellular Neurobiology, Goethe-University, Frankfurt am Main, Germany
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Endocytosis and endosomal trafficking play a multitude of roles in cellular function beyond regulating entry of essential nutrients. In this review, we discuss the cell biological principles of endosomal trafficking, the neuronal adaptations to endosomal organization, and the role of endosomal trafficking in neural development. In particular, we consider how cell fate decisions, polarity, migration, and axon outgrowth and guidance are influenced by five endosomal tricks: dynamic modulation of receptor levels by endocytosis and recycling, cargo-specific responses via cargo-specific endocytic regulators, cell-type-specific endocytic regulation, ligand-specific endocytic regulation, and endosomal regulation of ligand processing and trafficking.
Collapse
Affiliation(s)
- Chan Choo Yap
- Department of Neuroscience, University of Virginia, 409 Lane Road, Charlottesville, VA 22908, USA
| | | |
Collapse
|
42
|
Volknandt W, Karas M. Proteomic analysis of the presynaptic active zone. Exp Brain Res 2012; 217:449-61. [DOI: 10.1007/s00221-012-3031-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Accepted: 02/04/2012] [Indexed: 02/06/2023]
|
43
|
Guo Q, Li H, Gaddam SSK, Justice NJ, Robertson CS, Zheng H. Amyloid precursor protein revisited: neuron-specific expression and highly stable nature of soluble derivatives. J Biol Chem 2011; 287:2437-45. [PMID: 22144675 DOI: 10.1074/jbc.m111.315051] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
APP processing and amyloid-β production play a central role in Alzheimer disease pathogenesis. APP has been considered a ubiquitously expressed protein. In addition to amyloid-β, α- or β-secretase-dependent cleavage of APP also generates soluble secreted APP (APPsα or APPsβ, respectively). Interestingly, APPsβ has been shown to be subject to further cleavage to create an N-APP fragment that binds to the DR6 death receptor and mediates axon pruning and degeneration under trophic factor withdrawal conditions. By performing APP immunocytochemical staining, we found that, unexpectedly, many antibodies yielded nonspecific staining in APP-null samples. Screening of a series of antibodies allowed us to identify a rabbit monoclonal antibody Y188 that is highly specific for APP and prompted us to re-examine the expression, localization, and stability of endogenous APP and APPsβ in wild-type and in APPsβ knock-in mice, respectively. In contrast to earlier studies, we found that APP is specifically expressed in neurons and that its expression cannot be detected in major types of glial cells under basal or neuroinflammatory conditions. Both APPsα and APPsβ are highly stable in the central nervous system (CNS) and do not undergo further cleavage with or without trophic factor support. Our results clarify several key questions with regard to the fundamental properties of APP and offer critical cellular insights into the pathophysiology of APP.
Collapse
Affiliation(s)
- Qinxi Guo
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | |
Collapse
|
44
|
Aydin D, Weyer SW, Müller UC. Functions of the APP gene family in the nervous system: insights from mouse models. Exp Brain Res 2011; 217:423-34. [PMID: 21931985 DOI: 10.1007/s00221-011-2861-2] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Accepted: 08/30/2011] [Indexed: 06/16/2024]
Abstract
The amyloid precursor protein (APP) plays a key role in the pathogenesis of Alzheimer's disease (AD), as proteolytical cleavage of APP gives rise to the β-amyloid peptide which is deposited in the brains of Alzheimer patients. During the past years, intense research efforts have been directed at elucidating the physiological function(s) of APP and the question of whether a perturbation of these functions contributes to AD pathogenesis. Indeed, a growing body of evidence has accumulated supporting a role of APP and the two closely related homologues APLP1 and APLP2 in various aspects of nervous system development and function, in particular, for synapse formation and function. This review summarizes recent insights into the in vivo role of the APP gene family from mice lacking individual or combinations of APP family members, with particular emphasis on recently generated knockin mice to examine the in vivo relevance of distinct functional domains.
Collapse
Affiliation(s)
- Dorothee Aydin
- Department of Bioinformatics and Functional Genomics, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | | | | |
Collapse
|
45
|
Abstract
The amyloid precursor protein (APP) has been under intensive study in recent years, mainly due to its critical role in the pathogenesis of Alzheimer's disease (AD). β-Amyloid (Aβ) peptides generated from APP proteolytic cleavage can aggregate, leading to plaque formation in human AD brains. Point mutations of APP affecting Aβ production are found to be causal for hereditary early onset familial AD. It is very likely that elucidating the physiological properties of APP will greatly facilitate the understanding of its role in AD pathogenesis. A number of APP loss- and gain-of-function models have been established in model organisms including Caenorhabditis elegans, Drosophila, zebrafish and mouse. These in vivo models provide us valuable insights into APP physiological functions. In addition, several knock-in mouse models expressing mutant APP at a physiological level are available to allow us to study AD pathogenesis without APP overexpression. This article will review the current physiological and pathophysiological animal models of APP.
Collapse
|
46
|
Lasiecka ZM, Winckler B. Mechanisms of polarized membrane trafficking in neurons -- focusing in on endosomes. Mol Cell Neurosci 2011; 48:278-87. [PMID: 21762782 DOI: 10.1016/j.mcn.2011.06.013] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 06/21/2011] [Accepted: 06/25/2011] [Indexed: 12/13/2022] Open
Abstract
Neurons are polarized cells that have a complex and unique morphology: long processes (axons and dendrites) extending far from the cell body. In addition, the somatodendritic and axonal domains are further divided into specific subdomains, such as synapses (pre- and postsynaptic specializations), proximal and distal dendrites, axon initial segments, nodes of Ranvier, and axon growth cones. The striking asymmetry and complexity of neuronal cells are necessary for their function in receiving, processing and transferring electrical signals, with each domain playing a precise function in these processes. In order to establish and maintain distinct neuronal domains, mechanisms must exist for protein delivery to specific neuronal compartments, such that each compartment has the correct functional molecular composition. How polarized membrane domains are established and maintained is a long-standing question. Transmembrane proteins, such as receptors and adhesion molecules, can be transported to their proper membrane domains by several pathways. The biosynthetic secretory system delivers newly synthesized transmembrane proteins from the ER via the Golgi and trans-Golgi-network (TGN) to the plasma membrane. In addition, the endosomal system is critically involved in many instances in ensuring proper (re)targeting of membrane components because it can internalize and degrade mislocalized proteins, or recycle proteins from one domain to another. The endosomal system is thus crucial for establishing and maintaining neuronal polarity. In this review, we focus mainly on the intracellular compartments that serve as sorting stations for polarized transport, with particular emphasis on the emerging roles of endosomes.
Collapse
Affiliation(s)
- Zofia M Lasiecka
- Department of Neuroscience, University of Virginia Medical School, 409 Lane Rd. Extension, MR4-6116, Charlottesville, VA 22908, USA
| | | |
Collapse
|
47
|
Winckler B, Yap CC. Endocytosis and endosomes at the crossroads of regulating trafficking of axon outgrowth-modifying receptors. Traffic 2011; 12:1099-108. [PMID: 21535338 DOI: 10.1111/j.1600-0854.2011.01213.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In neurons, many receptors must be localized correctly to axons or dendrites for proper function. During development, receptors for nerve growth and guidance are targeted to axons and localized to growth cones where receptor activation by ligands results in promotion or inhibition of axon growth. Signaling outcomes downstream of ligand binding are determined by the location, levels and residence times of receptors on the neuronal plasma membrane. Therefore, the mechanisms controlling the trafficking of these receptors are crucial to the proper wiring of circuits. Membrane proteins accumulate on the axonal surface by multiple routes, including polarized sorting in the trans Golgi network, sorting in endosomes and removal by endocytosis. Endosomes also play important roles in the signaling pathways for both growth-promoting and -inhibiting molecules: signaling endosomes derived from endocytosis are important for signaling from growth cones to cell bodies. Growth-promoting neurotrophins and growth-inhibiting Nogo-A can use EHD4/Pincher-dependent endocytosis at the growth cone for their respective retrograde signaling. In addition to retrograde transport of endosomes, anterograde transport to axons in endosomes also occurs for several receptors, including the axon outgrowth-promoting cell adhesion molecule L1/NgCAM and TrkA. L1/NgCAM also depends on EHD4/Pincher-dependent endocytosis for its axonal polarization. In this review, we will focus on receptors whose trafficking has been reported to be modulated by the EHD4/Pincher family of endosomal regulators, namely L1/NgCAM, Trk and Nogo-A. We will first summarize the pathways underlying the axonal transport of these proteins and then discuss the potential roles of EHD4/Pincher in mediating their endocytosis.
Collapse
Affiliation(s)
- Bettina Winckler
- Department of Neuroscience, University of Virginia Medical School, MR4-6115, 409 Lane Road Ext., Charlottesville, VA 22936, USA.
| | | |
Collapse
|
48
|
Zheng H, Koo EH. Biology and pathophysiology of the amyloid precursor protein. Mol Neurodegener 2011; 6:27. [PMID: 21527012 PMCID: PMC3098799 DOI: 10.1186/1750-1326-6-27] [Citation(s) in RCA: 221] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2011] [Accepted: 04/28/2011] [Indexed: 01/22/2023] Open
Abstract
The amyloid precursor protein (APP) plays a central role in the pathophysiology of Alzheimer's disease in large part due to the sequential proteolytic cleavages that result in the generation of β-amyloid peptides (Aβ). Not surprisingly, the biological properties of APP have also been the subject of great interest and intense investigations. Since our 2006 review, the body of literature on APP continues to expand, thereby offering further insights into the biochemical, cellular and functional properties of this interesting molecule. Sophisticated mouse models have been created to allow in vivo examination of cell type-specific functions of APP together with the many functional domains. This review provides an overview and update on our current understanding of the pathobiology of APP.
Collapse
Affiliation(s)
- Hui Zheng
- Huffington Center on Aging and Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| | | |
Collapse
|