1
|
Fu Y, Zhai Q. High-gamma frequency flash stimulation as a possible cognitive facilitator in rat pups. Brain Res 2025; 1848:149314. [PMID: 39549826 DOI: 10.1016/j.brainres.2024.149314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 10/27/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024]
Abstract
High-gamma frequency flashes can enhance cognition by synchronizing neural oscillations in mammals. Early flash treatment promotes the development of improved cognitive functions in young children. However, it is unclear whether exposure to high-gamma frequency flashes in preschool-aged individuals affects cognition in preadolescents by regulating neural oscillations in the brain. Here, we aimed to investigate the effects of gamma-frequency flashes on cognitive ability. In this study, the effect of high-frequency flicker on cognitive performance was verified by behavioural experiments such as the open-field test and the water maze, but also proteomics. We found that external 40 Hz and 70 Hz frequency flashes synchronized neural oscillations at the corresponding frequencies in the primary visual cortex (V1) of rats. Rats that underwent 70 Hz flash intervention had better cognitive behavioural performance in the early stages of training. The 70 Hz flash frequency upregulated proteins associated with neuronal growth and differentiation, such as Snapin, FoxO3, Hspa12a, and Penk, and activated the MAPK signalling pathway, signalling pathway regulating stem cell pluripotency, and the neuroactive ligand-receptor interaction pathway. These proteins and pathways play important roles in cognitive functions. Our study revealed that 70 Hz flashes received by young children early in their development substantially promote the growth of cognitive capabilities in the brain. Exposure to 70 Hz flashes may be a new intervention method and a new strategy for improving cognition.
Collapse
Affiliation(s)
- Yu Fu
- Kundulun Center for Disease Control and Prevention, Inner Mongolia 014010, China
| | - Qingfeng Zhai
- School of Public Health, Shandong Second Medical University, Shandong 261021, China.
| |
Collapse
|
2
|
Salazar CJ, Diaz-Balzac CA, Wang Y, Rahman M, Grant BD, Bülow HE. RABR-1, an atypical Rab-related GTPase, cell-nonautonomously restricts somatosensory dendrite branching. Genetics 2024; 228:iyae113. [PMID: 39028768 PMCID: PMC11457943 DOI: 10.1093/genetics/iyae113] [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: 11/15/2023] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
Neurons are highly polarized cells with dendrites and axons. Dendrites, which receive sensory information or input from other neurons, often display elaborately branched morphologies. While mechanisms that promote dendrite branching have been widely studied, less is known about the mechanisms that restrict branching. Using the nematode Caenorhabditis elegans, we identify rabr-1 (for Rab-related gene 1) as a factor that restricts branching of the elaborately branched dendritic trees of PVD and FLP somatosensory neurons. Animals mutant for rabr-1 show excessively branched dendrites throughout development and into adulthood in areas where the dendrites overlay epidermal tissues. Phylogenetic analyses show that RABR-1 displays similarity to small GTPases of the Rab-type, although based on sequence alone, no clear vertebrate ortholog of RABR-1 can be identified. We find that rabr-1 is expressed and can function in epidermal tissues, suggesting that rabr-1 restricts dendritic branching cell-nonautonomously. Genetic experiments further indicate that for the formation of ectopic branches rabr-1 mutants require the genes of the Menorin pathway, which have been previously shown to mediate dendrite morphogenesis of somatosensory neurons. A translational reporter for RABR-1 reveals a subcellular localization to punctate, perinuclear structures, which correlates with endosomal and autophagosomal markers, but anticorrelates with lysosomal markers suggesting an amphisomal character. Point mutations in rabr-1 analogous to key residues of small GTPases suggest that rabr-1 functions in a GTP-bound form independently of GTPase activity. Taken together, rabr-1 encodes for an atypical small GTPase of the Rab-type that cell-nonautonomously restricts dendritic branching of somatosensory neurons, likely independently of GTPase activity.
Collapse
Affiliation(s)
| | - Carlos A Diaz-Balzac
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yu Wang
- Department of Molecular Biology and Biochemistry, Rutgers Center for Lipid Research, Rutgers University, Piscataway, NJ 08854, USA
| | - Maisha Rahman
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Barth D Grant
- Department of Molecular Biology and Biochemistry, Rutgers Center for Lipid Research, Rutgers University, Piscataway, NJ 08854, USA
| | - Hannes E Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
3
|
Parisi B, Esposito A, Castroflorio E, Bramini M, Pepe S, Marte A, Guarnieri FC, Valtorta F, Baldelli P, Benfenati F, Fassio A, Giovedì S. Apache is a neuronal player in autophagy required for retrograde axonal transport of autophagosomes. Cell Mol Life Sci 2024; 81:416. [PMID: 39367928 PMCID: PMC11455771 DOI: 10.1007/s00018-024-05441-7] [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: 11/29/2023] [Revised: 08/19/2024] [Accepted: 08/29/2024] [Indexed: 10/07/2024]
Abstract
Neurons are dependent on efficient quality control mechanisms to maintain cellular homeostasis and function due to their polarization and long-life span. Autophagy is a lysosomal degradative pathway that provides nutrients during starvation and recycles damaged and/or aged proteins and organelles. In neurons, autophagosomes constitutively form in distal axons and at synapses and are trafficked retrogradely to the cell soma to fuse with lysosomes for cargo degradation. How the neuronal autophagy pathway is organized and controlled remains poorly understood. Several presynaptic endocytic proteins have been shown to regulate both synaptic vesicle recycling and autophagy. Here, by combining electron, fluorescence, and live imaging microscopy with biochemical analysis, we show that the neuron-specific protein APache, a presynaptic AP-2 interactor, functions in neurons as an important player in the autophagy process, regulating the retrograde transport of autophagosomes. We found that APache colocalizes and co-traffics with autophagosomes in primary cortical neurons and that induction of autophagy by mTOR inhibition increases LC3 and APache protein levels at synaptic boutons. APache silencing causes a blockade of autophagic flux preventing the clearance of p62/SQSTM1, leading to a severe accumulation of autophagosomes and amphisomes at synaptic terminals and along neurites due to defective retrograde transport of TrkB-containing signaling amphisomes along the axons. Together, our data identify APache as a regulator of the autophagic cycle, potentially in cooperation with AP-2, and hypothesize that its dysfunctions contribute to the early synaptic impairments in neurodegenerative conditions associated with impaired autophagy.
Collapse
Affiliation(s)
- Barbara Parisi
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italia
- Present Affiliation: Department of Cell Biology, Universidad de Granada, Granada, Spain
| | - Alessandro Esposito
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italia
- IRCSS, Ospedale Policlinico San Martino, Viale Benedetto XV, 3, Genova, 16122, Italy
| | - Enrico Castroflorio
- Center for Synaptic Neuroscience and Technology, Italian Institute of Technology, Genoa, Italy
| | - Mattia Bramini
- Center for Synaptic Neuroscience and Technology, Italian Institute of Technology, Genoa, Italy
- Present Affiliation: Institute of Neuroscience, National Research Council (CNR), Vedano al Lambro, Italy
| | - Sara Pepe
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italia
- IRCCS, Ospedale Policlinico San Martino, Genova, Italia
| | - Antonella Marte
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italia
- IRCCS, Ospedale Policlinico San Martino, Genova, Italia
| | - Fabrizia C Guarnieri
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- IRCSS, Ospedale Policlinico San Martino, Viale Benedetto XV, 3, Genova, 16122, Italy
| | - Flavia Valtorta
- Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Pietro Baldelli
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italia
- IRCCS, Ospedale Policlinico San Martino, Genova, Italia
| | - Fabio Benfenati
- Center for Synaptic Neuroscience and Technology, Italian Institute of Technology, Genoa, Italy
- IRCCS, Ospedale Policlinico San Martino, Genova, Italia
| | - Anna Fassio
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italia
- IRCCS, Ospedale Policlinico San Martino, Genova, Italia
| | - Silvia Giovedì
- Dipartimento di Medicina Sperimentale, Università degli Studi di Genova, Genova, Italia.
- IRCCS, Ospedale Policlinico San Martino, Genova, Italia.
- Department of Experimental Medicine, University of Genoa, Viale Benedetto XV, 3, Genova, 16122, Italy.
| |
Collapse
|
4
|
Wisner SR, Chlebowski M, Mandal A, Mai D, Stein C, Petralia RS, Wang YX, Drerup CM. An initial HOPS-mediated fusion event is critical for autophagosome transport initiation from the axon terminal. Autophagy 2024; 20:2275-2296. [PMID: 38899385 PMCID: PMC11423661 DOI: 10.1080/15548627.2024.2366122] [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: 12/03/2023] [Revised: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024] Open
Abstract
In neurons, macroautophagy/autophagy is a frequent and critical process. In the axon, autophagy begins in the axon terminal, where most nascent autophagosomes form. After formation, autophagosomes must initiate transport to exit the axon terminal and move toward the cell body via retrograde transport. During retrograde transport these autophagosomes mature through repetitive fusion events. Complete lysosomal cargo degradation occurs largely in the cell body. The precipitating events to stimulate retrograde autophagosome transport have been debated but their importance is clear: disrupting neuronal autophagy or autophagosome transport is detrimental to neuronal health and function. We have identified the HOPS complex as essential for early autophagosome maturation and consequent initiation of retrograde transport from the axon terminal. In yeast and mammalian cells, HOPS controls fusion between autophagosomes and late endosomes with lysosomes. Using zebrafish strains with loss-of-function mutations in vps18 and vps41, core components of the HOPS complex, we found that disruption of HOPS eliminates autophagosome maturation and disrupts retrograde autophagosome transport initiation from the axon terminal. We confirmed this phenotype was due to loss of HOPS complex formation using an endogenous deletion of the HOPS binding domain in Vps18. Finally, using pharmacological inhibition of lysosomal proteases, we show that initiation of autophagosome retrograde transport requires autophagosome maturation. Together, our data demonstrate that HOPS-mediated fusion events are critical for retrograde autophagosome transport initiation through promoting autophagosome maturation. This reveals critical roles for the HOPS complex in neuronal autophagy which deepens our understanding of the cellular pathology of HOPS-complex linked neurodegenerative diseases.Abbreviations: CORVET: Class C core vacuole/endosome tethering; gRNA: guide RNA; HOPS: homotypic fusion and protein sorting; pLL: posterior lateral line; Vps18: VPS18 core subunit of CORVET and HOPS complexes; Vps41: VPS41 subunit of HOPS complex.
Collapse
Affiliation(s)
- Serena R. Wisner
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Madison Chlebowski
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Amrita Mandal
- National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Don Mai
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Chris Stein
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Ronald S. Petralia
- Advanced Imaging Core, National Institute of Deafness and Other Communication Disorders, NIH, Bethesda, MD, USA
| | - Ya-Xian Wang
- Advanced Imaging Core, National Institute of Deafness and Other Communication Disorders, NIH, Bethesda, MD, USA
| | - Catherine M. Drerup
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
5
|
Li J, Huang X, An Y, Chen X, Chen Y, Xu M, Shan H, Zhang M. The role of snapin in regulation of brain homeostasis. Neural Regen Res 2024; 19:1696-1701. [PMID: 38103234 PMCID: PMC10960280 DOI: 10.4103/1673-5374.389364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 09/15/2023] [Accepted: 10/08/2023] [Indexed: 12/18/2023] Open
Abstract
Brain homeostasis refers to the normal working state of the brain in a certain period, which is important for overall health and normal life activities. Currently, there is a lack of effective treatment methods for the adverse consequences caused by brain homeostasis imbalance. Snapin is a protein that assists in the formation of neuronal synapses and plays a crucial role in the normal growth and development of synapses. Recently, many researchers have reported the association between snapin and neurologic and psychiatric disorders, demonstrating that snapin can improve brain homeostasis. Clinical manifestations of brain disease often involve imbalances in brain homeostasis and may lead to neurological and behavioral sequelae. This article aims to explore the role of snapin in restoring brain homeostasis after injury or diseases, highlighting its significance in maintaining brain homeostasis and treating brain diseases. Additionally, it comprehensively discusses the implications of snapin in other extracerebral diseases such as diabetes and viral infections, with the objective of determining the clinical potential of snapin in maintaining brain homeostasis.
Collapse
Affiliation(s)
- Jiawen Li
- Shanghai Key Lab of Forensic Medicine, Key Lab of Forensic Science, Ministry of Justice, China (Academy of Forensic Science), Shanghai, China
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Xinqi Huang
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Yumei An
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Xueshi Chen
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Yiyang Chen
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Mingyuan Xu
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| | - Haiyan Shan
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu Province, China
| | - Mingyang Zhang
- Shanghai Key Lab of Forensic Medicine, Key Lab of Forensic Science, Ministry of Justice, China (Academy of Forensic Science), Shanghai, China
- Institute of Forensic Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
6
|
Xiong GJ, Sheng ZH. Presynaptic perspective: Axonal transport defects in neurodevelopmental disorders. J Cell Biol 2024; 223:e202401145. [PMID: 38568173 PMCID: PMC10988239 DOI: 10.1083/jcb.202401145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/05/2024] Open
Abstract
Disruption of synapse assembly and maturation leads to a broad spectrum of neurodevelopmental disorders. Presynaptic proteins are largely synthesized in the soma, where they are packaged into precursor vesicles and transported into distal axons to ensure precise assembly and maintenance of presynapses. Due to their morphological features, neurons face challenges in the delivery of presynaptic cargos to nascent boutons. Thus, targeted axonal transport is vital to build functional synapses. A growing number of mutations in genes encoding the transport machinery have been linked to neurodevelopmental disorders. Emerging lines of evidence have started to uncover presynaptic mechanisms underlying axonal transport defects, thus broadening the view of neurodevelopmental disorders beyond postsynaptic mechanisms. In this review, we discuss presynaptic perspectives of neurodevelopmental disorders by focusing on impaired axonal transport and disturbed assembly and maintenance of presynapses. We also discuss potential strategies for restoring axonal transport as an early therapeutic intervention.
Collapse
Affiliation(s)
- Gui-Jing Xiong
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
7
|
Moya-Alvarado G, Valero-Peña X, Aguirre-Soto A, Bustos FJ, Lazo OM, Bronfman FC. PLC-γ-Ca 2+ pathway regulates axonal TrkB endocytosis and is required for long-distance propagation of BDNF signaling. Front Mol Neurosci 2024; 17:1009404. [PMID: 38660384 PMCID: PMC11040097 DOI: 10.3389/fnmol.2024.1009404] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its tropomyosin receptor kinase B (TrkB) are important signaling proteins that regulate dendritic growth and maintenance in the central nervous system (CNS). After binding of BDNF, TrkB is endocytosed into endosomes and continues signaling within the cell soma, dendrites, and axon. In previous studies, we showed that BDNF signaling initiated in axons triggers long-distance signaling, inducing dendritic arborization in a CREB-dependent manner in cell bodies, processes that depend on axonal dynein and TrkB activities. The binding of BDNF to TrkB triggers the activation of different signaling pathways, including the ERK, PLC-γ and PI3K-mTOR pathways, to induce dendritic growth and synaptic plasticity. How TrkB downstream pathways regulate long-distance signaling is unclear. Here, we studied the role of PLC-γ-Ca2+ in BDNF-induced long-distance signaling using compartmentalized microfluidic cultures. We found that dendritic branching and CREB phosphorylation induced by axonal BDNF stimulation require the activation of PLC-γ in the axons of cortical neurons. Locally, in axons, BDNF increases PLC-γ phosphorylation and induces intracellular Ca2+ waves in a PLC-γ-dependent manner. In parallel, we observed that BDNF-containing signaling endosomes transport to the cell body was dependent on PLC-γ activity and intracellular Ca2+ stores. Furthermore, the activity of PLC-γ is required for BDNF-dependent TrkB endocytosis, suggesting a role for the TrkB/PLC-γ signaling pathway in axonal signaling endosome formation.
Collapse
Affiliation(s)
- Guillermo Moya-Alvarado
- Faculty of Biological Sciences, Pontificia Universidad Catolica de Chile (UC), Santiago, Chile
| | - Xavier Valero-Peña
- NeuroSignaling Laboratory, Institute of Biomedical Sciences (ICB), Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Alejandro Aguirre-Soto
- NeuroSignaling Laboratory, Institute of Biomedical Sciences (ICB), Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Fernando J. Bustos
- Constantin-Paton Research Laboratory, Institute of Biomedical Sciences (ICB), Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Oscar M. Lazo
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Francisca C. Bronfman
- NeuroSignaling Laboratory, Institute of Biomedical Sciences (ICB), Faculty of Medicine and Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
8
|
Nambiar A, Manjithaya R. Driving autophagy - the role of molecular motors. J Cell Sci 2024; 137:jcs260481. [PMID: 38329417 DOI: 10.1242/jcs.260481] [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] [Indexed: 02/09/2024] Open
Abstract
Most of the vesicular transport pathways inside the cell are facilitated by molecular motors that move along cytoskeletal networks. Autophagy is a well-explored catabolic pathway that is initiated by the formation of an isolation membrane known as the phagophore, which expands to form a double-membraned structure that captures its cargo and eventually moves towards the lysosomes for fusion. Molecular motors and cytoskeletal elements have been suggested to participate at different stages of the process as the autophagic vesicles move along cytoskeletal tracks. Dynein and kinesins govern autophagosome trafficking on microtubules through the sequential recruitment of their effector proteins, post-translational modifications and interactions with LC3-interacting regions (LIRs). In contrast, myosins are actin-based motors that participate in various stages of the autophagic flux, as well as in selective autophagy pathways. However, several outstanding questions remain with regard to how the dominance of a particular motor protein over another is controlled, and to the molecular mechanisms that underlie specific disease variants in motor proteins. In this Review, we aim to provide an overview of the role of molecular motors in autophagic flux, as well as highlight their dysregulation in diseases, such as neurodegenerative disorders and pathogenic infections, and ageing.
Collapse
Affiliation(s)
- Akshaya Nambiar
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
| | - Ravi Manjithaya
- Autophagy Laboratory, Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
| |
Collapse
|
9
|
Kuo HY, Chen SY, Huang RC, Takahashi H, Lee YH, Pang HY, Wu CH, Graybiel AM, Liu FC. Speech- and language-linked FOXP2 mutation targets protein motors in striatal neurons. Brain 2023; 146:3542-3557. [PMID: 37137515 PMCID: PMC10393416 DOI: 10.1093/brain/awad090] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/13/2023] [Accepted: 02/02/2023] [Indexed: 05/05/2023] Open
Abstract
Human speech and language are among the most complex motor and cognitive abilities. The discovery of a mutation in the transcription factor FOXP2 in KE family members with speech disturbances has been a landmark example of the genetic control of vocal communication in humans. Cellular mechanisms underlying this control have remained unclear. By leveraging FOXP2 mutation/deletion mouse models, we found that the KE family FOXP2R553H mutation directly disables intracellular dynein-dynactin 'protein motors' in the striatum by induction of a disruptive high level of dynactin1 that impairs TrkB endosome trafficking, microtubule dynamics, dendritic outgrowth and electrophysiological activity in striatal neurons alongside vocalization deficits. Dynactin1 knockdown in mice carrying FOXP2R553H mutations rescued these cellular abnormalities and improved vocalization. We suggest that FOXP2 controls vocal circuit formation by regulating protein motor homeostasis in striatal neurons, and that its disruption could contribute to the pathophysiology of FOXP2 mutation/deletion-associated speech disorders.
Collapse
Affiliation(s)
- Hsiao-Ying Kuo
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Anatomy and Cell Biology, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Shih-Yun Chen
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Rui-Chi Huang
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Hiroshi Takahashi
- Department of Neurology, National Hospital Organization, Tottori Medical Center, Tottori 689-0203, Japan
| | - Yen-Hui Lee
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Hao-Yu Pang
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Cheng-Hsi Wu
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Ann M Graybiel
- McGovern Institute for Brain Research and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Fu-Chin Liu
- Institute of Neuroscience, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| |
Collapse
|
10
|
Fogarty MJ, Dasgupta D, Khurram OU, Sieck GC. Chemogenetic inhibition of TrkB signalling reduces phrenic motor neuron survival and size. Mol Cell Neurosci 2023; 125:103847. [PMID: 36958643 PMCID: PMC10247511 DOI: 10.1016/j.mcn.2023.103847] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 03/07/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023] Open
Abstract
Brain derived neurotrophic factor (BDNF) signalling through its high-affinity tropomyosin receptor kinase B (TrkB) is known to have potent effects on motor neuron survival and morphology during development and in neurodegenerative diseases. Here, we employed a novel 1NMPP1 sensitive TrkBF616 rat model to evaluate the effect of 14 days inhibition of TrkB signalling on phrenic motor neurons (PhMNs). Adult female and male TrkBF616 rats were divided into 1NMPP1 or vehicle treated groups. Three days prior to treatment, PhMNs in both groups were initially labeled via intrapleural injection of Alexa-Fluor-647 cholera toxin B (CTB). After 11 days of treatment, retrograde axonal uptake/transport was assessed by secondary labeling of PhMNs by intrapleural injection of Alexa-Fluor-488 CTB. After 14 days of treatment, the spinal cord was excised 100 μm thick spinal sections containing PhMNs were imaged using two-channel confocal microscopy. TrkB inhibition reduced the total number of PhMNs by ∼16 %, reduced the mean PhMN somal surface areas by ∼25 %, impaired CTB uptake 2.5-fold and reduced the estimated PhMN dendritic surface area by ∼38 %. We conclude that inhibition of TrkB signalling alone in adult TrkBF616 rats is sufficient to lead to PhMN loss, morphological degeneration and deficits in retrograde axonal uptake/transport.
Collapse
Affiliation(s)
- Matthew J Fogarty
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Debanjali Dasgupta
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Obaid U Khurram
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA
| | - Gary C Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
11
|
Lazo OM, Schiavo G. Rab10 regulates the sorting of internalised TrkB for retrograde axonal transport. eLife 2023; 12:81532. [PMID: 36897066 PMCID: PMC10005780 DOI: 10.7554/elife.81532] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 02/15/2023] [Indexed: 03/11/2023] Open
Abstract
Neurons process real-time information from axon terminals to coordinate gene expression, growth, and plasticity. Inputs from distal axons are encoded as a stream of endocytic organelles, termed signalling endosomes, targeted to the soma. Formation of these organelles depends on target-derived molecules, such as brain-derived neurotrophic factor (BDNF), which is recognised by TrkB receptors on the plasma membrane, endocytosed, and transported to the cell body along the microtubules network. Notwithstanding its physiological and neuropathological importance, the mechanism controlling the sorting of TrkB to signalling endosomes is currently unknown. In this work, we use primary mouse neurons to uncover the small GTPase Rab10 as critical for TrkB sorting and propagation of BDNF signalling from axon terminals to the soma. Our data demonstrate that Rab10 defines a novel membrane compartment that is rapidly mobilised towards the axon terminal upon BDNF stimulation, enabling the axon to fine-tune retrograde signalling depending on BDNF availability at the synapse. These results help clarifying the neuroprotective phenotype recently associated to Rab10 polymorphisms in Alzheimer's disease and provide a new therapeutic target to halt neurodegeneration.
Collapse
Affiliation(s)
- Oscar Marcelo Lazo
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, University College LondonLondonUnited Kingdom
- UK Dementia Research Institute at UCLLondonUnited Kingdom
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases and UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, University College LondonLondonUnited Kingdom
- UK Dementia Research Institute at UCLLondonUnited Kingdom
| |
Collapse
|
12
|
Moya-Alvarado G, Tiburcio-Felix R, Ibáñez MR, Aguirre-Soto AA, Guerra MV, Wu C, Mobley WC, Perlson E, Bronfman FC. BDNF/TrkB signaling endosomes in axons coordinate CREB/mTOR activation and protein synthesis in the cell body to induce dendritic growth in cortical neurons. eLife 2023; 12:77455. [PMID: 36826992 PMCID: PMC9977295 DOI: 10.7554/elife.77455] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/12/2023] [Indexed: 02/25/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its receptors tropomyosin kinase receptor B (TrkB) and the p75 neurotrophin receptor (p75) are the primary regulators of dendritic growth in the CNS. After being bound by BDNF, TrkB and p75 are endocytosed into endosomes and continue signaling within the cell soma, dendrites, and axons. We studied the functional role of BDNF axonal signaling in cortical neurons derived from different transgenic mice using compartmentalized cultures in microfluidic devices. We found that axonal BDNF increased dendritic growth from the neuronal cell body in a cAMP response element-binding protein (CREB)-dependent manner. These effects were dependent on axonal TrkB but not p75 activity. Dynein-dependent BDNF-TrkB-containing endosome transport was required for long-distance induction of dendritic growth. Axonal signaling endosomes increased CREB and mTOR kinase activity in the cell body, and this increase in the activity of both proteins was required for general protein translation and the expression of Arc, a plasticity-associated gene, indicating a role for BDNF-TrkB axonal signaling endosomes in coordinating the transcription and translation of genes whose products contribute to learning and memory regulation.
Collapse
Affiliation(s)
- Guillermo Moya-Alvarado
- Department of Physiology, Faculty of Biological Sciences and Center for Aging and Regeneration), Pontificia Universidad Católica de Chile. Av. Libertador Bernardo O´HigginsSantiagoChile
| | - Reynaldo Tiburcio-Felix
- NeuroSignaling Lab (NESLab), Center for Aging and Regeneration (CARE-UC), Institute of Biomedical Sciences (ICB), Faculty of Medicine, and Faculty of Life Sciences, Universidad Andrés BelloSantiagoChile
| | - María Raquel Ibáñez
- NeuroSignaling Lab (NESLab), Center for Aging and Regeneration (CARE-UC), Institute of Biomedical Sciences (ICB), Faculty of Medicine, and Faculty of Life Sciences, Universidad Andrés BelloSantiagoChile
| | - Alejandro A Aguirre-Soto
- NeuroSignaling Lab (NESLab), Center for Aging and Regeneration (CARE-UC), Institute of Biomedical Sciences (ICB), Faculty of Medicine, and Faculty of Life Sciences, Universidad Andrés BelloSantiagoChile
| | - Miguel V Guerra
- Department of Physiology, Faculty of Biological Sciences and Center for Aging and Regeneration), Pontificia Universidad Católica de Chile. Av. Libertador Bernardo O´HigginsSantiagoChile,NeuroSignaling Lab (NESLab), Center for Aging and Regeneration (CARE-UC), Institute of Biomedical Sciences (ICB), Faculty of Medicine, and Faculty of Life Sciences, Universidad Andrés BelloSantiagoChile
| | - Chengbiao Wu
- Department of Neurosciences, University of California, San DiegoSan DiegoUnited States
| | - William C Mobley
- Department of Neurosciences, University of California, San DiegoSan DiegoUnited States
| | - Eran Perlson
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine; Sagol School of Neuroscience, Tel Aviv UniversityTel AvivIsrael
| | - Francisca C Bronfman
- NeuroSignaling Lab (NESLab), Center for Aging and Regeneration (CARE-UC), Institute of Biomedical Sciences (ICB), Faculty of Medicine, and Faculty of Life Sciences, Universidad Andrés BelloSantiagoChile
| |
Collapse
|
13
|
Fu X, Rao L, Li P, Liu X, Wang Q, Son AI, Gennerich A, Liu JSH. Doublecortin and JIP3 are neural-specific counteracting regulators of dynein-mediated retrograde trafficking. eLife 2022; 11:e82218. [PMID: 36476638 PMCID: PMC9799976 DOI: 10.7554/elife.82218] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 12/06/2022] [Indexed: 12/12/2022] Open
Abstract
Mutations in the microtubule (MT)-binding protein doublecortin (DCX) or in the MT-based molecular motor dynein result in lissencephaly. However, a functional link between DCX and dynein has not been defined. Here, we demonstrate that DCX negatively regulates dynein-mediated retrograde transport in neurons from Dcx-/y or Dcx-/y;Dclk1-/- mice by reducing dynein's association with MTs and disrupting the composition of the dynein motor complex. Previous work showed an increased binding of the adaptor protein C-Jun-amino-terminal kinase-interacting protein 3 (JIP3) to dynein in the absence of DCX. Using purified components, we demonstrate that JIP3 forms an active motor complex with dynein and its cofactor dynactin with two dyneins per complex. DCX competes with the binding of the second dynein, resulting in a velocity reduction of the complex. We conclude that DCX negatively regulates dynein-mediated retrograde transport through two critical interactions by regulating dynein binding to MTs and regulating the composition of the dynein motor complex.
Collapse
Affiliation(s)
- Xiaoqin Fu
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical UniversityWenzhouChina
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhouChina
- Key Laboratory of Perinatal Medicine of WenzhouWenzhouChina
| | - Lu Rao
- Department of Biochemistry and Gruss-Lipper Biophotonics Center, Albert Einstein College of MedicineBronxUnited States
| | - Peijun Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical UniversityWenzhouChina
- Key Laboratory of Structural Malformations in Children of Zhejiang ProvinceWenzhouChina
- Key Laboratory of Perinatal Medicine of WenzhouWenzhouChina
| | - Xinglei Liu
- Department of Biochemistry and Gruss-Lipper Biophotonics Center, Albert Einstein College of MedicineBronxUnited States
| | - Qi Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical UniversityWenzhouChina
| | - Alexander I Son
- Center for Neuroscience Research, Children's National Research Institute, Children's National HospitalWashingtonUnited States
| | - Arne Gennerich
- Department of Biochemistry and Gruss-Lipper Biophotonics Center, Albert Einstein College of MedicineBronxUnited States
| | - Judy Shih-Hwa Liu
- Department of Neurology, Department of Molecular Biology, Cell Biology, and Biochemistry, Brown UniversityProvidenceUnited States
| |
Collapse
|
14
|
Huang N, Sheng ZH. Microfluidic devices as model platforms of CNS injury-ischemia to study axonal regeneration by regulating mitochondrial transport and bioenergetic metabolism. CELL REGENERATION 2022; 11:33. [PMID: 36184647 PMCID: PMC9527262 DOI: 10.1186/s13619-022-00138-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/28/2022] [Indexed: 11/10/2022]
Abstract
Central nervous system (CNS) neurons typically fail to regenerate their axons after injury leading to neurological impairment. Axonal regeneration is a highly energy-demanding cellular program that requires local mitochondria to supply most energy within injured axons. Recent emerging lines of evidence have started to reveal that injury-triggered acute mitochondrial damage and local energy crisis contribute to the intrinsic energetic restriction that accounts for axon regeneration failure in the CNS. Characterizing and reprogramming bioenergetic signaling and mitochondrial maintenance after axon injury-ischemia is fundamental for developing therapeutic strategies that can restore local energy metabolism and thus facilitate axon regeneration. Therefore, establishing reliable and reproducible neuronal model platforms is critical for assessing axonal energetic metabolism and regeneration capacity after injury-ischemia. In this focused methodology article, we discuss recent advances in applying cutting-edge microfluidic chamber devices in combination with state-of-the-art live-neuron imaging tools to monitor axonal regeneration, mitochondrial transport, bioenergetic metabolism, and local protein synthesis in response to injury-ischemic stress in mature CNS neurons.
Collapse
Affiliation(s)
- Ning Huang
- grid.94365.3d0000 0001 2297 5165Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706 USA ,grid.43169.390000 0001 0599 1243Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, 710061 Shaanxi China ,grid.43169.390000 0001 0599 1243Institute of Neuroscience, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, 710061 Shaanxi China
| | - Zu-Hang Sheng
- grid.94365.3d0000 0001 2297 5165Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706 USA
| |
Collapse
|
15
|
Tosolini AP, Sleigh JN, Surana S, Rhymes ER, Cahalan SD, Schiavo G. BDNF-dependent modulation of axonal transport is selectively impaired in ALS. Acta Neuropathol Commun 2022; 10:121. [PMID: 35996201 PMCID: PMC9396851 DOI: 10.1186/s40478-022-01418-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 07/29/2022] [Indexed: 02/08/2023] Open
Abstract
Axonal transport ensures long-range delivery of essential cargoes between proximal and distal compartments, and is needed for neuronal development, function, and survival. Deficits in axonal transport have been detected at pre-symptomatic stages in the SOD1G93A and TDP-43M337V mouse models of amyotrophic lateral sclerosis (ALS), suggesting that impairments in this critical process are fundamental for disease pathogenesis. Strikingly, in ALS, fast motor neurons (FMNs) degenerate first whereas slow motor neurons (SMNs) are more resistant, and this is a currently unexplained phenomenon. The main aim of this investigation was to determine the effects of brain-derived neurotrophic factor (BDNF) on in vivo axonal transport in different α-motor neuron (MN) subtypes in wild-type (WT) and SOD1G93A mice. We report that despite displaying similar basal transport speeds, stimulation of wild-type MNs with BDNF enhances in vivo trafficking of signalling endosomes specifically in FMNs. This BDNF-mediated enhancement of transport was also observed in primary ventral horn neuronal cultures. However, FMNs display selective impairment of axonal transport in vivo in symptomatic SOD1G93A mice, and are refractory to BDNF stimulation, a phenotype that was also observed in primary embryonic SOD1G93A neurons. Furthermore, symptomatic SOD1G93A mice display upregulation of the classical non-pro-survival truncated TrkB and p75NTR receptors in muscles, sciatic nerves, and Schwann cells. Altogether, these data indicate that cell- and non-cell autonomous BDNF signalling is impaired in SOD1G93A MNs, thus identifying a new key deficit in ALS.
Collapse
Affiliation(s)
- Andrew P Tosolini
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK.
| | - James N Sleigh
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - Sunaina Surana
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - Elena R Rhymes
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK
| | - Stephen D Cahalan
- Comparative Neuromuscular Disease Laboratory, Department of Clinical Sciences and Services, Royal Veterinary College, University of London, London, NW1 0TU, UK
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, WC1N 3BG, UK.
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, WC1N 3BG, UK.
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK.
| |
Collapse
|
16
|
Red Raspberry Extract Decreases Depression-Like Behavior in Rats by Modulating Neuroinflammation and Oxidative Stress. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9943598. [PMID: 35818443 PMCID: PMC9270999 DOI: 10.1155/2022/9943598] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/23/2022] [Accepted: 05/27/2022] [Indexed: 11/28/2022]
Abstract
Objective Red raspberry serves as a proven natural product to produce anti-inflammatory, antioxidant, and anticancer functions, but limited findings are available on its effects on depression. This study, by using a chronic unpredictable mild stress- (CUMS-) induced depression model, thus investigated the effects and underlying mechanism of red raspberry extract (RRE) on depressive behavior, inflammation, and oxidative stress. Methods Different treatments were given after random grouping of Sprague-Dawley rats, including no intervention (control), CUMS induction, and CUMS+different concentrations of RRE, and subsequently, depression-like behavior tests were performed. HE staining was designed to observe the pathological damage of the hippocampal tissue in rats. The levels of oxidative stress, endocrine hormones, and inflammatory factors were determined by biochemical assay and ELISA, and gene expression (mRNA and protein) in the hippocampal tissue by qRT-PCR and Western blot. Results On completion of CUMS treatment, the rats showed severe depression-like behavior, with obvious hippocampal tissue damage, oxidative inflammatory response, and endocrine imbalance. Importantly, RRE treatment significantly improved such depression-like behavior and attenuated histopathological damage in CUMS rats when reducing inflammation and oxidative stress and endocrine imbalance with upregulation of glutathione (GSH), superoxide dismutase (SOD), and interleukin- (IL-) 10 and downregulation of adrenocorticotropic hormone (ACTH), corticosterone (CORT), malondialdehyde (MDA), IL-1β, cyclooxygenase- (COX-) 2, and human macrophage chemoattractant protein- (MCP-) 1. In addition, for CUMS rats, RRE was a contributor to increasingly expressed brain-derived neurotrophic factor (BDNF), neurotrophic tyrosine receptor kinase 2 (TrkB), and p-mTOR but inhibited p-GSK-3β expression in the hippocampal tissue. All the above antidepressant effects of RRE were concentration-dependent. Conclusion By regulating neuroinflammation, oxidative stress response, endocrine level, and BDNF/TrkB level, RRE showed potential efficacy in alleviating depression-like behavior and histopathological damage of hippocampal tissue in CUMS rats by regulating the GSK3β and mTOR signaling pathways.
Collapse
|
17
|
Abdollahi M, Fahnestock M. Nurr1 Is Not an Essential Regulator of BDNF in Mouse Cortical Neurons. Int J Mol Sci 2022; 23:6853. [PMID: 35743300 PMCID: PMC9224520 DOI: 10.3390/ijms23126853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 12/30/2022] Open
Abstract
Nurr1 and brain-derived neurotrophic factor (BDNF) play major roles in cognition. Nurr1 regulates BDNF in midbrain dopaminergic neurons and cerebellar granule cells. Nurr1 and BDNF are also highly expressed in the cerebral cortex, a brain area important in cognition. Due to Nurr1 and BDNF tissue specificity, the regulatory effect of Nurr1 on BDNF in different brain areas cannot be generalized. The relationship between Nurr1 and BDNF in the cortex has not been investigated previously. Therefore, we examined Nurr1-mediated BDNF regulation in cortical neurons in activity-dependent and activity-independent states. Mouse primary cortical neurons were treated with the Nurr1 agonist, amodiaquine (AQ). Membrane depolarization was induced by KCl or veratridine and reversed by nimodipine. AQ and membrane depolarization significantly increased Nurr1 (p < 0.001) and BDNF (pAQ < 0.001, pKCl < 0.01) as assessed by real-time qRT-PCR. However, Nurr1 knockdown did not affect BDNF gene expression in resting or depolarized neurons. Accordingly, the positive correlation between Nurr1 and BDNF expression in AQ and membrane depolarization experiments does not imply co-regulation because Nurr1 knockdown did not affect BDNF gene expression in resting or depolarized cortical neurons. Therefore, in contrast to midbrain dopaminergic neurons and cerebellar granule cells, Nurr1 does not regulate BDNF in cortical neurons.
Collapse
Affiliation(s)
- Mona Abdollahi
- Medical Sciences Graduate Program, Faculty of Health Sciences, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada;
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioral Neurosciences, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
18
|
The Rab11-regulated endocytic pathway and BDNF/TrkB signaling: Roles in plasticity changes and neurodegenerative diseases. Neurobiol Dis 2022; 171:105796. [PMID: 35728773 DOI: 10.1016/j.nbd.2022.105796] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/10/2022] [Accepted: 06/14/2022] [Indexed: 02/08/2023] Open
Abstract
Neurons are highly polarized cells that rely on the intracellular transport of organelles. This process is regulated by molecular motors such as dynein and kinesins and the Rab family of monomeric GTPases that together help move cargo along microtubules in dendrites, somas, and axons. Rab5-Rab11 GTPases regulate receptor trafficking along early-recycling endosomes, which is a process that determines the intracellular signaling output of different signaling pathways, including those triggered by BDNF binding to its tyrosine kinase receptor TrkB. BDNF is a well-recognized neurotrophic factor that regulates experience-dependent plasticity in different circuits in the brain. The internalization of the BDNF/TrkB complex results in signaling endosomes that allow local signaling in dendrites and presynaptic terminals, nuclear signaling in somas and dynein-mediated long-distance signaling from axons to cell bodies. In this review, we briefly discuss the organization of the endocytic pathway and how Rab11-recycling endosomes interact with other endomembrane systems. We further expand upon the roles of the Rab11-recycling pathway in neuronal plasticity. Then, we discuss the BDNF/TrkB signaling pathways and their functional relationships with the postendocytic trafficking of BDNF, including axonal transport, emphasizing the role of BDNF signaling endosomes, particularly Rab5-Rab11 endosomes, in neuronal plasticity. Finally, we discuss the evidence indicating that the dysfunction of the early-recycling pathway impairs BDNF signaling, contributing to several neurodegenerative diseases.
Collapse
|
19
|
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: 0.3] [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
|
20
|
Selective motor activation in organelle transport along axons. Nat Rev Mol Cell Biol 2022; 23:699-714. [DOI: 10.1038/s41580-022-00491-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2022] [Indexed: 12/17/2022]
|
21
|
Claes M, Geeraerts E, Plaisance S, Mentens S, Van den Haute C, De Groef L, Arckens L, Moons L. Chronic Chemogenetic Activation of the Superior Colliculus in Glaucomatous Mice: Local and Retrograde Molecular Signature. Cells 2022; 11:1784. [PMID: 35681479 PMCID: PMC9179903 DOI: 10.3390/cells11111784] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 05/20/2022] [Accepted: 05/22/2022] [Indexed: 12/13/2022] Open
Abstract
One important facet of glaucoma pathophysiology is axonal damage, which ultimately disrupts the connection between the retina and its postsynaptic brain targets. The concurrent loss of retrograde support interferes with the functionality and survival of the retinal ganglion cells (RGCs). Previous research has shown that stimulation of neuronal activity in a primary retinal target area-i.e., the superior colliculus-promotes RGC survival in an acute mouse model of glaucoma. To build further on this observation, we applied repeated chemogenetics in the superior colliculus of a more chronic murine glaucoma model-i.e., the microbead occlusion model-and performed bulk RNA sequencing on collicular lysates and isolated RGCs. Our study revealed that chronic target stimulation upon glaucomatous injury phenocopies the a priori expected molecular response: growth factors were pinpointed as essential transcriptional regulators both in the locally stimulated tissue and in distant, unstimulated RGCs. Strikingly, and although the RGC transcriptome revealed a partial reversal of the glaucomatous signature and an enrichment of pro-survival signaling pathways, functional rescue of injured RGCs was not achieved. By postulating various explanations for the lack of RGC neuroprotection, we aim to warrant researchers and drug developers for the complexity of chronic neuromodulation and growth factor signaling.
Collapse
Affiliation(s)
- Marie Claes
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; (M.C.); (E.G.); (S.M.)
| | - Emiel Geeraerts
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; (M.C.); (E.G.); (S.M.)
| | | | - Stephanie Mentens
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; (M.C.); (E.G.); (S.M.)
- Cellular Communication and Neurodegeneration Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium;
- Neuroplasticity and Neuroproteomics Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium;
| | - Chris Van den Haute
- Neurobiology and Gene Therapy Research Group, Department of Neurosciences, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium;
- KU Leuven Viral Vector Core, 3000 Leuven, Belgium
| | - Lies De Groef
- Cellular Communication and Neurodegeneration Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium;
| | - Lut Arckens
- Neuroplasticity and Neuroproteomics Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium;
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven Brain Institute, 3000 Leuven, Belgium; (M.C.); (E.G.); (S.M.)
| |
Collapse
|
22
|
Pradeepkiran JA, Hindle A, Kshirsagar S, Reddy PH. Are mitophagy enhancers therapeutic targets for Alzheimer's disease? Biomed Pharmacother 2022; 149:112918. [PMID: 35585708 PMCID: PMC9148418 DOI: 10.1016/j.biopha.2022.112918] [Citation(s) in RCA: 22] [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: 11/24/2021] [Revised: 03/18/2022] [Accepted: 03/30/2022] [Indexed: 01/09/2023] Open
Abstract
Healthy mitochondria are essential for functional bioenergetics, calcium signaling, and balanced redox homeostasis. Dysfunctional mitochondria are a central aspect of aging and neurodegenerative diseases such as Alzheimer's disease (AD). The formation and accumulation of amyloid beta (Aβ) and hyperphosphorylated tau (P-tau) play large roles in the cellular changes seen in AD, including mitochondrial dysfunction, synaptic damage, neuronal loss, and defective mitophagy. Mitophagy is the cellular process whereby damaged mitochondria are selectively removed, and it plays an important role in mitochondrial quality control. Dysfunctional mitochondria are associated with increased reactive oxygen species and increased levels of Aβ, P-tau and Drp1, which together trigger mitophagy and autophagy. Impaired mitophagy causes the progressive accumulation of defective organelles and damaged mitochondria, and it has been hypothesized that the restoration of mitophagy may offer therapeutic benefits to AD patients. This review highlights the challenges of pharmacologically inducing mitophagy through two different signaling cascades: 1) The PINK1/parkin-dependent pathway and 2) the PINK1/parkin-independent pathway, with an emphasis on abnormal mitochondrial interactions with Aβ and P-Tau, which alter mitophagy in an age-dependent manner. This article also summarizes recent studies on the effects of mitophagy enhancers, including urolithin A, NAD+, actinonin, and tomatidine, on mutant APP/Aβ and mutant Tau. Findings from our lab have revealed that mitophagy enhancers can suppress APP/Aβ-induced and mutant Tau-induced mitochondrial and synaptic dysfunctions in mouse and cell line models of AD. Finally, we discuss the mechanisms underlying the beneficial health effects of mitophagy enhancers like urolithin A, NAD+, resveratrol and spermidine in AD.
Collapse
Affiliation(s)
| | - Ashly Hindle
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Sudhir Kshirsagar
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
23
|
Bonanni R, Cariati I, Tarantino U, D’Arcangelo G, Tancredi V. Physical Exercise and Health: A Focus on Its Protective Role in Neurodegenerative Diseases. J Funct Morphol Kinesiol 2022; 7:jfmk7020038. [PMID: 35645300 PMCID: PMC9149968 DOI: 10.3390/jfmk7020038] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 02/07/2023] Open
Abstract
Scientific evidence has demonstrated the power of physical exercise in the prevention and treatment of numerous chronic and/or age-related diseases, such as musculoskeletal, metabolic, and cardiovascular disorders. In addition, regular exercise is known to play a key role in the context of neurodegenerative diseases, as it helps to reduce the risk of their onset and counteracts their progression. However, the underlying molecular mechanisms have not yet been fully elucidated. In this regard, neurotrophins, such as brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), glia cell line-derived neurotrophic factor (GDNF), neurotrophin-3 (NT-3), and neurotrophin-4 (NT-4), have been suggested as key mediators of brain health benefits, as they are involved in neurogenesis, neuronal survival, and synaptic plasticity. The production of these neurotrophic factors, known to be increased by physical exercise, is downregulated in neurodegenerative disorders, suggesting their fundamental importance in maintaining brain health. However, the mechanism by which physical exercise promotes the production of neurotrophins remains to be understood, posing limits on their use for the development of potential therapeutic strategies for the treatment of neurodegenerative diseases. In this literature review, we analyzed the most recent evidence regarding the relationship between physical exercise, neurotrophins, and brain health, providing an overview of their involvement in the onset and progression of neurodegeneration.
Collapse
Affiliation(s)
- Roberto Bonanni
- Department of Clinical Sciences and Translational Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (R.B.); (U.T.)
| | - Ida Cariati
- Department of Clinical Sciences and Translational Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (R.B.); (U.T.)
- Correspondence:
| | - Umberto Tarantino
- Department of Clinical Sciences and Translational Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (R.B.); (U.T.)
- Department of Orthopaedics and Traumatology, “Policlinico Tor Vergata” Foundation, 00133 Rome, Italy
- Centre of Space Bio-Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (G.D.); (V.T.)
| | - Giovanna D’Arcangelo
- Centre of Space Bio-Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (G.D.); (V.T.)
- Department of Systems Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy
| | - Virginia Tancredi
- Centre of Space Bio-Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy; (G.D.); (V.T.)
- Department of Systems Medicine, “Tor Vergata” University of Rome, 00133 Rome, Italy
| |
Collapse
|
24
|
Vermehren-Schmaedick A, Olah MJ, Ramunno-Johnson D, Lidke KA, Cohen MS, Vu TQ. Molecular-Scale Dynamics of Long Range Retrograde Brain-Derived Neurotrophic Factor Transport Shaped by Cellular Spatial Context. Front Neurosci 2022; 16:835815. [PMID: 35431786 PMCID: PMC9008462 DOI: 10.3389/fnins.2022.835815] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/03/2022] [Indexed: 12/11/2022] Open
Abstract
Retrograde neurotrophin (NT) transport is a specialized form of signal transduction used to conduct information from axons to the cell bodies of central and peripheral nervous system neurons. It is activated upon NT-Trk receptor binding, NT-Trk internalization into signaling endosomes, and their motion along the axon toward the cell body. Brain-derived neurotrophic factor (BDNF) is an abundant NT that modulates key brain and spinal cord functions, and defects in BDNF trafficking are associated with neuronal death, neurodegenerative diseases and in nerve injury. Decades of study have yielded impressive progress in elucidating NT retrograde transport; however, much information remains unclear. For example, while it is known that NT function is dependent on tight control of NT-receptor intracellular trafficking, data describing the precise spatiotemporal molecular dynamics of their axonal to somatic transport are lacking. In past work, we showed the use of discrete, photo-bleaching-resistant quantum dot (QD)-BNDF probes to activate and track BDNF-TrkB receptor internalization; this revealed a rich diversity of molecular motions that intracellular BDNF signaling endosomes undergo within the soma of nodose ganglia sensory neurons. Here, we used combined techniques of discrete QD-BDNF tracking with compartmented microfluidic chambers to characterize retrograde BDNF-TrkB transport over long-ranging distances of primary dorsal root ganglion sensory neuronal axons. Our new findings show that axonal retrograde motion is comprised of heterogeneous mixtures of diffusive behaviors, pauses, and variations in net molecular-motor-dependent transport speeds. Notably, specific molecular dynamic features such as NT speed were dependent on spatial context that could be categorized in distance from distal axons and proximity to the soma and were not entirely dictated by active motor transport speed. The important implication is recognition that NT-receptor retrograde transport is comprised of molecular dynamics, which change over the course of long-range trafficking to shape overall transport and possibly signaling.
Collapse
Affiliation(s)
- Anke Vermehren-Schmaedick
- Department of Biomedical Engineering, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, United States
| | - Mark J. Olah
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, United States
| | - Damien Ramunno-Johnson
- Department of Biomedical Engineering, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| | - Keith A. Lidke
- Department of Physics and Astronomy, University of New Mexico, Albuquerque, NM, United States
| | - Michael S. Cohen
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, United States
| | - Tania Q. Vu
- Department of Biomedical Engineering, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
25
|
Interaction between TRPML1 and p62 in Regulating Autophagosome-Lysosome Fusion and Impeding Neuroaxonal Dystrophy in Alzheimer’s Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8096009. [PMID: 35116093 PMCID: PMC8807035 DOI: 10.1155/2022/8096009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 12/10/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022]
Abstract
The loss of transient receptor potential mucolipin 1 (TRPML1), an endosomal and lysosomal Ca2+-releasing channel, has been implicated in neurodegenerative disorders. Mounting evidence have shown that TRPML1 could clear intraneuronal amyloid-β (Aβ), which triggers a hypothesis that TRPML1 activation may be beneficial for axonal transport in Alzheimer's disease (AD). In this work, the functional roles of TRPML1 were studied in the APP/PS1 transgenic mice and Aβ1-42-stimulated hippocampal neurons HT22. We found that lentivirus-mediated overexpression of TRPML1 was shown to promote an accumulation of autolysosomes and increase brain-derived neurotrophic factor (BDNF) transportation to the nucleus, suggesting an axon-protective function. More importantly, we found that TRPML1 also increased p62 that interacted with dynein. Lentivirus-mediated knockdown of p62 or inhibition of dynein by ciliobrevin D stimulation was found to reduce autolysosome formation and nuclear accumulation of BDNF in HT22 cells with Aβ1-42 stimulation. Inhibition of p62 by XRK3F2 stimulation was observed to promote the death of hippocampal neurons of the APP/PS1 transgenic mice. TRPML1 recruited dynein by interacting with p62 to promote the autophagosome-lysosome fusion to mediate BDNF nuclear translocation to impede axon dystrophy in mice with Alzheimer-like phenotypes. In summary, these results demonstrate the presence of a TRPML1/p62/dynein regulatory network in AD, and activation of TRPML1 is required for axon protection to prevent neuroaxonal dystrophy.
Collapse
|
26
|
Scaramuzzino C, Cuoc EC, Pla P, Humbert S, Saudou F. Calcineurin and huntingtin form a calcium-sensing machinery that directs neurotrophic signals to the nucleus. SCIENCE ADVANCES 2022; 8:eabj8812. [PMID: 34985962 PMCID: PMC8730605 DOI: 10.1126/sciadv.abj8812] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
When a neurotrophin binds at the presynapse, it sends survival signals all the way to the nucleus on signaling endosomes. These endosomes fuel their own journey with on-board glycolysis—but how is that journey initiated and maintained? Using microfluidic devices and mice, we find that the calcium released upon brain-derived neurotrophic factor (BDNF) binding to its receptor, tropomyosin receptor kinase B (TrkB), is sensed by calcineurin on the cytosolic face of the endosome. Calcineurin dephosphorylates huntingtin, the BDNF scaffold, which sets the endosome moving in a retrograde direction. In an in vitro reconstituted microtubule transport system, controlled calcium uncaging prompts purified vesicles to move to the microtubule minus end. We observed similar retrograde waves of TrkA- and epidermal growth factor receptor (EGFR)-bearing endosomes. Signaling endosomes in neurons thus carry not only their own fuel, but their own navigational system.
Collapse
|
27
|
Chiang NN, Lin TH, Teng YS, Sun YC, Chang KH, Lin CY, Hsieh-Li HM, Su MT, Chen CM, Lee-Chen GJ. Flavones 7,8-DHF, Quercetin, and Apigenin Against Tau Toxicity via Activation of TRKB Signaling in ΔK280 Tau RD-DsRed SH-SY5Y Cells. Front Aging Neurosci 2022; 13:758895. [PMID: 34975454 PMCID: PMC8714935 DOI: 10.3389/fnagi.2021.758895] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 11/17/2021] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease with memory loss and cognitive decline. Neurofibrillary tangles (NFTs) formed by hyperphosphorylated Tau protein are one of the pathological hallmarks of several neurodegenerative diseases including AD. Heat shock protein family B (small) member 1 (HSPB1) is a molecular chaperone that promotes the correct folding of other proteins in response to environmental stress. Nuclear factor erythroid 2-like 2 (NRF2), a redox-regulated transcription factor, is the master regulator of the cellular response to excess reactive oxygen species. Tropomyosin-related kinase B (TRKB) is a membrane-bound receptor that, upon binding brain-derived neurotrophic factor (BDNF), phosphorylates itself to initiate downstream signaling for neuronal survival and axonal growth. In this study, four natural flavones such as 7,8-dihydroxyflavone (7,8-DHF), wogonin, quercetin, and apigenin were evaluated for Tau aggregation inhibitory activity and neuroprotection in SH-SY5Y neuroblastoma. Among the tested flavones, 7,8-DHF, quercetin, and apigenin reduced Tau aggregation, oxidative stress, and caspase-1 activity as well as improved neurite outgrowth in SH-SY5Y cells expressing ΔK280 TauRD-DsRed folding reporter. Treatments with 7,8-DHF, quercetin, and apigenin rescued the reduced HSPB1 and NRF2 and activated TRKB-mediated extracellular signal-regulated kinase (ERK) signaling to upregulate cAMP-response element binding protein (CREB) and its downstream antiapoptotic BCL2 apoptosis regulator (BCL2). Knockdown of TRKB attenuated the neuroprotective effects of these three flavones. Our results suggest 7,8-DHF, quercetin, and apigenin targeting HSPB1, NRF2, and TRKB to reduce Tau aggregation and protect cells against Tau neurotoxicity and may provide new treatment strategies for AD.
Collapse
Affiliation(s)
- Ni-Ni Chiang
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Te-Hsien Lin
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Yu-Shan Teng
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ying-Chieh Sun
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Chung-Yin Lin
- Medical Imaging Research Center, Institute for Radiological Research, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Hsiu Mei Hsieh-Li
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Ming-Tsan Su
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University School of Medicine, Taoyuan, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei, Taiwan
| |
Collapse
|
28
|
Cai Q, Ganesan D. Regulation of neuronal autophagy and the implications in neurodegenerative diseases. Neurobiol Dis 2022; 162:105582. [PMID: 34890791 PMCID: PMC8764935 DOI: 10.1016/j.nbd.2021.105582] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/22/2021] [Accepted: 12/06/2021] [Indexed: 01/03/2023] Open
Abstract
Neurons are highly polarized and post-mitotic cells with the specific requirements of neurotransmission accompanied by high metabolic demands that create a unique challenge for the maintenance of cellular homeostasis. Thus, neurons rely heavily on autophagy that constitutes a key quality control system by which dysfunctional cytoplasmic components, protein aggregates, and damaged organelles are sequestered within autophagosomes and then delivered to the lysosome for degradation. While mature lysosomes are predominantly located in the soma of neurons, the robust, constitutive biogenesis of autophagosomes occurs in the synaptic terminal via a conserved pathway that is required to maintain synaptic integrity and function. Following formation, autophagosomes fuse with late endosomes and then are rapidly and efficiently transported by the microtubule-based cytoplasmic dynein motor along the axon toward the soma for lysosomal clearance. In this review, we highlight the recent knowledge of the roles of autophagy in neuronal health and disease. We summarize the available evidence about the normal functions of autophagy as a protective factor against neurodegeneration and discuss the mechanism underlying neuronal autophagy regulation. Finally, we describe how autophagy function is affected in major neurodegenerative diseases with a special focus on Alzheimer's disease, Parkinson's disease, and Amyotrophic Lateral Sclerosis.
Collapse
|
29
|
Wang T, Meunier FA. Live-Cell Superresolution Imaging of Retrograde Axonal Trafficking Using Pulse-Chase Labeling in Cultured Hippocampal Neurons. Methods Mol Biol 2022; 2473:101-128. [PMID: 35819762 DOI: 10.1007/978-1-0716-2209-4_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The entanglement of long axons found in cultured dissociated hippocampal neurons restricts the analysis of the machinery underlying directed axonal trafficking. Further, hippocampal neurons exhibit "en passant" presynapses that may confound the analysis of long-range retrograde axonal transport. To solve these issues, we and others have developed microfluid-based methods to specifically follow the fates of the retrograde axonal cargoes following pulse-chase labeling by super-resolution live-cell imaging, and automatically tracking their directed transport and analyzing their kinetical properties. These methods have allowed us to visualize the trafficking of fluorescently tagged signaling endosomes and autophagosomes derived from axonal terminals and resolve their localizations and movements with high spatial and temporal accuracy. In this chapter, we describe how to use a commercially available microfluidic device to enable the labeling and tracking of retrograde axonal carriers, including (1) how to culture and transfect rat hippocampal neurons in the microfluidic device; (2) how to perform pulse-chase to label specific populations of retrograde axonal carriers; and (3) how to conduct the automatic tracking and data analysis using open-source software.
Collapse
Affiliation(s)
- Tong Wang
- Center for Brain Science, School of Life Science and Technology, Shanghaitech University, Shanghai, China.
| | - Frédéric A Meunier
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
30
|
Microfabricated Devices for Confocal Microscopy on Biological Samples. Methods Mol Biol 2021. [PMID: 34028712 DOI: 10.1007/978-1-0716-1402-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Microfabricated devices have found applications in a range of biomedical research problems in recent years, with thousands of research papers published and multiple commercial devices now available. This chapter is intended to provide an overview of the available options for devices compatible with confocal microscopy, including an overview of fabrication techniques and some examples of device use. Although there are times when off-the-shelf devices are well suited for the problem at hand, in some cases customized devices are necessary or more convenient. Protocols for researchers who wish to make their own devices are outlined below; although fabricating templates for devices requires some specialized equipment, making PDMS or hydrogel devices from templates can be done in a standard laboratory setting.
Collapse
|
31
|
Schnöder L, Tomic I, Schwindt L, Helm D, Rettel M, Schulz-Schaeffer W, Krause E, Rettig J, Fassbender K, Liu Y. P38α-MAPK phosphorylates Snapin and reduces Snapin-mediated BACE1 transportation in APP-transgenic mice. FASEB J 2021; 35:e21691. [PMID: 34118085 DOI: 10.1096/fj.202100017r] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 04/18/2021] [Accepted: 05/07/2021] [Indexed: 11/11/2022]
Abstract
Amyloid β peptide (Aβ) is the major pathogenic molecule in Alzheimer's disease (AD). BACE1 enzyme is essential for the generation of Aβ. Deficiency of p38α-MAPK in neurons increases lysosomal degradation of BACE1 and decreases Aβ deposition in the brain of APP-transgenic mice. However, the mechanisms mediating effects of p38α-MAPK are largely unknown. In this study, we used APP-transgenic mice and cultured neurons and observed that deletion of p38α-MAPK specifically in neurons decreased phosphorylation of Snapin at serine, increased retrograde transportation of BACE1 in axons and reduced BACE1 at synaptic terminals, which suggests that p38α-MAPK deficiency promotes axonal transportation of BACE1 from its predominant locations, axonal terminals, to lysosomes in the cell body. In vitro kinase assay revealed that p38α-MAPK directly phosphorylates Snapin. By further performing mass spectrometry analysis and site-directed mutagenic experiments in SH-SY5Y cell lines, we identified serine residue 112 as a p38α-MAPK-phosphorylating site on Snapin. Replacement of serine 112 with alanine did abolish p38α-MAPK knockdown-induced reduction of BACE1 activity and protein level, and transportation to lysosomes in SH-SY5Y cells. Taken together, our study suggests that activation of p38α-MAPK phosphorylates Snapin and inhibits the retrograde transportation of BACE1 in axons, which might exaggerate amyloid pathology in AD brain.
Collapse
Affiliation(s)
- Laura Schnöder
- Department of Neurology, Saarland University, Homburg/Saar, Germany.,German Institute for Dementia Prevention (DIDP), Saarland University, Homburg/Saar, Germany
| | - Inge Tomic
- Department of Neurology, Saarland University, Homburg/Saar, Germany.,German Institute for Dementia Prevention (DIDP), Saarland University, Homburg/Saar, Germany
| | - Laura Schwindt
- Department of Neurology, Saarland University, Homburg/Saar, Germany.,German Institute for Dementia Prevention (DIDP), Saarland University, Homburg/Saar, Germany
| | - Dominic Helm
- European Molecular Biology Laboratory, Proteomics Core Facility, Heidelberg, Germany
| | - Mandy Rettel
- European Molecular Biology Laboratory, Proteomics Core Facility, Heidelberg, Germany
| | | | - Elmar Krause
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Jens Rettig
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, Homburg, Germany
| | - Klaus Fassbender
- Department of Neurology, Saarland University, Homburg/Saar, Germany.,German Institute for Dementia Prevention (DIDP), Saarland University, Homburg/Saar, Germany
| | - Yang Liu
- Department of Neurology, Saarland University, Homburg/Saar, Germany.,German Institute for Dementia Prevention (DIDP), Saarland University, Homburg/Saar, Germany
| |
Collapse
|
32
|
Jiang M, Kuang Z, He Y, Cao Y, Yu T, Cheng J, Liu W, Wang W. SNAPIN Regulates Cell Cycle Progression to Promote Pancreatic β Cell Growth. Front Endocrinol (Lausanne) 2021; 12:624309. [PMID: 34194388 DOI: 10.3389/fendo.2021.624309] [cited] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 05/13/2021] [Indexed: 01/26/2025] Open
Abstract
In diabetes mellitus, death of β cell in the pancreas occurs throughout the development of the disease, with loss of insulin production. The maintenance of β cell number is essential to maintaining normoglycemia. SNAPIN has been found to regulate insulin secretion, but whether it induces β cell proliferation remains to be elucidated. This study aimed to explore the physiological roles of SNAPIN in β cell proliferation. SNAPIN expression increases with the age of mice and SNAPIN is down-regulated in diabetes. KEGG pathway and GO analysis showed that SNAPIN- interacting proteins were enriched in cell cycle regulation. B cell cycle was arrested in the S phase, and cell proliferation was inhibited after SNAPIN knockdown. The expression of CDK2, CDK4 and CCND1 proteins in the S phase of the cell cycle were reduced after SNAPIN knockdown, whereas they were increased after overexpression of SNAPIN. In addition, insulin protein and mRNA levels also increased or decreased after SNAPIN knockdown or overexpression, respectively. Conclusions: Our data indicate that SNAPIN mediates β cells proliferation and insulin secretion, and provide evidences that SNAPIN might be a pharmacotherapeutic target for diabetes mellitus.
Collapse
Affiliation(s)
- Mengxue Jiang
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhijian Kuang
- Fujian Provincial KeyLaboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Yaohui He
- Fujian Provincial KeyLaboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Yin Cao
- Fujian Provincial KeyLaboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Tingyan Yu
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jidong Cheng
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wen Liu
- Fujian Provincial KeyLaboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Wei Wang
- Department of Endocrinology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
33
|
Understanding amphisomes. Biochem J 2021; 478:1959-1976. [PMID: 34047789 DOI: 10.1042/bcj20200917] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/27/2021] [Accepted: 05/06/2021] [Indexed: 12/14/2022]
Abstract
Amphisomes are intermediate/hybrid organelles produced through the fusion of endosomes with autophagosomes within cells. Amphisome formation is an essential step during a sequential maturation process of autophagosomes before their ultimate fusion with lysosomes for cargo degradation. This process is highly regulated with multiple protein machineries, such as SNAREs, Rab GTPases, tethering complexes, and ESCRTs, are involved to facilitate autophagic flux to proceed. In neurons, autophagosomes are robustly generated in axonal terminals and then rapidly fuse with late endosomes to form amphisomes. This fusion event allows newly generated autophagosomes to gain retrograde transport motility and move toward the soma, where proteolytically active lysosomes are predominantly located. Amphisomes are not only the products of autophagosome maturation but also the intersection of the autophagy and endo-lysosomal pathways. Importantly, amphisomes can also participate in non-canonical functions, such as retrograde neurotrophic signaling or autophagy-based unconventional secretion by fusion with the plasma membrane. In this review, we provide an updated overview of the recent discoveries and advancements on the molecular and cellular mechanisms underlying amphisome biogenesis and the emerging roles of amphisomes. We discuss recent developments towards the understanding of amphisome regulation as well as the implications in the context of major neurodegenerative diseases, with a comparative focus on Alzheimer's disease and Parkinson's disease.
Collapse
|
34
|
Camblor-Perujo S, Kononenko NL. Brain-specific functions of the endocytic machinery. FEBS J 2021; 289:2219-2246. [PMID: 33896112 DOI: 10.1111/febs.15897] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/29/2021] [Indexed: 12/12/2022]
Abstract
Endocytosis is an essential cellular process required for multiple physiological functions, including communication with the extracellular environment, nutrient uptake, and signaling by the cell surface receptors. In a broad sense, endocytosis is accomplished through either constitutive or ligand-induced invagination of the plasma membrane, which results in the formation of the plasma membrane-retrieved endocytic vesicles, which can either be sent for degradation to the lysosomes or recycled back to the PM. This additional function of endocytosis in membrane retrieval has been adopted by excitable cells, such as neurons, for membrane equilibrium maintenance at synapses. The last two decades were especially productive with respect to the identification of brain-specific functions of the endocytic machinery, which additionally include but not limited to regulation of neuronal differentiation and migration, maintenance of neuron morphology and synaptic plasticity, and prevention of neurotoxic aggregates spreading. In this review, we highlight the current knowledge of brain-specific functions of endocytic machinery with a specific focus on three brain cell types, neuronal progenitor cells, neurons, and glial cells.
Collapse
Affiliation(s)
| | - Natalia L Kononenko
- CECAD Cluster of Excellence, University of Cologne, Germany.,Center for Physiology & Pathophysiology, Medical Faculty, University of Cologne, Germany
| |
Collapse
|
35
|
Pathak A, Clark S, Bronfman FC, Deppmann CD, Carter BD. Long-distance regressive signaling in neural development and disease. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2021; 10:e382. [PMID: 32391977 PMCID: PMC7655682 DOI: 10.1002/wdev.382] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 03/23/2020] [Accepted: 04/06/2020] [Indexed: 02/06/2023]
Abstract
Nervous system development proceeds via well-orchestrated processes involving a balance between progressive and regressive events including stabilization or elimination of axons, synapses, and even entire neurons. These progressive and regressive events are driven by functionally antagonistic signaling pathways with the dominant pathway eventually determining whether a neural element is retained or removed. Many of these developmental sculpting events are triggered by final target innervation necessitating a long-distance mode of communication. While long-distance progressive signaling has been well characterized, particularly for neurotrophic factors, there remains relatively little known about how regressive events are triggered from a distance. Here we discuss the emergent phenomenon of long-distance regressive signaling pathways. In particular, we will cover (a) progressive and regressive cues known to be employed after target innervation, (b) the mechanisms of long-distance signaling from an endosomal platform, (c) recent evidence that long-distance regressive cues emanate from platforms like death receptors or repulsive axon guidance receptors, and (d) evidence that these pathways are exploited in pathological scenarios. This article is categorized under: Nervous System Development > Vertebrates: General Principles Signaling Pathways > Global Signaling Mechanisms Establishment of Spatial and Temporal Patterns > Cytoplasmic Localization.
Collapse
Affiliation(s)
- Amrita Pathak
- Department of Biochemistry and Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Shayla Clark
- Neuroscience Graduate Program, University of Virginia, Charlottesville, Virginia
| | - Francisca C. Bronfman
- Institute of Biomedical Sciences (ICB), Faculty of Medicine, Faculty of Life Science, Universidad Andres Bello, Santiago, Chile
| | - Christopher D. Deppmann
- Departments of Biology, Cell Biology, Biomedical Engineering, and Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Bruce D. Carter
- Department of Biochemistry and Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee
| |
Collapse
|
36
|
Kuijpers M, Azarnia Tehran D, Haucke V, Soykan T. The axonal endolysosomal and autophagic systems. J Neurochem 2021; 158:589-602. [PMID: 33372296 DOI: 10.1111/jnc.15287] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/26/2022]
Abstract
Neurons, because of their elaborate morphology and the long distances between distal axons and the soma as well as their longevity, pose special challenges to autophagy and to the endolysosomal system, two of the main degradative routes for turnover of defective proteins and organelles. Autophagosomes sequester cytoplasmic or organellar cargos by engulfing them into their lumen before fusion with degradative lysosomes enriched in neuronal somata and participate in retrograde signaling to the soma. Endosomes are mainly involved in the sorting, recycling, or lysosomal turnover of internalized or membrane-bound macromolecules to maintain axonal membrane homeostasis. Lysosomes and the multiple shades of lysosome-related organelles also serve non-degradative roles, for example, in nutrient signaling and in synapse formation. Recent years have begun to shed light on the distinctive organization of the autophagy and endolysosomal systems in neurons, in particular their roles in axons. We review here our current understanding of the localization, distribution, and growing list of functions of these organelles in the axon in health and disease and outline perspectives for future research.
Collapse
Affiliation(s)
- Marijn Kuijpers
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | | | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany.,Freie Universität Berlin, Faculty of Biology, Chemistry, Berlin, Germany.,Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Tolga Soykan
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| |
Collapse
|
37
|
Abstract
During the development of the nervous system, neurons respond to diffusible cues secreted by target cells. Because such target-derived factors regulate development, maturation, and maintenance of axons as well as somatodendritic compartments, signals initiated at distal axons must be retrogradely transmitted toward cell bodies. Neurotrophins, including the nerve growth factor (NGF), provide one of the best-known examples of target-derived growth factors. The cell biological processes of endocytosis and retrograde trafficking of their Trk receptors from growth cones to cell bodies are key mechanisms by which target-derived neurotrophins influence neurons. Evidence accumulated over the past several decades has begun to uncover the molecular mechanisms of formation, transport, and biological functions of these specialized endosomes called "signaling endosomes."
Collapse
|
38
|
Enlargement of early endosomes and traffic jam in basal forebrain cholinergic neurons in Alzheimer's disease. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:207-218. [PMID: 34225963 DOI: 10.1016/b978-0-12-819975-6.00011-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
While a handful of neurotransmitter systems including cholinergic, norepinephrinergic, and serotonergic undergo significant degeneration in Alzheimer's disease, the cholinergic system has been the prime target for research and therapy. The cholinergic system in the basal forebrain is strategically located to impose significant modulatory effects on vast cortical and subcortical regions of the brain. Numerous studies have established a strong link between neurotrophin signaling and basal forebrain cholinergic neuron degeneration in several neurodegenerative disorders. Evidence presented during the last few years points to the effects of endosomal pathology and primarily unidirectional traffic jam. Hence, formulating new therapies, e.g., to reduce local production of β C-terminal fragments and preventing changes in endosomal morphology have become attractive potential therapeutic strategies to restore cholinergic neurons and their neuromodulatory function. While it is not expected that restoring the cholinergic system function will fully mitigate cognitive dysfunction in Alzheimer's disease, pivotal aspects of cognition including attention-deficit during the prodromal stages might well be at disposal for corrective measures.
Collapse
|
39
|
Jiang M, Kuang Z, He Y, Cao Y, Yu T, Cheng J, Liu W, Wang W. SNAPIN Regulates Cell Cycle Progression to Promote Pancreatic β Cell Growth. Front Endocrinol (Lausanne) 2021; 12:624309. [PMID: 34194388 PMCID: PMC8237857 DOI: 10.3389/fendo.2021.624309] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 05/13/2021] [Indexed: 02/05/2023] Open
Abstract
In diabetes mellitus, death of β cell in the pancreas occurs throughout the development of the disease, with loss of insulin production. The maintenance of β cell number is essential to maintaining normoglycemia. SNAPIN has been found to regulate insulin secretion, but whether it induces β cell proliferation remains to be elucidated. This study aimed to explore the physiological roles of SNAPIN in β cell proliferation. SNAPIN expression increases with the age of mice and SNAPIN is down-regulated in diabetes. KEGG pathway and GO analysis showed that SNAPIN- interacting proteins were enriched in cell cycle regulation. B cell cycle was arrested in the S phase, and cell proliferation was inhibited after SNAPIN knockdown. The expression of CDK2, CDK4 and CCND1 proteins in the S phase of the cell cycle were reduced after SNAPIN knockdown, whereas they were increased after overexpression of SNAPIN. In addition, insulin protein and mRNA levels also increased or decreased after SNAPIN knockdown or overexpression, respectively. Conclusions: Our data indicate that SNAPIN mediates β cells proliferation and insulin secretion, and provide evidences that SNAPIN might be a pharmacotherapeutic target for diabetes mellitus.
Collapse
Affiliation(s)
- Mengxue Jiang
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhijian Kuang
- Fujian Provincial KeyLaboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Yaohui He
- Fujian Provincial KeyLaboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Yin Cao
- Fujian Provincial KeyLaboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
| | - Tingyan Yu
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jidong Cheng
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Jidong Cheng, ; Wen Liu, ; Wei Wang,
| | - Wen Liu
- Fujian Provincial KeyLaboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, China
- *Correspondence: Jidong Cheng, ; Wen Liu, ; Wei Wang,
| | - Wei Wang
- Department of Endocrinology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- *Correspondence: Jidong Cheng, ; Wen Liu, ; Wei Wang,
| |
Collapse
|
40
|
Andres-Alonso M, Kreutz MR, Karpova A. Autophagy and the endolysosomal system in presynaptic function. Cell Mol Life Sci 2020; 78:2621-2639. [PMID: 33340068 PMCID: PMC8004491 DOI: 10.1007/s00018-020-03722-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 11/17/2020] [Accepted: 11/24/2020] [Indexed: 12/11/2022]
Abstract
The complex morphology of neurons, the specific requirements of synaptic neurotransmission and the accompanying metabolic demands create a unique challenge for proteostasis. The main machineries for neuronal protein synthesis and degradation are localized in the soma, while synaptic junctions are found at vast distances from the cell body. Sophisticated mechanisms must, therefore, ensure efficient delivery of newly synthesized proteins and removal of faulty proteins. These requirements are exacerbated at presynaptic sites, where the demands for protein turnover are especially high due to synaptic vesicle release and recycling that induces protein damage in an intricate molecular machinery, and where replacement of material is hampered by the extreme length of the axon. In this review, we will discuss the contribution of the two major pathways in place, autophagy and the endolysosomal system, to presynaptic protein turnover and presynaptic function. Although clearly different in their biogenesis, both pathways are characterized by cargo collection and transport into distinct membrane-bound organelles that eventually fuse with lysosomes for cargo degradation. We summarize the available evidence with regard to their degradative function, their regulation by presynaptic machinery and the cargo for each pathway. Finally, we will discuss the interplay of both pathways in neurons and very recent findings that suggest non-canonical functions of degradative organelles in synaptic signalling and plasticity.
Collapse
Affiliation(s)
- Maria Andres-Alonso
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
- Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Michael R Kreutz
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- Leibniz Group 'Dendritic Organelles and Synaptic Function', Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany.
- Center for Behavioral Brain Sciences, Otto Von Guericke University, Magdeburg, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany.
| | - Anna Karpova
- Research Group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- Center for Behavioral Brain Sciences, Otto Von Guericke University, Magdeburg, Germany.
| |
Collapse
|
41
|
Halakos EG, Connell AJ, Glazewski L, Wei S, Mason RW. Bottom up proteomics identifies neuronal differentiation pathway networks activated by cathepsin inhibition treatment in neuroblastoma cells that are enhanced by concurrent 13-cis retinoic acid treatment. J Proteomics 2020; 232:104068. [PMID: 33278663 DOI: 10.1016/j.jprot.2020.104068] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/16/2020] [Accepted: 11/29/2020] [Indexed: 12/19/2022]
Abstract
Neuroblastoma is the second most common pediatric cancer involving the peripheral nervous system in which stage IVS metastatic tumors regress due to spontaneous differentiation. 13-cis retinoic acid (13-cis RA) is currently used in the clinic for its differentiation effects and although it improves outcomes, relapse is seen in half of high-risk patients. Combinatorial therapies have been shown to be more effective in oncotherapy and since cathepsin inhibition reduces tumor growth, we explored the potential of coupling 13-cis RA with a cathepsin inhibitor (K777) to enhance therapeutic efficacy against neuroblastoma. Shotgun proteomics was used to identify proteins affected by K777 and dual (13-cis RA/K777) treatment in neuroblastoma SK-N-SH cells. Cathepsin inhibition was more effective in increasing proteins involved in neuronal differentiation and neurite outgrowth than 13-cis RA alone, but the combination of both treatments enhanced the neuronal differentiation effect. SIGNIFICANCE: As neuroblastoma can spontaneously differentiate, determining which proteins are involved in differentiation can guide development of more accurate diagnostic markers and more effective treatments. In this study, we established a differentiation proteomic map of SK-N-SH cells treated with a cathepsin inhibitor (K777) and K777/13-cis RA (dual). Bioinformatic analysis revealed these treatments enhanced neuronal differentiation and axonogenesis pathways. The most affected proteins in these pathways may become valuable biomarkers of efficacy of drugs designed to enhance differentiation of neuroblastoma [1].
Collapse
Affiliation(s)
- Effie G Halakos
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Andrew J Connell
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Lisa Glazewski
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA
| | - Shuo Wei
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| | - Robert W Mason
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
42
|
Overhoff M, De Bruyckere E, Kononenko NL. Mechanisms of neuronal survival safeguarded by endocytosis and autophagy. J Neurochem 2020; 157:263-296. [PMID: 32964462 DOI: 10.1111/jnc.15194] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/21/2020] [Accepted: 09/08/2020] [Indexed: 12/11/2022]
Abstract
Multiple aspects of neuronal physiology crucially depend on two cellular pathways, autophagy and endocytosis. During endocytosis, extracellular components either unbound or recognized by membrane-localized receptors (termed "cargo") become internalized into plasma membrane-derived vesicles. These can serve to either recycle the material back to the plasma membrane or send it for degradation to lysosomes. Autophagy also uses lysosomes as a terminal degradation point, although instead of degrading the plasma membrane-derived cargo, autophagy eliminates detrimental cytosolic material and intracellular organelles, which are transported to lysosomes by means of double-membrane vesicles, referred to as autophagosomes. Neurons, like all non-neuronal cells, capitalize on autophagy and endocytosis to communicate with the environment and maintain protein and organelle homeostasis. Additionally, the highly polarized, post-mitotic nature of neurons made them adopt these two pathways for cell-specific functions. These include the maintenance of the synaptic vesicle pool in the pre-synaptic terminal and the long-distance transport of signaling molecules. Originally discovered independently from each other, it is now clear that autophagy and endocytosis are closely interconnected and share several common participating molecules. Considering the crucial role of autophagy and endocytosis in cell type-specific functions in neurons, it is not surprising that defects in both pathways have been linked to the pathology of numerous neurodegenerative diseases. In this review, we highlight the recent knowledge of the role of endocytosis and autophagy in neurons with a special focus on synaptic physiology and discuss how impairments in genes coding for autophagy and endocytosis proteins can cause neurodegeneration.
Collapse
Affiliation(s)
- Melina Overhoff
- CECAD Cluster of Excellence, Institute for Genetics, University of Cologne, Cologne, Germany
| | - Elodie De Bruyckere
- CECAD Cluster of Excellence, Institute for Genetics, University of Cologne, Cologne, Germany
| | - Natalia L Kononenko
- CECAD Cluster of Excellence, Institute for Genetics, University of Cologne, Cologne, Germany
| |
Collapse
|
43
|
The Rab5-Rab11 Endosomal Pathway is Required for BDNF-Induced CREB Transcriptional Regulation in Hippocampal Neurons. J Neurosci 2020; 40:8042-8054. [PMID: 32928890 DOI: 10.1523/jneurosci.2063-19.2020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 09/02/2020] [Accepted: 09/08/2020] [Indexed: 12/20/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a key regulator of the morphology and connectivity of central neurons. We have previously shown that BDNF/TrkB signaling regulates the activity and mobility of the GTPases Rab5 and Rab11, which in turn determine the postendocytic sorting of signaling TrkB receptors. Moreover, decreased Rab5 or Rab11 activity inhibits BDNF-induced dendritic branching. Whether Rab5 or Rab11 activity is important for local events only or for regulating nuclear signaling and gene expression is unknown. Here, we investigated, in rat hippocampal neuronal cultures derived from embryos of unknown sex, whether BDNF-induced signaling cascades are altered when early and recycling endosomes are disrupted by the expression of dominant-negative mutants of Rab5 and Rab11. The activity of both Rab5 and Rab11 was required for sustained activity of Erk1/2 and nuclear CREB phosphorylation, and increased transcription of a BDNF-dependent program of gene expression containing CRE binding sites, which includes activity-regulated genes such as Arc, Dusp1, c-fos, Egr1, and Egr2, and growth and survival genes such as Atf3 and Gem Based on our results, we propose that early and recycling endosomes provide a platform for the integration of neurotrophic signaling from the plasma membrane to the nucleus in neurons, and that this mechanism is likely to regulate neuronal plasticity and survival.SIGNIFICANCE STATEMENT BDNF is a neurotrophic factor that regulates plastic changes in the brain, including dendritic growth. The cellular and molecular mechanisms underlying this process are not completely understood. Our results uncover the cellular requirements that central neurons possess to integrate the plasma membrane into nuclear signaling in neurons. Our results indicate that the endosomal pathway is required for the signaling cascade initiated by BDNF and its receptors at the plasma membrane to modulate BDNF-dependent gene expression and neuronal dendritic growth mediated by the CREB transcription factor. CREB is a key transcription factor regulating circuit development and learning and memory.
Collapse
|
44
|
Bowman SL, Bi-Karchin J, Le L, Marks MS. The road to lysosome-related organelles: Insights from Hermansky-Pudlak syndrome and other rare diseases. Traffic 2020; 20:404-435. [PMID: 30945407 DOI: 10.1111/tra.12646] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/02/2019] [Accepted: 04/02/2019] [Indexed: 12/11/2022]
Abstract
Lysosome-related organelles (LROs) comprise a diverse group of cell type-specific, membrane-bound subcellular organelles that derive at least in part from the endolysosomal system but that have unique contents, morphologies and functions to support specific physiological roles. They include: melanosomes that provide pigment to our eyes and skin; alpha and dense granules in platelets, and lytic granules in cytotoxic T cells and natural killer cells, which release effectors to regulate hemostasis and immunity; and distinct classes of lamellar bodies in lung epithelial cells and keratinocytes that support lung plasticity and skin lubrication. The formation, maturation and/or secretion of subsets of LROs are dysfunctional or entirely absent in a number of hereditary syndromic disorders, including in particular the Hermansky-Pudlak syndromes. This review provides a comprehensive overview of LROs in humans and model organisms and presents our current understanding of how the products of genes that are defective in heritable diseases impact their formation, motility and ultimate secretion.
Collapse
Affiliation(s)
- Shanna L Bowman
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jing Bi-Karchin
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Linh Le
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael S Marks
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania.,Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
45
|
Zhou F, Xiong X, Li S, Liang J, Zhang X, Tian M, Li X, Gao M, Tang L, Li Y. Enhanced autophagic retrograde axonal transport by dynein intermediate chain upregulation improves Aβ clearance and cognitive function in APP/PS1 double transgenic mice. Aging (Albany NY) 2020; 12:12142-12159. [PMID: 32584265 PMCID: PMC7343509 DOI: 10.18632/aging.103382] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 05/23/2020] [Indexed: 02/07/2023]
Abstract
Autophagosome accumulation is observed in the distal axons of Alzheimer disease (AD) patients and AD animal models, suggesting that deficient retrograde transport and impaired autophagic clearance of beta-amyloid (A β) contribute to AD pathogenesis. Expression of the retrograde axonal transport-related protein dynein intermediate chain (DIC) is also reduced in AD patients, but the contributions of DIC to AD pathology remain elusive. This study investigated the effects of DIC expression levels on cognitive function, autophagosome axonal transport, and A β clearance in the APP/PS1 double transgenic mouse model of AD. Autophagic activity was enhanced in the hippocampus of young (3-month-old) AD mice, as evidenced by greater expression of autophagosome markers, lysosome markers, axonal transport motors (including DIC), and dynein regulatory proteins. The expression levels of autophagosome markers remained elevated, whereas those of autophagic and axonal transport proteins decreased progressively with age, accompanied by spatial learning and memory deficits, axonal autophagosome accumulation, and A β deposition. Knockdown of DIC exacerbated while overexpression improved axonal transport, autophagosome maturation, Aβ clearance, and spatial learning and memory in aged AD mice. Our study provides evidence that age-dependent failure of axonal autophagic flux contributes to AD-associated neuropathology and cognitive deficits, suggesting DIC as a potential therapeutic target for AD.
Collapse
Affiliation(s)
- Fanlin Zhou
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China.,Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Xiaomin Xiong
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Shijie Li
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Jie Liang
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xiong Zhang
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Mingyuan Tian
- Department of Endocrinology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Xiaoju Li
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Minna Gao
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Li Tang
- Department of Pathophysiology, Chongqing Medical University, Chongqing 400016, China
| | - Yu Li
- Institute of Neuroscience, School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China.,Key Laboratory for Biorheological Science and Technology of Ministry of Education, Chongqing University, Chongqing University Cancer Hospital, Chongqing 400044, China
| |
Collapse
|
46
|
Tempes A, Weslawski J, Brzozowska A, Jaworski J. Role of dynein-dynactin complex, kinesins, motor adaptors, and their phosphorylation in dendritogenesis. J Neurochem 2020; 155:10-28. [PMID: 32196676 DOI: 10.1111/jnc.15010] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 02/24/2020] [Accepted: 03/13/2020] [Indexed: 12/21/2022]
Abstract
One of the characteristic features of different classes of neurons that is vital for their proper functioning within neuronal networks is the shape of their dendritic arbors. To properly develop dendritic trees, neurons need to accurately control the intracellular transport of various cellular cargo (e.g., mRNA, proteins, and organelles). Microtubules and motor proteins (e.g., dynein and kinesins) that move along microtubule tracks play an essential role in cargo sorting and transport to the most distal ends of neurons. Equally important are motor adaptors, which may affect motor activity and specify cargo that is transported by the motor. Such transport undergoes very dynamic fine-tuning in response to changes in the extracellular environment and synaptic transmission. Such regulation is achieved by the phosphorylation of motors, motor adaptors, and cargo, among other mechanisms. This review focuses on the contribution of the dynein-dynactin complex, kinesins, their adaptors, and the phosphorylation of these proteins in the formation of dendritic trees by maturing neurons. We primarily review the effects of the motor activity of these proteins in dendrites on dendritogenesis. We also discuss less anticipated mechanisms that contribute to dendrite growth, such as dynein-driven axonal transport and non-motor functions of kinesins.
Collapse
Affiliation(s)
- Aleksandra Tempes
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Jan Weslawski
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Agnieszka Brzozowska
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Jacek Jaworski
- Laboratory of Molecular and Cellular Neurobiology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| |
Collapse
|
47
|
Regulation of BDNF-TrkB Signaling and Potential Therapeutic Strategies for Parkinson's Disease. J Clin Med 2020; 9:jcm9010257. [PMID: 31963575 PMCID: PMC7019526 DOI: 10.3390/jcm9010257] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/19/2019] [Accepted: 01/15/2020] [Indexed: 12/20/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and its receptor tropomyosin-related kinase receptor type B (TrkB) are widely distributed in multiple regions of the human brain. Specifically, BDNF/TrkB is highly expressed and activated in the dopaminergic neurons of the substantia nigra and plays a critical role in neurophysiological processes, including neuro-protection and maturation and maintenance of neurons. The activation as well as dysfunction of the BDNF-TrkB pathway are associated with neurodegenerative diseases. The expression of BDNF/TrkB in the substantia nigra is significantly reduced in Parkinson's Disease (PD) patients. This review summarizes recent progress in the understanding of the cellular and molecular roles of BNDF/TrkB signaling and its isoform, TrkB.T1, in Parkinson's disease. We have also discussed the effects of current therapies on BDNF/TrkB signaling in Parkinson's disease patients and the mechanisms underlying the mutation-mediated acquisition of resistance to therapies for Parkinson's disease.
Collapse
|
48
|
SIPA1L2 controls trafficking and local signaling of TrkB-containing amphisomes at presynaptic terminals. Nat Commun 2019; 10:5448. [PMID: 31784514 PMCID: PMC6884526 DOI: 10.1038/s41467-019-13224-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 10/28/2019] [Indexed: 12/31/2022] Open
Abstract
Amphisomes are organelles of the autophagy pathway that result from the fusion of autophagosomes with late endosomes. While biogenesis of autophagosomes and late endosomes occurs continuously at axon terminals, non-degradative roles of autophagy at boutons are barely described. Here, we show that in neurons BDNF/TrkB traffick in amphisomes that signal locally at presynaptic boutons during retrograde transport to the soma. This is orchestrated by the Rap GTPase-activating (RapGAP) protein SIPA1L2, which connects TrkB amphisomes to a dynein motor. The autophagosomal protein LC3 regulates RapGAP activity of SIPA1L2 and controls retrograde trafficking and local signaling of TrkB. Following induction of presynaptic plasticity, amphisomes dissociate from dynein at boutons enabling local signaling and promoting transmitter release. Accordingly, sipa1l2 knockout mice show impaired BDNF-dependent presynaptic plasticity. Taken together, the data suggest that in hippocampal neurons, TrkB-signaling endosomes are in fact amphisomes that during retrograde transport have local signaling capacity in the context of presynaptic plasticity. There is growing evidence that autophagy might serve specialized functions in neurons besides its role in protein homeostasis. In this study, authors demonstrate that axonal retrograde transport of BDNF/TrkB in neuronal amphisomes is involved in plasticity-relevant local signaling at presynaptic boutons and that SIPA1L2, a member of the SIPA1L family of neuronal RapGAPs, associates via LC3b to TrkB-containing amphisomes to regulate its motility and signaling at the axon terminals
Collapse
|
49
|
Tuttle A, Drerup CM, Marra M, McGraw H, Nechiporuk AV. Retrograde Ret signaling controls sensory pioneer axon outgrowth. eLife 2019; 8:46092. [PMID: 31476133 PMCID: PMC6718271 DOI: 10.7554/elife.46092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022] Open
Abstract
The trafficking mechanisms and transcriptional targets downstream of long-range neurotrophic factor ligand/receptor signaling that promote axon growth are incompletely understood. Zebrafish carrying a null mutation in a neurotrophic factor receptor, Ret, displayed defects in peripheral sensory axon growth cone morphology and dynamics. Ret receptor was highly enriched in sensory pioneer neurons and Ret51 isoform was required for pioneer axon outgrowth. Loss-of-function of a cargo adaptor, Jip3, partially phenocopied Ret axonal defects, led to accumulation of activated Ret in pioneer growth cones, and reduced retrograde Ret51 transport. Jip3 and Ret51 were also retrogradely co-transported, ultimately suggesting Jip3 is a retrograde adapter of active Ret51. Finally, loss of Ret reduced transcription and growth cone localization of Myosin-X, an initiator of filopodial formation. These results show a specific role for Ret51 in pioneer axon growth, and suggest a critical role for long-range retrograde Ret signaling in regulating growth cone dynamics through downstream transcriptional changes.
Collapse
Affiliation(s)
- Adam Tuttle
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, United States
| | - Catherine M Drerup
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, United States
| | - Molly Marra
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, United States.,Neuroscience Graduate Program, Oregon Health & Science University, Portland, United States
| | - Hillary McGraw
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, United States
| | - Alex V Nechiporuk
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, United States
| |
Collapse
|
50
|
Liu Y, Zhang L, Li W, Li Y, Liu J, Zhang S, Pin G, Song S, Ray PF, Arnoult C, Cho C, Garcia-Reyes B, Knippschild U, Strauss JF, Zhang Z. The sperm-associated antigen 6 interactome and its role in spermatogenesis. Reproduction 2019; 158:181-197. [PMID: 31146259 PMCID: PMC7368494 DOI: 10.1530/rep-18-0522] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 05/30/2019] [Indexed: 12/18/2022]
Abstract
Mammalian SPAG6, the orthologue of Chlamydomonas reinhardtii PF16, is a component of the central apparatus of the '9 + 2' axoneme that controls ciliary/flagellar motility, including sperm motility. Recent studies revealed that SPAG6 has functions beyond its role in the central apparatus. Hence, we reexamined the role of SPAG6 in male fertility. In wild-type mice, SPAG6 was present in cytoplasmic vesicles in spermatocytes, the acrosome of round and elongating spermatids and the manchette of elongating spermatids. Spag6-deficient testes showed abnormal spermatogenesis, with abnormalities in male germ cell morphology consistent with the multi-compartment pattern of SPAG6 localization. The armadillo repeat domain of mouse SPAG6 was used as a bait in a yeast two-hybrid screen, and several proteins with diverse functions appeared multiple times, including Snapin, SPINK2 and COPS5. Snapin has a similar localization to SPAG6 in male germ cells, and SPINK2, a key protein in acrosome biogenesis, was dramatically reduced in Spag6-deficient mice which have defective acrosomes. SPAG16L, another SPAG6-binding partner, lost its localization to the manchette in Spag6-deficient mice. Our findings demonstrate that SPAG6 is a multi-functional protein that not only regulates sperm motility, but also plays roles in spermatogenesis in multiple cellular compartments involving multiple protein partners.
Collapse
Affiliation(s)
- Yunhao Liu
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Ling Zhang
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Wei Li
- Department of Physiology, Wayne State University, Detroit, MI, 48201
| | - Yuhong Li
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Junpin Liu
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Shiyang Zhang
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Guanglun Pin
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Shizhen Song
- School of Public Health, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, Hubei, 430065
| | - Pierre F Ray
- Genetic Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| | - Christophe Arnoult
- Genetic Epigenetic and Therapies of Infertility, Institute for Advanced Biosciences, Inserm U1209, CNRS UMR 5309, Université Grenoble Alpes, Grenoble, France
| | - Chunghee Cho
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju 500-712, Korea
| | - Balbina Garcia-Reyes
- Department of General and Visceral Surgery, Ulm University, Albert-Einstein-Allee 23, D-89081, Ulm, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Ulm University, Albert-Einstein-Allee 23, D-89081, Ulm, Germany
| | - Jerome F. Strauss
- Department of Obstetrics/Gynecology, Virginia Commonwealth University, Richmond, VA, 23298
| | - Zhibing Zhang
- Department of Physiology, Wayne State University, Detroit, MI, 48201
- Department of Obstetrics/Gynecology, Wayne State University, Detroit, MI, 48201
| |
Collapse
|