1
|
Khan R, Rehman NU, Thangappan R, Saritha A, Sangaraju S. Advances in Ga 2O 3-based memristor devices, modeling, properties, and applications for low power neuromorphic computing. NANOSCALE 2025; 17:11152-11190. [PMID: 40230314 DOI: 10.1039/d4nr04865b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
About a decade ago, gallium oxide (Ga2O3) was found to be a very attractive ultrawide-bandgap (4.6-4.9 eV) semiconductor for next-generation low-power devices. Ga2O3 materials have attracted a lot of scientific and technical interest because of their outstanding properties and numerous application opportunities in the field of semiconductor based memristor technology. This review is focused on Ga2O3 thin-film memristors for smart technologies. The capacitance behavior of memristors is very important for adapting nonlinear memristor responses. Also, this comprehensive review explores in depth the ideas, device construction, and manufacturing procedures for Ga2O3-based memristor devices. To improve the device's behavior and performance improvement, a detailed analysis of many modeling and simulation techniques is given. Also, advanced characterization techniques, such as electrical, structural, and thermal evaluations, for studying artificial optoelectronic synaptic characteristics, which are important for use in computational neuroscience, are discussed in detail. The synaptic activities revealed that learning and memory processes were aided by potentiation and depression similar to those found in biological synapses. The most notable accomplishment is the realization of quaternary memory storage in a single device. This idea is supported by empirical evidence and simulations, which demonstrate the possibility of storing and maintaining multiple memory states. This study establishes oxide semiconductor memristors as a doorway to quaternary memory storage and improved synaptic functioning, paving the way for optoelectronic synaptic devices with greater memory capacity.
Collapse
Affiliation(s)
- Rajwali Khan
- National Water and Energy Center, United Arab Emirates University, Al Ain, 15551, United Arab Emirates.
- Department of Physics, University of Lakki Marwat, Lakki Marwat, 28530, KP, Pakistan
| | - Naveed Ur Rehman
- Department of Physics, University of Lakki Marwat, Lakki Marwat, 28530, KP, Pakistan
| | - R Thangappan
- Advanced Functional Materials for Energy Research Lab, Department of Energy Science & Technology, Periyar University, Salem-636011, Tamil Nadu, India
| | - Appukuttan Saritha
- Department of Chemistry, Amrita Vishwa Vidyapeetham, Amritapuri, Kollam, Kerala, India
| | - Sambasivam Sangaraju
- National Water and Energy Center, United Arab Emirates University, Al Ain, 15551, United Arab Emirates.
| |
Collapse
|
2
|
Amin M, Gragnoli C. Secondary analysis of GenRED data (Genetics of Recurrent Early-Onset major Depression) using MERLIN. Eur Arch Psychiatry Clin Neurosci 2025:10.1007/s00406-025-02014-y. [PMID: 40285827 DOI: 10.1007/s00406-025-02014-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 04/05/2025] [Indexed: 04/29/2025]
Abstract
The familial relevance of depression is a critical aspect, with evidence suggesting a substantial hereditary component. Investigating the inheritance patterns within families can provide valuable insights into the genetic underpinnings of depression. To gain a new perspective on the genetics underpinning of depression, we conducted a novel analysis of the Genetics of Recurrent Early-onset Depression (GenRED) dataset including 683 U.S. Caucasian families, each with one proband with recurrent early-onset major depression and at least one sibling with depression. Using the MERLIN tool, we identified 37 genomic markers with nominal significance linkage to early-onset recurrent depression. The findings differ from prior analyses of GenRED using a different analytical tool. Our findings highlight the variability and potential discrepancies that can arise from using different analytical tools on the same dataset. Given the chance of microsatellites undergoing duplication, mutations, and amplification errors, our two-point analysis can be more robust compared to the previous multipoint analysis. Also, one limitation is genetic admixture; studies with more homogenous ethnic groups are warranted.
Collapse
Affiliation(s)
- Mutaz Amin
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Al-Neelain University, Khartoum, 11121, Sudan
| | - Claudia Gragnoli
- Division of Endocrinology, Department of Medicine, Creighton University School of Medicine, Omaha, NE, 68124, USA.
- Division of Endocrinology, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
- Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, 17033, USA.
- Klinik für Endokrinologie, Diabetologie und Klinische Ernährung, Universitätsspital Zürich, Zürich, 8091, Switzerland.
- Molecular Biology Laboratory, Bios Biotech Multi-Diagnostic Health Center, Rome, 00197, Italy.
| |
Collapse
|
3
|
Tymanskyj SR, Escorce A, Karthikeyan S, Ma L. Optogenetic control of receptor-mediated growth cone dynamics in neurons. Mol Biol Cell 2025; 36:br5. [PMID: 39705378 PMCID: PMC11809317 DOI: 10.1091/mbc.e23-07-0268] [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: 07/26/2023] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/22/2024] Open
Abstract
Development of neuronal connections is spatially and temporally controlled by extracellular cues which often activate their cognate cell surface receptors and elicit localized cellular responses. Here, we demonstrate the use of an optogenetic tool to activate receptor signaling locally to induce actin-mediated growth cone remodeling in neurons. Based on the light-induced interaction between Cryptochrome 2 (CRY2) and CIB1, we generated a bicistronic vector to co-expresses CRY2 fused to the intracellular domain of a guidance receptor and a membrane-anchored CIB1. When expressed in primary neurons, activation of the growth inhibitory PlexA4 receptor induced growth cone collapse, while activation of the growth stimulating TrkA receptor increased growth cone size. Moreover, local activation of either receptor not only elicited the predicted response in light-activated growth cones but also an opposite response in neighboring no-light-exposed growth cones of the same neuron. Finally, this tool was used to reorient growth cones toward or away from the site of light activation and to stimulate local actin polymerization for branch initiation along axonal shafts. These studies demonstrate the use of an optogenetic tool for precise spatial and temporal control of receptor signaling in neurons and support its future application in investigating cellular mechanisms of neuronal development and plasticity.
Collapse
Affiliation(s)
- Stephen R. Tymanskyj
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107
| | - Althea Escorce
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107
| | - Siddharth Karthikeyan
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107
| | - Le Ma
- Department of Neuroscience, Jefferson Center for Synaptic Biology, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107
| |
Collapse
|
4
|
Nakagawa N. The neuronal Golgi in neural circuit formation and reorganization. Front Neural Circuits 2024; 18:1504422. [PMID: 39703196 PMCID: PMC11655203 DOI: 10.3389/fncir.2024.1504422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024] Open
Abstract
The Golgi apparatus is a central hub in the intracellular secretory pathway. By positioning in the specific intracellular region and transporting materials to spatially restricted compartments, the Golgi apparatus contributes to the cell polarity establishment and morphological specification in diverse cell types. In neurons, the Golgi apparatus mediates several essential steps of initial neural circuit formation during early brain development, such as axon-dendrite polarization, neuronal migration, primary dendrite specification, and dendritic arbor elaboration. Moreover, neuronal activity-dependent remodeling of the Golgi structure enables morphological changes in neurons, which provides the cellular basis of circuit reorganization during postnatal critical period. In this review, I summarize recent findings illustrating the unique Golgi positioning and its developmental dynamics in various types of neurons. I also discuss the upstream regulators for the Golgi positioning in neurons, and functional roles of the Golgi in neural circuit formation and reorganization. Elucidating how Golgi apparatus sculpts neuronal connectivity would deepen our understanding of the cellular/molecular basis of neural circuit development and plasticity.
Collapse
Affiliation(s)
- Naoki Nakagawa
- Laboratory of Mammalian Neural Circuits, National Institute of Genetics, Mishima, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Japan
| |
Collapse
|
5
|
Gay SM, Chartampila E, Lord JS, Grizzard S, Maisashvili T, Ye M, Barker NK, Mordant AL, Mills CA, Herring LE, Diering GH. Developing forebrain synapses are uniquely vulnerable to sleep loss. Proc Natl Acad Sci U S A 2024; 121:e2407533121. [PMID: 39441640 PMCID: PMC11536182 DOI: 10.1073/pnas.2407533121] [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: 04/15/2024] [Accepted: 09/19/2024] [Indexed: 10/25/2024] Open
Abstract
Sleep is an essential behavior that supports lifelong brain health and cognition. Neuronal synapses are a major target for restorative sleep function and a locus of dysfunction in response to sleep deprivation (SD). Synapse density is highly dynamic during development, becoming stabilized with maturation to adulthood, suggesting sleep exerts distinct synaptic functions between development and adulthood. Importantly, problems with sleep are common in neurodevelopmental disorders including autism spectrum disorder (ASD). Moreover, early life sleep disruption in animal models causes long-lasting changes in adult behavior. Divergent plasticity engaged during sleep necessarily implies that developing and adult synapses will show differential vulnerability to SD. To investigate distinct sleep functions and mechanisms of vulnerability to SD across development, we systematically examined the behavioral and molecular responses to acute SD between juvenile (P21 to P28), adolescent (P42 to P49), and adult (P70 to P100) mice of both sexes. Compared to adults, juveniles lack robust adaptations to SD, precipitating cognitive deficits in the novel object recognition task. Subcellular fractionation, combined with proteome and phosphoproteome analysis revealed the developing synapse is profoundly vulnerable to SD, whereas adults exhibit comparative resilience. SD in juveniles, and not older mice, aberrantly drives induction of synapse potentiation, synaptogenesis, and expression of perineuronal nets. Our analysis further reveals the developing synapse as a putative node of convergence between vulnerability to SD and ASD genetic risk. Together, our systematic analysis supports a distinct developmental function of sleep and reveals how sleep disruption impacts key aspects of brain development, providing insights for ASD susceptibility.
Collapse
Affiliation(s)
- Sean M. Gay
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Elissavet Chartampila
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Julia S. Lord
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Sawyer Grizzard
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Tekla Maisashvili
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Michael Ye
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Natalie K. Barker
- University of North Carolina Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Angie L. Mordant
- University of North Carolina Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - C. Allie Mills
- University of North Carolina Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Laura E. Herring
- University of North Carolina Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
| | - Graham H. Diering
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Neuroscience Center, University of North Carolina at Chapel Hill, Chapel Hill, NC27599
- Carolina Institute for Developmental Disabilities, Carrboro, NC27510
| |
Collapse
|
6
|
Dasgupta A, Reagor CC, Paik SP, Snow LM, Jacobo A, Hudspeth AJ. Semaphorin7A patterns neural circuitry in the lateral line of the zebrafish. eLife 2024; 12:RP89926. [PMID: 39133541 PMCID: PMC11318972 DOI: 10.7554/elife.89926] [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: 08/13/2024] Open
Abstract
In a developing nervous system, axonal arbors often undergo complex rearrangements before neural circuits attain their final innervation topology. In the lateral line sensory system of the zebrafish, developing sensory axons reorganize their terminal arborization patterns to establish precise neural microcircuits around the mechanosensory hair cells. However, a quantitative understanding of the changes in the sensory arbor morphology and the regulators behind the microcircuit assembly remain enigmatic. Here, we report that Semaphorin7A (Sema7A) acts as an important mediator of these processes. Utilizing a semi-automated three-dimensional neurite tracing methodology and computational techniques, we have identified and quantitatively analyzed distinct topological features that shape the network in wild-type and Sema7A loss-of-function mutants. In contrast to those of wild-type animals, the sensory axons in Sema7A mutants display aberrant arborizations with disorganized network topology and diminished contacts to hair cells. Moreover, ectopic expression of a secreted form of Sema7A by non-hair cells induces chemotropic guidance of sensory axons. Our findings propose that Sema7A likely functions both as a juxtracrine and as a secreted cue to pattern neural circuitry during sensory organ development.
Collapse
Affiliation(s)
- Agnik Dasgupta
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller UniversityNew York CityUnited States
| | - Caleb C Reagor
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller UniversityNew York CityUnited States
- Tri-Institutional PhD Program in Computational Biology and MedicineNew YorkUnited States
| | - Sang Peter Paik
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller UniversityNew York CityUnited States
| | - Lauren M Snow
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller UniversityNew York CityUnited States
| | - Adrian Jacobo
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller UniversityNew York CityUnited States
- Chan Zuckerberg Biohub San FranciscoSan FranciscoUnited States
| | - AJ Hudspeth
- Howard Hughes Medical Institute and Laboratory of Sensory Neuroscience, The Rockefeller UniversityNew York CityUnited States
| |
Collapse
|
7
|
Chitre K, Kairamkonda S, Dwivedi MK, Yadav S, Kumar V, Sikdar SK, Nongthomba U. Beadex, the Drosophila LIM only protein, is required for the growth of the larval neuromuscular junction. J Neurophysiol 2024; 132:418-432. [PMID: 38838299 DOI: 10.1152/jn.00064.2024] [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: 02/12/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/07/2024] Open
Abstract
The appropriate growth of the neurons, accurate organization of their synapses, and successful neurotransmission are indispensable for sensorimotor activities. These processes are highly dynamic and tightly regulated. Extensive genetic, molecular, physiological, and behavioral studies have identified many molecular candidates and investigated their roles in various neuromuscular processes. In this article, we show that Beadex (Bx), the Drosophila LIM only (LMO) protein, is required for motor activities and neuromuscular growth of Drosophila. The larvae bearing Bx7, a null allele of Bx, and the RNAi-mediated neuronal-specific knockdown of Bx show drastically reduced crawling behavior, a diminished synaptic span of the neuromuscular junctions (NMJs) and an increased spontaneous neuronal firing with altered motor patterns in the central pattern generators (CPGs). Microarray studies identified multiple targets of Beadex that are involved in different cellular and molecular pathways, including those associated with the cytoskeleton and mitochondria that could be responsible for the observed neuromuscular defects. With genetic interaction studies, we further show that Highwire (Hiw), a negative regulator of synaptic growth at the NMJs, negatively regulates Bx, as the latter's deficiency was able to rescue the phenotype of the Hiw null mutant, HiwDN. Thus, our data indicate that Beadex functions downstream of Hiw to regulate the larval synaptic growth and physiology.NEW & NOTEWORTHY A novel role for Beadex (Bx) regulates the larval neuromuscular junction (NMJ) structure and function in a tissue-specific manner. Bx is expressed in a subset of Toll-6-expressing neurons and is involved in regulating synaptic span and physiology, possibly through its negative interaction with Highwire (Hiw). The findings of this study provide insights into the molecular mechanisms underlying NMJ development and function and warrant further investigation to understand the role of Bx in these processes fully.
Collapse
Affiliation(s)
- Kripa Chitre
- Department of Development Biology and Genetics (DBG), Indian Institute of Science (IISc), Bangalore, India
| | - Subhash Kairamkonda
- Department of Development Biology and Genetics (DBG), Indian Institute of Science (IISc), Bangalore, India
| | - Manish Kumar Dwivedi
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal, India
| | - Saumitra Yadav
- Molecular Biophysics Unit (MBU), Indian Institute of Science (IISc), Bangalore, India
| | - Vimlesh Kumar
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER), Bhopal, India
| | - Sujit K Sikdar
- Molecular Biophysics Unit (MBU), Indian Institute of Science (IISc), Bangalore, India
| | - Upendra Nongthomba
- Department of Development Biology and Genetics (DBG), Indian Institute of Science (IISc), Bangalore, India
| |
Collapse
|
8
|
Gauberg J, Moreno KB, Jayaraman K, Abumeri S, Jenkins S, Salazar AM, Meharena HS, Glasgow SM. Spinal motor neuron development and metabolism are transcriptionally regulated by Nuclear Factor IA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600888. [PMID: 38979382 PMCID: PMC11230388 DOI: 10.1101/2024.06.26.600888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Neural circuits governing all motor behaviors in vertebrates rely on the proper development of motor neurons and their precise targeting of limb muscles. Transcription factors are essential for motor neuron development, regulating their specification, migration, and axonal targeting. While transcriptional regulation of the early stages of motor neuron specification is well-established, much less is known about the role of transcription factors in the later stages of maturation and terminal arborization. Defining the molecular mechanisms of these later stages is critical for elucidating how motor circuits are constructed. Here, we demonstrate that the transcription factor Nuclear Factor-IA (NFIA) is required for motor neuron positioning, axonal branching, and neuromuscular junction formation. Moreover, we find that NFIA is required for proper mitochondrial function and ATP production, providing a new and important link between transcription factors and metabolism during motor neuron development. Together, these findings underscore the critical role of NFIA in instructing the assembly of spinal circuits for movement.
Collapse
|
9
|
Sharma V, Chander Sharma P, Reang J, Yadav V, Kumar Tonk R, Majeed J, Sharma K. Impact of GSK-3β and CK-1δ on Wnt signaling pathway in alzheimer disease: A dual target approach. Bioorg Chem 2024; 147:107378. [PMID: 38643562 DOI: 10.1016/j.bioorg.2024.107378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 04/02/2024] [Accepted: 04/14/2024] [Indexed: 04/23/2024]
Abstract
Alzheimer's disease (AD) is an enigmatic neurological illness that offers few treatment options. Recent exploration has highlighted the crucial connection of the Wnt signaling pathway in AD pathogenesis, shedding light on potential therapeutic targets. The present study focuses on the dual targeting of glycogen synthase kinase-3β (GSK-3β) and casein kinase-1δ (CK-1δ) within the framework of the Wnt signaling pathway as a possible technique for AD intervention. GSK-3β and CK-1δ are multifunctional kinases known for their roles in tau hyperphosphorylation, amyloid processing, and synaptic dysfunction, all of which are major hallmarks of Alzheimer's disease. They are intricately linked to Wnt signaling, which plays a pivotal part in sustaining neuronal function and synaptic plasticity. Dysregulation of the Wnt pathway in AD contributes to cognitive decline and neurodegeneration. This review delves into the molecular mechanisms by which GSK-3β and CK-1δ impact the Wnt signaling pathway, elucidating their roles in AD pathogenesis. We discuss the potential of small-molecule inhibitors along with their SAR studies along with the multi-targetd approach targeting GSK-3β and CK-1δ to modulate Wnt signaling and mitigate AD-related pathology. In summary, the dual targeting of GSK-3β and CK-1δ within the framework of the Wnt signaling pathway presents an innovative and promising avenue for future AD therapies, offering new hope for patients and caregivers in the quest to combat this challenging condition.
Collapse
Affiliation(s)
- Vinita Sharma
- Department of Pharmaceutical Chemistry, SPS, DPSRU, New Delhi, 110017, India
| | | | - Jurnal Reang
- Department of Pharmaceutical Chemistry, SPS, DPSRU, New Delhi, 110017, India
| | - Vivek Yadav
- Department of Pharmaceutical Chemistry, SPS, DPSRU, New Delhi, 110017, India
| | - Rajiv Kumar Tonk
- Department of Pharmaceutical Chemistry, SPS, DPSRU, New Delhi, 110017, India
| | - Jaseela Majeed
- School of Allied Health Sciences and Management, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
| | - Kalicharan Sharma
- Department of Pharmaceutical Chemistry, SPS, DPSRU, New Delhi, 110017, India; Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
10
|
Aiken J, Holzbaur ELF. Spastin locally amplifies microtubule dynamics to pattern the axon for presynaptic cargo delivery. Curr Biol 2024; 34:1687-1704.e8. [PMID: 38554708 PMCID: PMC11042977 DOI: 10.1016/j.cub.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 01/10/2024] [Accepted: 03/08/2024] [Indexed: 04/02/2024]
Abstract
Neurons rely on the long-range trafficking of synaptic components to form and maintain the complex neural networks that encode the human experience. With a single neuron capable of forming thousands of distinct en passant synapses along its axon, spatially precise delivery of the necessary synaptic components is paramount. How these synapses are patterned, as well as how the efficient delivery of synaptic components is regulated, remains largely unknown. Here, we reveal a novel role for the microtubule (MT)-severing enzyme spastin in locally enhancing MT polymerization to influence presynaptic cargo pausing and retention along the axon. In human neurons derived from induced pluripotent stem cells (iPSCs), we identify sites stably enriched for presynaptic components along the axon prior to the robust assembly of mature presynapses apposed by postsynaptic contacts. These sites are capable of cycling synaptic vesicles, are enriched with spastin, and are hotspots for new MT growth and synaptic vesicle precursor (SVP) pausing/retention. The disruption of neuronal spastin level or activity, by CRISPRi-mediated depletion, transient overexpression, or pharmacologic inhibition of enzymatic activity, interrupts the localized enrichment of dynamic MT plus ends and diminishes SVP accumulation. Using an innovative human heterologous synapse model, where microfluidically isolated human axons recognize and form presynaptic connections with neuroligin-expressing non-neuronal cells, we reveal that neurons deficient for spastin do not achieve the same level of presynaptic component accumulation as control neurons. We propose a model where spastin acts locally as an amplifier of MT polymerization to pattern specific regions of the axon for synaptogenesis and guide synaptic cargo delivery.
Collapse
Affiliation(s)
- Jayne Aiken
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Erika L F Holzbaur
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
11
|
Chang YC, Gao Y, Lee JY, Peng YJ, Langen J, Chang KT. Identification of secretory autophagy as a mechanism modulating activity-induced synaptic remodeling. Proc Natl Acad Sci U S A 2024; 121:e2315958121. [PMID: 38588427 PMCID: PMC11032469 DOI: 10.1073/pnas.2315958121] [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: 09/14/2023] [Accepted: 02/23/2024] [Indexed: 04/10/2024] Open
Abstract
The ability of neurons to rapidly remodel their synaptic structure and strength in response to neuronal activity is highly conserved across species and crucial for complex brain functions. However, mechanisms required to elicit and coordinate the acute, activity-dependent structural changes across synapses are not well understood, as neurodevelopment and structural plasticity are tightly linked. Here, using an RNAi screen in Drosophila against genes affecting nervous system functions in humans, we uncouple cellular processes important for synaptic plasticity and synapse development. We find mutations associated with neurodegenerative and mental health disorders are 2-times more likely to affect activity-induced synaptic remodeling than synapse development. We report that while both synapse development and activity-induced synaptic remodeling at the fly NMJ require macroautophagy (hereafter referred to as autophagy), bifurcation in the autophagy pathway differentially impacts development and synaptic plasticity. We demonstrate that neuronal activity enhances autophagy activation but diminishes degradative autophagy, thereby driving the pathway towards autophagy-based secretion. Presynaptic knockdown of Snap29, Sec22, or Rab8, proteins implicated in the secretory autophagy pathway, is sufficient to abolish activity-induced synaptic remodeling. This study uncovers secretory autophagy as a transsynaptic signaling mechanism modulating synaptic plasticity.
Collapse
Affiliation(s)
- Yen-Ching Chang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Yuan Gao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Joo Yeun Lee
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Yi-Jheng Peng
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Jennifer Langen
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Karen T. Chang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| |
Collapse
|
12
|
Gay SM, Chartampila E, Lord JS, Grizzard S, Maisashvili T, Ye M, Barker NK, Mordant AL, Mills CA, Herring LE, Diering GH. Developing forebrain synapses are uniquely vulnerable to sleep loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.06.565853. [PMID: 37986967 PMCID: PMC10659326 DOI: 10.1101/2023.11.06.565853] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Sleep is an essential behavior that supports lifelong brain health and cognition. Neuronal synapses are a major target for restorative sleep function and a locus of dysfunction in response to sleep deprivation (SD). Synapse density is highly dynamic during development, becoming stabilized with maturation to adulthood, suggesting sleep exerts distinct synaptic functions between development and adulthood. Importantly, problems with sleep are common in neurodevelopmental disorders including autism spectrum disorder (ASD). Moreover, early life sleep disruption in animal models causes long lasting changes in adult behavior. Different plasticity engaged during sleep necessarily implies that developing and adult synapses will show differential vulnerability to SD. To investigate distinct sleep functions and mechanisms of vulnerability to SD across development, we systematically examined the behavioral and molecular responses to acute SD between juvenile (P21-28), adolescent (P42-49) and adult (P70-100) mice of both sexes. Compared to adults, juveniles lack robust adaptations to SD, precipitating cognitive deficits in the novel object recognition test. Subcellular fractionation, combined with proteome and phosphoproteome analysis revealed the developing synapse is profoundly vulnerable to SD, whereas adults exhibit comparative resilience. SD in juveniles, and not older mice, aberrantly drives induction of synapse potentiation, synaptogenesis, and expression of peri-neuronal nets. Our analysis further reveals the developing synapse as a convergent node between vulnerability to SD and ASD genetic risk. Together, our systematic analysis supports a distinct developmental function of sleep and reveals how sleep disruption impacts key aspects of brain development, providing mechanistic insights for ASD susceptibility.
Collapse
|
13
|
Ko MY, Park H, Chon S, Lee B, Cha S, Hyun S, Ka M. Prenatal Di-methoxyethyl phthalate exposure impairs cortical neurogenesis and synaptic activity in the mice. Brain Pathol 2024; 34:e13221. [PMID: 37903655 PMCID: PMC10901619 DOI: 10.1111/bpa.13221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/14/2023] [Indexed: 11/01/2023] Open
Abstract
Di-methoxyethyl phthalate (DMEP) is a well-known environmentally prevalent endocrine disruptor and may be associated with neurodevelopmental disorders including attention deficit/hyperactivity disorder and intellectual disability. However, the regulatory mechanisms leading to these neurodevelopmental disorders are still poorly understood. Here, we demonstrate that prenatal DMEP exposure causes abnormal brain morphology and function in the mice. DMEP (50 mg/kg) was chronically administered to pregnant mice orally once a day starting on embryonic day 0 (E0) to breast-feeding cessation for the fetus. We found that prenatal DMEP exposure significantly reduced the number of neurons in the parietal cortex by impairing neurogenesis and gliogenesis during the developing cortex. Moreover, we found that prenatal DMEP exposure impaired dendritic spine architectures and synaptic activity in the parietal cortex. Finally, prenatal DMEP exposure in mice induces hyperactivity and reduces anxiety behaviors. Altogether, our study demonstrates that prenatal DMEP exposure leads to abnormal behaviors via impairment of neurogenesis and synaptic activity.
Collapse
Affiliation(s)
- Moon Yi Ko
- Department of Advanced Toxicology ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Heejin Park
- Department of Advanced Toxicology ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
- Collage of Veterinary of MedicineJeonbuk National UniversityJeonjuRepublic of Korea
| | - Sun‐Hwa Chon
- Department of Advanced Toxicology ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Byoung‐Seok Lee
- Department of Advanced Toxicology ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Sin‐Woo Cha
- Department of Advanced Toxicology ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Sung‐Ae Hyun
- Department of Advanced Toxicology ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| | - Minhan Ka
- Department of Advanced Toxicology ResearchKorea Institute of ToxicologyDaejeonRepublic of Korea
| |
Collapse
|
14
|
Chang YC, Gao Y, Lee JY, Langen J, Chang KT. Identification of secretory autophagy as a novel mechanism modulating activity-induced synaptic remodeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.06.560931. [PMID: 38328055 PMCID: PMC10849665 DOI: 10.1101/2023.10.06.560931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
The ability of neurons to rapidly remodel their synaptic structure and strength in response to neuronal activity is highly conserved across species and crucial for complex brain functions. However, mechanisms required to elicit and coordinate the acute, activity-dependent structural changes across synapses are not well understood. Here, using an RNAi screen in Drosophila against genes affecting nervous system functions in humans, we uncouple cellular processes important for synaptic plasticity from synapse development. We find mutations associated with neurodegenerative and mental health disorders are 2-times more likely to affect activity-induced synaptic remodeling than synapse development. We further demonstrate that neuronal activity stimulates autophagy activation but diminishes degradative autophagy, thereby driving the pathway towards autophagy-based secretion. Presynaptic knockdown of Snap29, Sec22, or Rab8, proteins implicated in the secretory autophagy pathway, is sufficient to abolish activity-induced synaptic remodeling. This study uncovers secretory autophagy as a novel trans-synaptic signaling mechanism modulating structural plasticity.
Collapse
|
15
|
Napier M, Reynolds K, Scott AL. Glial-mediated dysregulation of neurodevelopment in Fragile X Syndrome. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 173:187-215. [PMID: 37993178 DOI: 10.1016/bs.irn.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Abstract
Astrocytes are highly involved in a multitude of developmental processes that are known to be dysregulated in Fragile X Syndrome. Here, we examine these processes individually and review the roles astrocytes play in contributing to the pathology of this syndrome. As a growing area of interest in the field, new and exciting insight is continually emerging. Understanding these glial-mediated roles is imperative for elucidating the underlying molecular mechanisms at play, not only in Fragile X Syndrome, but also other ASD-related disorders. Understanding these roles will be central to the future development of effective, clinically-relevant treatments of these disorders.
Collapse
Affiliation(s)
- M Napier
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - K Reynolds
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada; Department of Neuroscience, Tufts University School of Medicine, Boston, United States
| | - A L Scott
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Canada; Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada.
| |
Collapse
|
16
|
Keine C, Radulovic T, Al-Yaari M, Young SM. Confocal Imaging and 3D Reconstruction to Determine How Genetic Perturbations Impact Presynaptic Morphology at the Mouse Calyx of Held. Bio Protoc 2023; 13:e4799. [PMID: 37849785 PMCID: PMC10577601 DOI: 10.21769/bioprotoc.4799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/05/2023] [Accepted: 06/08/2023] [Indexed: 10/19/2023] Open
Abstract
Neurons communicate via synapses-specialized structures that consist of a presynaptic terminal of one neuron and a postsynaptic terminal of another. As knowledge is emerging that mutations in molecules that regulate synaptic function underpin many neurological disorders, it is crucial to elucidate the molecular mechanisms regulating synaptic function to understand synaptic strength, plasticity, modulation, and pathology, which ultimately impact neuronal circuit output and behavior. The presynaptic calyx of Held is a large glutamatergic presynaptic terminal in the auditory brainstem, which due to its accessibility and the possibility to selectively perform molecular perturbations on it, is an ideal model to study the role of presynaptic proteins in regulating synaptic function. In this protocol, we describe the use of confocal imaging and three-dimensional reconstruction of the calyx of Held to assess alterations in gross morphology following molecular perturbation. Using viral-vector delivery to perform molecular perturbations at distinct developmental time points, we provide a fast and cost-effective method to investigate how presynaptic proteins regulate gross morphology such as surface area and synapse volume throughout the lifetime of a neuronal circuit. Key features Confocal imaging and 3D reconstruction of presynaptic terminals. Used with a virus-mediated expression of mEGFP to achieve efficient, cell-type specific labeling of the presynaptic compartment. Protocol was developed with the calyx of Held but is suitable for pre- and postsynaptic compartments of various neurons across multiple mammalian and invertebrate species.
Collapse
Affiliation(s)
- Christian Keine
- Department of Anatomy and Cell Biology, University of
Iowa, Iowa City, IA, USA
- Department of Human Medicine, University of
Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Oldenburg,
Germany
| | - Tamara Radulovic
- Department of Anatomy and Cell Biology, University of
Iowa, Iowa City, IA, USA
- Department of Human Medicine, University of
Oldenburg, Oldenburg, Germany
- Research Center Neurosensory Science, Oldenburg,
Germany
| | - Mohammed Al-Yaari
- Department of Anatomy and Cell Biology, University of
Iowa, Iowa City, IA, USA
| | - Samuel M. Young
- Department of Anatomy and Cell Biology, University of
Iowa, Iowa City, IA, USA
- Department of Otolaryngology, Iowa Neuroscience
Institute, University of Iowa, Iowa City, IA, USA
| |
Collapse
|
17
|
Murphy KE, Wade SD, Sperringer JE, Mohan V, Duncan BW, Zhang EY, Pak Y, Lutz D, Schachner M, Maness PF. The L1 cell adhesion molecule constrains dendritic spine density in pyramidal neurons of the mouse cerebral cortex. Front Neuroanat 2023; 17:1111525. [PMID: 37007644 PMCID: PMC10062527 DOI: 10.3389/fnana.2023.1111525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
A novel function for the L1 cell adhesion molecule, which binds the actin adaptor protein Ankyrin was identified in constraining dendritic spine density on pyramidal neurons in the mouse neocortex. In an L1-null mouse mutant increased spine density was observed on apical but not basal dendrites of pyramidal neurons in diverse cortical areas (prefrontal cortex layer 2/3, motor cortex layer 5, visual cortex layer 4. The Ankyrin binding motif (FIGQY) in the L1 cytoplasmic domain was critical for spine regulation, as demonstrated by increased spine density and altered spine morphology in the prefrontal cortex of a mouse knock-in mutant (L1YH) harboring a tyrosine (Y) to histidine (H) mutation in the FIGQY motif, which disrupted L1-Ankyrin association. This mutation is a known variant in the human L1 syndrome of intellectual disability. L1 was localized by immunofluorescence staining to spine heads and dendrites of cortical pyramidal neurons. L1 coimmunoprecipitated with Ankyrin B (220 kDa isoform) from lysates of wild type but not L1YH forebrain. This study provides insight into the molecular mechanism of spine regulation and underscores the potential for this adhesion molecule to regulate cognitive and other L1-related functions that are abnormal in the L1 syndrome.
Collapse
Affiliation(s)
- Kelsey E. Murphy
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
- Carolina Institute of Developmental Disabilities, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
| | - Sarah D. Wade
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
- Carolina Institute of Developmental Disabilities, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
| | - Justin E. Sperringer
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
- Carolina Institute of Developmental Disabilities, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
| | - Vishwa Mohan
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
- Carolina Institute of Developmental Disabilities, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
| | - Bryce W. Duncan
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
- Carolina Institute of Developmental Disabilities, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
| | - Erin Y. Zhang
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
- Carolina Institute of Developmental Disabilities, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
| | - Yubin Pak
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
- Carolina Institute of Developmental Disabilities, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
| | - David Lutz
- Division of Neuroanatomy and Molecular Brain Research, Ruhr University-Bochum, Bochum, Germany
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscatawy, NJ, United States
| | - Patricia F. Maness
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
- Carolina Institute of Developmental Disabilities, University of North Carolina School of Medicine at Chapel Hill, Chapel Hill, NC, United States
- *Correspondence: Patricia F. Maness
| |
Collapse
|
18
|
Thapliyal S, Arendt KL, Lau AG, Chen L. Retinoic acid-gated BDNF synthesis in neuronal dendrites drives presynaptic homeostatic plasticity. eLife 2022; 11:e79863. [PMID: 36515276 PMCID: PMC9797192 DOI: 10.7554/elife.79863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022] Open
Abstract
Homeostatic synaptic plasticity is a non-Hebbian synaptic mechanism that adjusts synaptic strength to maintain network stability while achieving optimal information processing. Among the molecular mediators shown to regulate this form of plasticity, synaptic signaling through retinoic acid (RA) and its receptor, RARα, has been shown to be critically involved in the homeostatic adjustment of synaptic transmission in both hippocampus and sensory cortices. In this study, we explore the molecular mechanism through which postsynaptic RA and RARα regulates presynaptic neurotransmitter release during prolonged synaptic inactivity at mouse glutamatertic synapses. We show that RARα binds to a subset of dendritically sorted brain-derived neurotrophic factor (Bdnf) mRNA splice isoforms and represses their translation. The RA-mediated translational de-repression of postsynaptic BDNF results in the retrograde activation of presynaptic tropomyosin receptor kinase B (TrkB) receptors, facilitating presynaptic homeostatic compensation through enhanced presynaptic release. Together, our study illustrates an RA-mediated retrograde synaptic signaling pathway through which postsynaptic protein synthesis during synaptic inactivity drives compensatory changes at the presynaptic site.
Collapse
Affiliation(s)
- Shruti Thapliyal
- Departments of Neurosurgery, Neuropsychiatry and Behavioral Sciences, Stanford University School of MedicineStanfordUnited States
| | - Kristin L Arendt
- Departments of Neurosurgery, Neuropsychiatry and Behavioral Sciences, Stanford University School of MedicineStanfordUnited States
| | - Anthony G Lau
- Departments of Neurosurgery, Neuropsychiatry and Behavioral Sciences, Stanford University School of MedicineStanfordUnited States
| | - Lu Chen
- Departments of Neurosurgery, Neuropsychiatry and Behavioral Sciences, Stanford University School of MedicineStanfordUnited States
| |
Collapse
|
19
|
Untiveros G, Raskind A, Linares L, Dotti A, Strizzi L. Netrin-1 Stimulates Migration of Neogenin Expressing Aggressive Melanoma Cells. Int J Mol Sci 2022; 23:12751. [PMID: 36361539 PMCID: PMC9658020 DOI: 10.3390/ijms232112751] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 12/09/2024] Open
Abstract
Netrin-1 is a neural guidance factor that regulates migration and positioning of neural crest-derived cells during embryonic development. Depending on the type of Netrin-1 receptor expression, cells are either attracted or repulsed by Netrin-1. Postnatal expression of Netrin-1 is detected in brain, colon, liver, and kidney, which are common sites of cancer metastasis, including melanoma. Thus, understanding the dynamics between Netrin-1 and its receptors could explain the attraction of melanoma towards these Netrin-1-expressing tissues. Here, we investigate whether the Netrin-1-attractive receptor Neogenin can affect migration of melanoma cells towards a Netrin-1 source. Results from Western blot (WB) analysis show higher expression of Neogenin in aggressive compared to non-aggressive melanoma cells. Cell migration experiments show increased migration of Neogenin-expressing aggressive melanoma cells towards exogenous, soluble recombinant human Netrin-1 and towards a Netrin-1-expressing cell line. Furthermore, WB reveals ERK1/2 activation and increased N-cadherin expression in Neogenin-expressing aggressive melanoma cells treated with rhNetrin-1. Moreover, treatment with anti-Neogenin blocking antibody caused decreased migration towards Netrin-1-expressing cells and reduced ERK1/2 activity in Neogenin-expressing aggressive melanoma cells. These results suggest Neogenin may play a role during migration of melanoma cells towards Netrin-1 via ERK1/2 signaling.
Collapse
Affiliation(s)
- Gustavo Untiveros
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
| | - Aleksandr Raskind
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | - Laura Linares
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
| | - Alessandro Dotti
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | - Luigi Strizzi
- Department of Pathology, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
| |
Collapse
|
20
|
Harreguy MB, Tanvir Z, Shah E, Simprevil B, Tran TS, Haspel G. Semaphorin signaling restricts neuronal regeneration in C. elegans. Front Cell Dev Biol 2022; 10:814160. [PMID: 36325362 PMCID: PMC9618706 DOI: 10.3389/fcell.2022.814160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
Extracellular signaling proteins serve as neuronal growth cone guidance molecules during development and are well positioned to be involved in neuronal regeneration and recovery from injury. Semaphorins and their receptors, the plexins, are a family of conserved proteins involved in development that, in the nervous system, are axonal guidance cues mediating axon pathfinding and synapse formation. The Caenorhabditis elegans genome encodes for three semaphorins and two plexin receptors: the transmembrane semaphorins, SMP-1 and SMP-2, signal through their receptor, PLX-1, while the secreted semaphorin, MAB-20, signals through PLX-2. Here, we evaluate the locomotion behavior of knockout animals missing each of the semaphorins and plexins and the neuronal morphology of plexin knockout animals; we described the cellular expression pattern of the promoters of all plexins in the nervous system of C. elegans; and we evaluated their effect on the regrowth and reconnection of motoneuron neurites and the recovery of locomotion behavior following precise laser microsurgery. Regrowth and reconnection were more prevalent in the absence of each plexin, while recovery of locomotion surpassed regeneration in all genotypes.
Collapse
Affiliation(s)
- Maria B Harreguy
- New Jersey Institute of Technology, Department of Biological Sciences, Newark, NJ, United States
- Rutgers University, Department of Biological Sciences, Newark, NJ, United States
| | - Zainab Tanvir
- New Jersey Institute of Technology, Department of Biological Sciences, Newark, NJ, United States
- Rutgers University, Department of Biological Sciences, Newark, NJ, United States
| | - Esha Shah
- New Jersey Institute of Technology, Department of Biological Sciences, Newark, NJ, United States
| | - Blandine Simprevil
- New Jersey Institute of Technology, Department of Biological Sciences, Newark, NJ, United States
- City College of New York (CUNY), New York, NY, United States
| | - Tracy S Tran
- New Jersey Institute of Technology, Department of Biological Sciences, Newark, NJ, United States
- Rutgers University, Department of Biological Sciences, Newark, NJ, United States
| | - Gal Haspel
- New Jersey Institute of Technology, Department of Biological Sciences, Newark, NJ, United States
- Rutgers University, Department of Biological Sciences, Newark, NJ, United States
- Mercer University School of Medicine, Department of Biomedical Sciences, Columbus, GA, United States
- *Correspondence: Gal Haspel, ,
| |
Collapse
|
21
|
Hong J, Lu X, Wang J, Jiang M, Liu Q, Lin J, Sun W, Zhang J, Shi Y, Liu X. Triphenyl phosphate disturbs placental tryptophan metabolism and induces neurobehavior abnormal in male offspring. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 243:113978. [PMID: 36007322 DOI: 10.1016/j.ecoenv.2022.113978] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/28/2022] [Accepted: 08/13/2022] [Indexed: 06/15/2023]
Abstract
Epidemiological studies have shown that prenatal triphenyl phosphate (TPhP) exposure is related to abnormal neurobehavior in children. However, the neurodevelopmental toxicity of TPhP in mammals is limited. To study the neurodevelopmental toxicity of TPhP in mammals and investigate the underlying mechanism, we used a mouse intrauterine TPhP exposure model. We measured the inflammatory factors (IL-6, TNFα) and NFκB levels, and tryptophan metabolism in placentae, detected the fetal brain transcriptome, hippocampal neuron development and neurobehavioral in the male offspring. The results showed that the protein level of IL-6, TNFα and NFκB in the placenta of the TPhP treatment group (1, 5 mg/kg) were significantly increased. Change of the protein level of these pro-inflammatory factors in maternal serum or fetal brain was not observed. Expression of genes along tryptophan-serotonin metabolism pathway were significantly decreased. While, the concentration of 5-HT levels in the placenta or fetal brain were significantly increased. Consistent with the increased 5-HT, the Nissl body was reduced in the hippocampus of treatment group. The expression of serotonergic neuron gene markers including Tph2, Htr1A, Htr2A, Pet1 and Lmx1b in the hippocampus of treatment group was significantly decreased. The neurobehavioral test showed that TPhP decreased center time that represent anxiety-like behavior, and reduced learning and memory in male offspring. Meanwhile, expression of genes along tryptophan-kynurenine metabolism pathway were significantly increased. The result of the transcriptome analysis of fetal brain showed that the differentially expressed genes are mainly involved in the transcription regulation of DNA as a template in the nucleus, and the enriched pathways are mainly signal pathways regulated by axon guidance and neurotrophic factors, dopaminergic and cholinergic synapses, suggest that not only serotonergic neuronal was affected. Overall, this study demonstrates that TPhP has the potential to induce placental inflammatory response in the placenta, disturb placental tryptophan metabolism, compromise the neuronal development and synaptic transmission, and cause abnormal neurobehavior in male offspring.
Collapse
Affiliation(s)
- Jiabin Hong
- School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong 523-808, China
| | - Xiaoxun Lu
- School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong 523-808, China
| | - Jieyu Wang
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Mengzhu Jiang
- School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong 523-808, China
| | - Qian Liu
- School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong 523-808, China
| | - Juntong Lin
- School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong 523-808, China
| | - Wenjing Sun
- China-America Cancer Research Institute, Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Guangdong Medical University, Dongguan 523808, Guangdong, China
| | - Jing Zhang
- School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong 523-808, China
| | - Yanwei Shi
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Xiaoshan Liu
- School of Public Health, Dongguan Key Laboratory of Environmental Medicine, Guangdong Medical University, Guangdong 523-808, China.
| |
Collapse
|
22
|
Rastegar-Moghaddam SH, Bigham M, Hosseini M, Ebrahimzadeh-Bideskan A, Malvandi AM, Mohammadipour A. Grape seed extract effects on hippocampal neurogenesis, synaptogenesis and dark neurons production in old mice. Can this extract improve learning and memory in aged animals? Nutr Neurosci 2022; 25:1962-1972. [PMID: 33970818 DOI: 10.1080/1028415x.2021.1918983] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND During the elderly, hippocampal neurogenesis and synaptogenesis reduce and dark neurons (DNs) increase, leading to cognitive impairment. It is believed that natural products can protect the neural cells and system by protecting from damages or promoting regeneration. Therefore, the effects of grape seed extract (GSE) on the hippocampus of aged mice were investigated in this study. METHODS twelve old mice were divided into two groups of control and GSE. Animals in the GSE group received 300 mg/kg of GSE for eight weeks via gavage. At the end of treatment, cognition performance was evaluated by Morris water maze (MWM) and passive avoidance tests. Hippocampal neurogenesis, synaptogenesis and DNs production were evaluated with immunohistochemistry and histological evaluations on 5-micron coronal tissue sections. RESULTS The hippocampal mean number of double cortin positive cells (DCX+) per unit area, as well as synaptophysin expression in the GSE group, were significantly higher than the control group (p < 0.01). The frequency of DNs in the GSE group was lower than the control group (p < 0.05). Behavioral tests showed that GSE improves memory and learning performance. CONCLUSION Consuming GSE in the elderly can potentially alleviate the age-related reduction of hippocampal neurogenesis and synaptogenesis. It is also able to decrease hippocampal DNs production and increase memory and learning.
Collapse
Affiliation(s)
| | - Maryam Bigham
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hosseini
- Division of Neurocognitive Sciences, Psychiatry and Behavioral Sciences Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Ebrahimzadeh-Bideskan
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Abbas Mohammadipour
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
23
|
Lee HN, Hyeon SJ, Kim H, Sim KM, Kim Y, Ju J, Lee J, Wang Y, Ryu H, Seong J. Decreased FAK activity and focal adhesion dynamics impair proper neurite formation of medium spiny neurons in Huntington's disease. Acta Neuropathol 2022; 144:521-536. [PMID: 35857122 DOI: 10.1007/s00401-022-02462-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/11/2022] [Accepted: 06/25/2022] [Indexed: 11/29/2022]
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by a polyglutamine expansion in the protein huntingtin (HTT) [55]. While the final pathological consequence of HD is the neuronal cell death in the striatum region of the brain, it is still unclear how mutant HTT (mHTT) causes synaptic dysfunctions at the early stage and during the progression of HD. Here, we discovered that the basal activity of focal adhesion kinase (FAK) is severely reduced in a striatal HD cell line, a mouse model of HD, and the human post-mortem brains of HD patients. In addition, we observed with a FRET-based FAK biosensor [59] that neurotransmitter-induced FAK activation is decreased in HD striatal neurons. Total internal reflection fluorescence (TIRF) imaging revealed that the reduced FAK activity causes the impairment of focal adhesion (FA) dynamics, which further leads to the defect in filopodial dynamics causing the abnormally increased number of immature neurites in HD striatal neurons. Therefore, our results suggest that the decreased FAK and FA dynamics in HD impair the proper formation of neurites, which is crucial for normal synaptic functions [52]. We further investigated the molecular mechanism of FAK inhibition in HD and surprisingly discovered that mHTT strongly associates with phosphatidylinositol 4,5-biphosphate, altering its normal distribution at the plasma membrane, which is crucial for FAK activation [14, 60]. Therefore, our results provide a novel molecular mechanism of FAK inhibition in HD along with its pathological mechanism for synaptic dysfunctions during the progression of HD.
Collapse
Affiliation(s)
- Hae Nim Lee
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of Converging Science and Technology, Kyung Hee University, Seoul, 02453, Republic of Korea
| | - Seung Jae Hyeon
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Heejung Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of Converging Science and Technology, Kyung Hee University, Seoul, 02453, Republic of Korea
| | - Kyoung Mi Sim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Yunha Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongmin Ju
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Junghee Lee
- Department of Neurology, Boston University Alzheimer's Disease Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Yingxiao Wang
- Department of Bioengineering, University of California, San Diego, CA, 92093, USA
| | - Hoon Ryu
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
| | - Jihye Seong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea.
- Department of Converging Science and Technology, Kyung Hee University, Seoul, 02453, Republic of Korea.
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea.
| |
Collapse
|
24
|
Koppers M, Holt CE. Receptor-Ribosome Coupling: A Link Between Extrinsic Signals and mRNA Translation in Neuronal Compartments. Annu Rev Neurosci 2022; 45:41-61. [DOI: 10.1146/annurev-neuro-083021-110015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Axons receive extracellular signals that help to guide growth and synapse formation during development and to maintain neuronal function and survival during maturity. These signals relay information via cell surface receptors that can initiate local intracellular signaling at the site of binding, including local messenger RNA (mRNA) translation. Direct coupling of translational machinery to receptors provides an attractive way to activate this local mRNA translation and change the local proteome with high spatiotemporal resolution. Here, we first discuss the increasing evidence that different external stimuli trigger translation of specific subsets of mRNAs in axons via receptors and thus play a prominent role in various processes in both developing and mature neurons. We then discuss the receptor-mediated molecular mechanisms that regulate local mRNA translational with a focus on direct receptor-ribosome coupling. We advance the idea that receptor-ribosome coupling provides several advantages over other translational regulation mechanisms and is a common mechanism in cell communication. Expected final online publication date for the Annual Review of Neuroscience, Volume 45 is July 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Max Koppers
- Department of Biology, Division of Cell Biology, Neurobiology and Biophysics, Utrecht University, Utrecht, The Netherlands
| | - Christine E. Holt
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
25
|
Takei N, Yokomaku D, Yamada T, Nagano T, Kakita A, Namba H, Ushiki T, Takahashi H, Nawa H. EGF Downregulates Presynaptic Maturation and Suppresses Synapse Formation In Vitro and In Vivo. Neurochem Res 2022; 47:2632-2644. [PMID: 34984589 DOI: 10.1007/s11064-021-03524-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 11/27/2022]
Abstract
Neuronal differentiation, maturation, and synapse formation are regulated by various growth factors. Here we show that epidermal growth factor (EGF) negatively regulates presynaptic maturation and synapse formation. In cortical neurons, EGF maintained axon elongation and reduced the sizes of growth cones in culture. Furthermore, EGF decreased the levels of presynaptic molecules and number of presynaptic puncta, suggesting that EGF inhibits neuronal maturation. The reduction of synaptic sites is confirmed by the decreased frequencies of miniature EPSCs. In vivo analysis revealed that while peripherally administrated EGF decreased the levels of presynaptic molecules and numbers of synaptophysin-positive puncta in the prefrontal cortices of neonatal rats, EGF receptor inhibitors upregulated these indexes, suggesting that endogenous EGF receptor ligands suppress presynaptic maturation. Electron microscopy further revealed that EGF decreased the numbers, but not the sizes, of synaptic structures in vivo. These findings suggest that endogenous EGF and/or other EGF receptor ligands negatively modulates presynaptic maturation and synapse formation.
Collapse
Affiliation(s)
- Nobuyuki Takei
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan.
- Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata, Japan.
| | - Daisaku Yokomaku
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Takaho Yamada
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Department of Hematology, Endocrinology and Metabolism, Niigata University Faculty of Medicine, Niigata, Japan
| | - Tadasato Nagano
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
- Department of Health and Nutrition, Faculty of Human Life Studies, University of Niigata Prefecture, Niigata, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hisaaki Namba
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
- Department of Physiological Science, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
| | - Tatsuo Ushiki
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hitoshi Takahashi
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Hiroyuki Nawa
- Department of Molecular Neurobiology, Brain Research Institute, Niigata University, Niigata, Japan
- Department of Physiological Science, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
26
|
Wu J, Zhu ZY, Fan ZW, Chen Y, Yang RY, Li Y. Downregulation of EphB2 by RNA interference attenuates glial/fibrotic scar formation and promotes axon growth. Neural Regen Res 2022; 17:362-369. [PMID: 34269211 PMCID: PMC8463997 DOI: 10.4103/1673-5374.317988] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
The rapid formation of a glial/fibrotic scar is one of the main factors hampering axon growth after spinal cord injury. The bidirectional EphB2/ephrin-B2 signaling of the fibroblast-astrocyte contact-dependent interaction is a trigger for glial/fibrotic scar formation. In the present study, a new in vitro model was produced by coculture of fibroblasts and astrocytes wounded by scratching to mimic glial/fibrotic scar-like structures using an improved slide system. After treatment with RNAi to downregulate EphB2, changes in glial/fibrotic scar formation and the growth of VSC4.1 motoneuron axons were examined. Following RNAi treatment, fibroblasts and astrocytes dispersed without forming a glial/fibrotic scar-like structure. Furthermore, the expression levels of neurocan, NG2 and collagen I in the coculture were reduced, and the growth of VSC4.1 motoneuron axons was enhanced. These findings suggest that suppression of EphB2 expression by RNAi attenuates the formation of a glial/fibrotic scar and promotes axon growth. This study was approved by the Laboratory Animal Ethics Committee of Jiangsu Province, China (approval No. 2019-0506-002) on May 6, 2019.
Collapse
Affiliation(s)
- Jian Wu
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Zhen-Yu Zhu
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Zhi-Wei Fan
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Ying Chen
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Ri-Yun Yang
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| | - Yi Li
- Department of Histology and Embryology, Medical College, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
27
|
Zhang S, Jiao Z, Zhao X, Sun M, Feng X. Environmental exposure to 17β-trenbolone during adolescence inhibits social interaction in male mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 289:117710. [PMID: 34243057 DOI: 10.1016/j.envpol.2021.117710] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 06/10/2021] [Accepted: 07/02/2021] [Indexed: 06/13/2023]
Abstract
Puberty is a critical period for growth and development. This period is sensitive to external stimuli, which ultimately affects the development of nerves and the formation of social behaviour. 17β-Trenbolone (17β-TBOH) is an endocrine disrupting chemicals (EDCs), which had been widely reported in aquatic vertebrates. But there is little known about the effects of 17β-TBOH on mammals, especially on adolescent neurodevelopment. In this study, we found that 17β-TBOH acute 1 h exposure can cause the activation of the dopamine circuit in pubertal male balb/c mice. At present, there is little known about the effects of puberty exposure of endocrine disruptors on these neurons/nerve pathways. Through a series of behavioural tests, exposure to 80 μgkg-1 d-1 of 17β-TBOH during adolescence increased the anxiety-like behaviour of mice and reduced the control of wheel-running behaviour and the response of social interaction behaviour. The results of TH immunofluorescence staining showed that exposure to 17β-TBOH reduced dopamine axon growth in the medial prefrontal cortex (mPFC). In addition, the results of real-time PCR showed that exposure to 17β-TBOH not only down-regulated the expression of dopamine axon development genes, but also affected the balance of excitatory/inhibitory signals in mPFC. In this research, we reveal the effects of 17β-TBOH exposure during adolescence on mammalian behaviour and neurodevelopment, and provide a reference for studying the origin of adolescent diseases.
Collapse
Affiliation(s)
- Shaozhi Zhang
- College of Life Science, The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Zihao Jiao
- The Institute of Robotics and Automatic Information Systems, Nankai University, Tianjin, 300071, China
| | - Xin Zhao
- The Institute of Robotics and Automatic Information Systems, Nankai University, Tianjin, 300071, China
| | - Mingzhu Sun
- The Institute of Robotics and Automatic Information Systems, Nankai University, Tianjin, 300071, China
| | - Xizeng Feng
- College of Life Science, The Key Laboratory of Bioactive Materials, Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
28
|
Blockus H, Rolotti SV, Szoboszlay M, Peze-Heidsieck E, Ming T, Schroeder A, Apostolo N, Vennekens KM, Katsamba PS, Bahna F, Mannepalli S, Ahlsen G, Honig B, Shapiro L, de Wit J, Losonczy A, Polleux F. Synaptogenic activity of the axon guidance molecule Robo2 underlies hippocampal circuit function. Cell Rep 2021; 37:109828. [PMID: 34686348 PMCID: PMC8605498 DOI: 10.1016/j.celrep.2021.109828] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 07/06/2021] [Accepted: 09/22/2021] [Indexed: 01/03/2023] Open
Abstract
Synaptic connectivity within adult circuits exhibits a remarkable degree of cellular and subcellular specificity. We report that the axon guidance receptor Robo2 plays a role in establishing synaptic specificity in hippocampal CA1. In vivo, Robo2 is present and required postsynaptically in CA1 pyramidal neurons (PNs) for the formation of excitatory (E) but not inhibitory (I) synapses, specifically in proximal but not distal dendritic compartments. In vitro approaches show that the synaptogenic activity of Robo2 involves a trans-synaptic interaction with presynaptic Neurexins, as well as binding to its canonical extracellular ligand Slit. In vivo 2-photon Ca2+ imaging of CA1 PNs during spatial navigation in awake behaving mice shows that preventing Robo2-dependent excitatory synapse formation cell autonomously during development alters place cell properties of adult CA1 PNs. Our results identify a trans-synaptic complex linking the establishment of synaptic specificity to circuit function.
Collapse
Affiliation(s)
- Heike Blockus
- Department of Neuroscience, Columbia University, New York, NY 10027, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Sebi V Rolotti
- Department of Neuroscience, Columbia University, New York, NY 10027, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Miklos Szoboszlay
- Department of Neuroscience, Columbia University, New York, NY 10027, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Eugénie Peze-Heidsieck
- Department of Neuroscience, Columbia University, New York, NY 10027, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Tiffany Ming
- Department of Neuroscience, Columbia University, New York, NY 10027, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Anna Schroeder
- VIB Center for Brain and Disease Research, Herestraat 49, 3000 Leuven, Belgium; Department of Neurosciences, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Nuno Apostolo
- VIB Center for Brain and Disease Research, Herestraat 49, 3000 Leuven, Belgium; Department of Neurosciences, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Kristel M Vennekens
- VIB Center for Brain and Disease Research, Herestraat 49, 3000 Leuven, Belgium; Department of Neurosciences, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Phinikoula S Katsamba
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Fabiana Bahna
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Seetha Mannepalli
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Goran Ahlsen
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA
| | - Barry Honig
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA; Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Lawrence Shapiro
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA
| | - Joris de Wit
- VIB Center for Brain and Disease Research, Herestraat 49, 3000 Leuven, Belgium; Department of Neurosciences, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Attila Losonczy
- Department of Neuroscience, Columbia University, New York, NY 10027, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA.
| | - Franck Polleux
- Department of Neuroscience, Columbia University, New York, NY 10027, USA; Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA; Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA.
| |
Collapse
|
29
|
Bang S, Hwang KS, Jeong S, Cho IJ, Choi N, Kim J, Kim HN. Engineered neural circuits for modeling brain physiology and neuropathology. Acta Biomater 2021; 132:379-400. [PMID: 34157452 DOI: 10.1016/j.actbio.2021.06.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/16/2021] [Accepted: 06/14/2021] [Indexed: 12/14/2022]
Abstract
The neural circuits of the central nervous system are the regulatory pathways for feeling, motion control, learning, and memory, and their dysfunction is closely related to various neurodegenerative diseases. Despite the growing demand for the unraveling of the physiology and functional connectivity of the neural circuits, their fundamental investigation is hampered because of the inability to access the components of neural circuits and the complex microenvironment. As an alternative approach, in vitro human neural circuits show principles of in vivo human neuronal circuit function. They allow access to the cellular compartment and permit real-time monitoring of neural circuits. In this review, we summarize recent advances in reconstituted in vitro neural circuits using engineering techniques. To this end, we provide an overview of the fabrication techniques and methods for stimulation and measurement of in vitro neural circuits. Subsequently, representative examples of in vitro neural circuits are reviewed with a particular focus on the recapitulation of structures and functions observed in vivo, and we summarize their application in the study of various brain diseases. We believe that the in vitro neural circuits can help neuroscience and the neuropharmacology. STATEMENT OF SIGNIFICANCE: Despite the growing demand to unravel the physiology and functional connectivity of the neural circuits, the studies on the in vivo neural circuits are frequently limited due to the poor accessibility. Furthermore, single neuron-based analysis has an inherent limitation in that it does not reflect the full spectrum of the neural circuit physiology. As an alternative approach, in vitro engineered neural circuit models have arisen because they can recapitulate the structural and functional characteristics of in vivo neural circuits. These in vitro neural circuits allow the mimicking of dysregulation of the neural circuits, including neurodegenerative diseases and traumatic brain injury. Emerging in vitro engineered neural circuits will provide a better understanding of the (patho-)physiology of neural circuits.
Collapse
Affiliation(s)
- Seokyoung Bang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Kyeong Seob Hwang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Sohyeon Jeong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Il-Joo Cho
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea; School of Electrical and Electronics Engineering, Yonsei University, Seoul 03722, Republic of Korea; Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea.
| | - Jongbaeg Kim
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea.
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea.
| |
Collapse
|
30
|
Paredes-Redondo A, Harley P, Maniati E, Ryan D, Louzada S, Meng J, Kowala A, Fu B, Yang F, Liu P, Marino S, Pourquié O, Muntoni F, Wang J, Lieberam I, Lin YY. Optogenetic modeling of human neuromuscular circuits in Duchenne muscular dystrophy with CRISPR and pharmacological corrections. SCIENCE ADVANCES 2021; 7:eabi8787. [PMID: 34516770 PMCID: PMC8442926 DOI: 10.1126/sciadv.abi8787] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 07/20/2021] [Indexed: 05/13/2023]
Abstract
Duchenne muscular dystrophy (DMD) is caused by dystrophin gene mutations leading to skeletal muscle weakness and wasting. Dystrophin is enriched at the neuromuscular junction (NMJ), but how NMJ abnormalities contribute to DMD pathogenesis remains unclear. Here, we combine transcriptome analysis and modeling of DMD patient-derived neuromuscular circuits with CRISPR-corrected isogenic controls in compartmentalized microdevices. We show that NMJ volumes and optogenetic motor neuron–stimulated myofiber contraction are compromised in DMD neuromuscular circuits, which can be rescued by pharmacological inhibition of TGFβ signaling, an observation validated in a 96-well human neuromuscular circuit coculture assay. These beneficial effects are associated with normalization of dysregulated gene expression in DMD myogenic transcriptomes affecting NMJ assembly (e.g., MUSK) and axon guidance (e.g., SLIT2 and SLIT3). Our study provides a new human microphysiological model for investigating NMJ defects in DMD and assessing candidate drugs and suggests that enhancing neuromuscular connectivity may be an effective therapeutic strategy.
Collapse
Affiliation(s)
- Amaia Paredes-Redondo
- Centre for Genomics and Child Health, Blizard
Institute, Barts and the London School of Medicine and Dentistry, Queen Mary
University of London, 4 Newark Street, London E1 2AT, UK
- Stem Cell Laboratory, National Bowel Research Centre,
Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen
Mary University of London, 2 Newark Street, London E1 2AT, UK
- Centre for Predictive in vitro Model, Queen Mary
University of London, Mile End Road, London E1 4NS, UK
| | - Peter Harley
- Centre for Stem Cells and Regenerative Medicine, MRC
Centre for Neurodevelopmental Disorders, and Centre for Developmental
Neurobiology, King’s College London, London, UK
| | - Eleni Maniati
- Centre for Cancer Genomics and Computational Biology,
Barts Cancer Institute, Queen Mary University of London, London, UK
| | - David Ryan
- Wellcome Sanger Institute, Wellcome Genome Campus,
Hinxton, Cambridge CB10 1SA, UK
| | - Sandra Louzada
- Wellcome Sanger Institute, Wellcome Genome Campus,
Hinxton, Cambridge CB10 1SA, UK
| | - Jinhong Meng
- UCL Great Ormond Street Institute of Child Health, 30
Guilford Street, London WC1N 1EH, UK
| | - Anna Kowala
- Centre for Genomics and Child Health, Blizard
Institute, Barts and the London School of Medicine and Dentistry, Queen Mary
University of London, 4 Newark Street, London E1 2AT, UK
- Stem Cell Laboratory, National Bowel Research Centre,
Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen
Mary University of London, 2 Newark Street, London E1 2AT, UK
- Centre for Predictive in vitro Model, Queen Mary
University of London, Mile End Road, London E1 4NS, UK
| | - Beiyuan Fu
- Wellcome Sanger Institute, Wellcome Genome Campus,
Hinxton, Cambridge CB10 1SA, UK
| | - Fengtang Yang
- Wellcome Sanger Institute, Wellcome Genome Campus,
Hinxton, Cambridge CB10 1SA, UK
| | - Pentao Liu
- School of Biomedical Sciences, Stem Cell and
Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, The
University of Hong Kong, Hong Kong, China
| | - Silvia Marino
- Centre for Genomics and Child Health, Blizard
Institute, Barts and the London School of Medicine and Dentistry, Queen Mary
University of London, 4 Newark Street, London E1 2AT, UK
| | - Olivier Pourquié
- Department of Genetics and Department of Pathology,
Brigham and Women’s Hospital, Harvard Medical School, 60 Fenwood Road,
Boston, MA, USA
| | - Francesco Muntoni
- UCL Great Ormond Street Institute of Child Health, 30
Guilford Street, London WC1N 1EH, UK
- NIHR Biomedical Research Centre, Great Ormond
Street Hospital, Great Ormond Street, London, UK
| | - Jun Wang
- Centre for Cancer Genomics and Computational Biology,
Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Ivo Lieberam
- Centre for Stem Cells and Regenerative Medicine, MRC
Centre for Neurodevelopmental Disorders, and Centre for Developmental
Neurobiology, King’s College London, London, UK
| | - Yung-Yao Lin
- Centre for Genomics and Child Health, Blizard
Institute, Barts and the London School of Medicine and Dentistry, Queen Mary
University of London, 4 Newark Street, London E1 2AT, UK
- Stem Cell Laboratory, National Bowel Research Centre,
Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen
Mary University of London, 2 Newark Street, London E1 2AT, UK
- Centre for Predictive in vitro Model, Queen Mary
University of London, Mile End Road, London E1 4NS, UK
| |
Collapse
|
31
|
Townes-Anderson E, Halasz E, Wang W, Zarbin M. Coming of Age for the Photoreceptor Synapse. Invest Ophthalmol Vis Sci 2021; 62:24. [PMID: 34550300 PMCID: PMC8475281 DOI: 10.1167/iovs.62.12.24] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Purpose To discuss the potential contribution of rod and cone synapses to the loss of visual function in retinal injury and disease. Methods The published literature and the authors' own work were reviewed. Results Retinal detachment is used as a case study of rod spherule and cone pedicle plasticity after injury. Both rod and cone photoreceptors terminals are damaged after detachment although the structural changes observed are only partially overlapping. For second-order neurons, only those associated with rod spherules respond consistently to injury by remodeling. Examination of signaling pathways involved in plasticity of conventional synapses and in neural development has been and may continue to be productive in discovering novel therapeutic targets. Rho kinase (ROCK) inhibition is an example of therapy that may reduce synaptic damage by preserving normal synaptic structure of rod and cone cells. Conclusions We hypothesize that synaptic damage contributes to poor visual restoration after otherwise successful anatomical repair of retinal detachment. A similar situation may exist for patients with degenerative retinal disease. Thus, synaptic structure and function should be routinely studied, as this information may disclose therapeutic strategies to mitigate visual loss.
Collapse
Affiliation(s)
- Ellen Townes-Anderson
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey, United States
| | - Eva Halasz
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, New Jersey, United States
| | - Weiwei Wang
- Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard University, Boston, Massachusetts, United States
| | - Marco Zarbin
- Institute of Ophthalmology and Visual Science, Rutgers New Jersey Medical School, Newark, New Jersey, United States
| |
Collapse
|
32
|
Wang P, Yin R, Wang S, Zhou T, Zhang Y, Xiao M, Wang H, Xu G. Effects of Repetitive Transcranial Magnetic Stimulation (rTMS) and Treadmill Training on Recovery of Motor Function in a Rat Model of Partial Spinal Cord Injury. Med Sci Monit 2021; 27:e931601. [PMID: 34304239 PMCID: PMC8317583 DOI: 10.12659/msm.931601] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 03/24/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND This study aimed to investigate the effects of repetitive transcranial magnetic stimulation (rTMS) and treadmill training (TT) on motor function recovery in rats with partial spinal cord injury (SCI). MATERIAL AND METHODS Sixty rats with moderate partial SCI at the 9th thoracic vertebral level induced by a Louisville Injury System Apparatus impactor were randomly allocated to 5 groups: Sham surgery (Intact); Sham rTMS without TT (S-rTMS/Non-TT); Sham rTMS with TT (S-rTMS/TT); rTMS without TT (rTMS/Non-TT); and rTMS with TT (rTMS/TT). Interventions commenced 8 days after SCI and continued for 8 weeks. Outcomes studied were Basso, Beattie, and Bresnahan locomotor scale scores, grid walking test, and biochemical analysis of the brain-derived neurotrophic factor (BDNF), synapsin I (SYN), and postsynaptic density protein-95 (PSD-95) in the motor cortex and spinal cord. RESULTS The rTMS/TT contributed to greater Basso, Beattie, and Bresnahan scores compared with the S-rTMS/Non-TT (P<0.01), S-rTMS/TT (P<0.05), and rTMS/Non-TT (P<0.05), and showed obviously reduced numbers of foot drops compared with the S-rTMS/Non-TT (P<0.05). The rTMS/TT significantly increased the expressions of BDNF, SYN, and PSD-95 compared with the S-rTMS/Non-TT, both in the motor cortex (P<0.01, P<0.01, P<0.001, respectively) and spinal cord (P<0.001, P<0.01, P<0.05, respectively). CONCLUSIONS In a modified rat model of SCI, combined rTMS with TT improved motor function, indicating that this combined approach promoted adaptive neuroplasticity between the motor cortex and the spinal cord. A combined app roach to improving motor function following SCI requires further evaluation to determine the possible clinical applications.
Collapse
Affiliation(s)
- Pei Wang
- School of Rehabilitation Medicine, Nanjing Medical University, Center of Rehabilitation Medicine, 1 affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
- Department of Rehabilitation Medicine, Jiangsu Shengze Hospital, Nanjing Medical University, Suzhou, Jiangsu, PR China
| | - Ruian Yin
- School of Rehabilitation Medicine, Nanjing Medical University, Center of Rehabilitation Medicine, 1 affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Shuangyan Wang
- School of Rehabilitation Medicine, Nanjing Medical University, Center of Rehabilitation Medicine, 1 affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Ting Zhou
- Department of Rehabilitation Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Yongjie Zhang
- Department of Human Anatomy, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Ming Xiao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | - Hongxing Wang
- Department of Rehabilitation Medicine, Jiangsu Shengze Hospital, Nanjing Medical University, Suzhou, Jiangsu, PR China
- Department of Rehabilitation Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, PR China
| | - Guangxu Xu
- School of Rehabilitation Medicine, Nanjing Medical University, Center of Rehabilitation Medicine, 1 affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, PR China
| |
Collapse
|
33
|
Lee CY, Hyun SA, Ko MY, Kim HR, Rho J, Kim KK, Kim WY, Ka M. Maternal Bisphenol A (BPA) Exposure Alters Cerebral Cortical Morphogenesis and Synaptic Function in Mice. Cereb Cortex 2021; 31:5598-5612. [PMID: 34171088 DOI: 10.1093/cercor/bhab183] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/15/2022] Open
Abstract
Early-life exposure to bisphenol A (BPA), synthetic compound used in polycarbonate plastic, is associated with altered cognitive and emotional behavior later in life. However, the brain mechanism underlying the behavioral deficits is unknown. Here, we show that maternal BPA exposure disrupted self-renewal and differentiation of neural progenitors during cortical development. The BPA exposure reduced the neuron number, whereas it increased glial cells in the cerebral cortex. Also, synaptic formation and transmission in the cerebral cortex were suppressed after maternal BPA exposure. These changes appeared to be associated with autophagy as a gene ontology analysis of RNA-seq identified an autophagy domain in the BPA condition. Mouse behavioral tests revealed that maternal BPA caused hyperactivity and social deficits in adult offspring. Together, these results suggest that maternal BPA exposure leads to abnormal cortical architecture and function likely by activating autophagy.
Collapse
Affiliation(s)
- Chang Youn Lee
- Substance Abuse Pharmacology Group, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Sung-Ae Hyun
- Substance Abuse Pharmacology Group, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea.,Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Moon Yi Ko
- Substance Abuse Pharmacology Group, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea.,Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Hye Ryeong Kim
- Substance Abuse Pharmacology Group, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| | - Jaerang Rho
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Kee K Kim
- Department of Biochemistry, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Woo-Yang Kim
- Department of Biological Sciences, Kent State University, Kent, OH 44242, USA
| | - Minhan Ka
- Substance Abuse Pharmacology Group, Korea Institute of Toxicology, KRICT, Daejeon 34114, Republic of Korea
| |
Collapse
|
34
|
Teo S, Salinas PC. Wnt-Frizzled Signaling Regulates Activity-Mediated Synapse Formation. Front Mol Neurosci 2021; 14:683035. [PMID: 34194299 PMCID: PMC8236581 DOI: 10.3389/fnmol.2021.683035] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/17/2021] [Indexed: 11/26/2022] Open
Abstract
The formation of synapses is a tightly regulated process that requires the coordinated assembly of the presynaptic and postsynaptic sides. Defects in synaptogenesis during development or in the adult can lead to neurodevelopmental disorders, neurological disorders, and neurodegenerative diseases. In order to develop therapeutic approaches for these neurological conditions, we must first understand the molecular mechanisms that regulate synapse formation. The Wnt family of secreted glycoproteins are key regulators of synapse formation in different model systems from invertebrates to mammals. In this review, we will discuss the role of Wnt signaling in the formation of excitatory synapses in the mammalian brain by focusing on Wnt7a and Wnt5a, two Wnt ligands that play an in vivo role in this process. We will also discuss how changes in neuronal activity modulate the expression and/or release of Wnts, resulting in changes in the localization of surface levels of Frizzled, key Wnt receptors, at the synapse. Thus, changes in neuronal activity influence the magnitude of Wnt signaling, which in turn contributes to activity-mediated synapse formation.
Collapse
Affiliation(s)
| | - Patricia C. Salinas
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| |
Collapse
|
35
|
Aerobic Exercise Induces Alternative Splicing of Neurexins in Frontal Cortex. J Funct Morphol Kinesiol 2021; 6:jfmk6020048. [PMID: 34072692 PMCID: PMC8261640 DOI: 10.3390/jfmk6020048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 12/02/2022] Open
Abstract
Aerobic exercise (AE) is known to produce beneficial effects on brain health by improving plasticity, connectivity, and cognitive functions, but the underlying molecular mechanisms are still limited. Neurexins (Nrxns) are a family of presynaptic cell adhesion molecules that are important in synapsis formation and maturation. In vertebrates, three-neurexin genes (NRXN1, NRXN2, and NRXN3) have been identified, each encoding for α and β neurexins, from two independent promoters. Moreover, each Nrxns gene (1-3) has several alternative exons and produces many splice variants that bind to a large variety of postsynaptic ligands, playing a role in trans-synaptic specification, strength, and plasticity. In this study, we investigated the impact of a continuous progressive (CP) AE program on alternative splicing (AS) of Nrxns on two brain regions: frontal cortex (FC) and hippocampus. We showed that exercise promoted Nrxns1-3 AS at splice site 4 (SS4) both in α and β isoforms, inducing a switch from exon-excluded isoforms (SS4-) to exon-included isoforms (SS4+) in FC but not in hippocampus. Additionally, we showed that the same AE program enhanced the expression level of other genes correlated with synaptic function and plasticity only in FC. Altogether, our findings demonstrated the positive effect of CP AE on FC in inducing molecular changes underlying synaptic plasticity and suggested that FC is possibly a more sensitive structure than hippocampus to show molecular changes.
Collapse
|
36
|
León A, Aparicio GI, Scorticati C. Neuronal Glycoprotein M6a: An Emerging Molecule in Chemical Synapse Formation and Dysfunction. Front Synaptic Neurosci 2021; 13:661681. [PMID: 34017241 PMCID: PMC8129562 DOI: 10.3389/fnsyn.2021.661681] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/07/2021] [Indexed: 12/27/2022] Open
Abstract
The cellular and molecular mechanisms underlying neuropsychiatric and neurodevelopmental disorders show that most of them can be categorized as synaptopathies-or damage of synaptic function and plasticity. Synaptic formation and maintenance are orchestrated by protein complexes that are in turn regulated in space and time during neuronal development allowing synaptic plasticity. However, the exact mechanisms by which these processes are managed remain unknown. Large-scale genomic and proteomic projects led to the discovery of new molecules and their associated variants as disease risk factors. Neuronal glycoprotein M6a, encoded by the GPM6A gene is emerging as one of these molecules. M6a has been involved in neuron development and synapse formation and plasticity, and was also recently proposed as a gene-target in various neuropsychiatric disorders where it could also be used as a biomarker. In this review, we provide an overview of the structure and molecular mechanisms by which glycoprotein M6a participates in synapse formation and maintenance. We also review evidence collected from patients carrying mutations in the GPM6A gene; animal models, and in vitro studies that together emphasize the relevance of M6a, particularly in synapses and in neurological conditions.
Collapse
Affiliation(s)
| | | | - Camila Scorticati
- Instituto de Investigaciones Biotecnológicas “Rodolfo A. Ugalde”, Universidad Nacional de San Martín and Consejo Nacional de Investigaciones Científicas y Técnicas (IIBio-UNSAM-CONICET), Buenos Aires, Argentina
| |
Collapse
|
37
|
Perrone-Capano C, Volpicelli F, Penna E, Chun JT, Crispino M. Presynaptic protein synthesis and brain plasticity: From physiology to neuropathology. Prog Neurobiol 2021; 202:102051. [PMID: 33845165 DOI: 10.1016/j.pneurobio.2021.102051] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 03/14/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022]
Abstract
To form and maintain extremely intricate and functional neural circuitry, mammalian neurons are typically endowed with highly arborized dendrites and a long axon. The synapses that link neurons to neurons or to other cells are numerous and often too remote for the cell body to make and deliver new proteins to the right place in time. Moreover, synapses undergo continuous activity-dependent changes in their number and strength, establishing the basis of neural plasticity. The innate dilemma is then how a highly complex neuron provides new proteins for its cytoplasmic periphery and individual synapses to support synaptic plasticity. Here, we review a growing body of evidence that local protein synthesis in discrete sites of the axon and presynaptic terminals plays crucial roles in synaptic plasticity, and that deregulation of this local translation system is implicated in various pathologies of the nervous system.
Collapse
Affiliation(s)
- Carla Perrone-Capano
- Department of Pharmacy, University of Naples Federico II, Naples, Italy; Institute of Genetics and Biophysics "Adriano Buzzati Traverso", CNR, Naples, Italy.
| | | | - Eduardo Penna
- Department of Biology, University of Naples Federico II, Naples, Italy.
| | - Jong Tai Chun
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, Naples, Italy.
| | - Marianna Crispino
- Department of Biology, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
38
|
Abstract
Stroke is a debilitating disease. Current effective therapies for stroke recovery are limited to neurorehabilitation. Most stroke recovery occurs in a limited and early time window. Many of the mechanisms of spontaneous recovery after stroke parallel mechanisms of normal learning and memory. While various efforts are in place to identify potential drug targets, an emerging approach is to understand biological correlates between learning and stroke recovery. This review assesses parallels between biological changes at the molecular, structural, and functional levels during learning and recovery after stroke, with a focus on drug and cellular targets for therapeutics.
Collapse
Affiliation(s)
- Mary Teena Joy
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - S. Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
39
|
Hayashi S, Hoerder-Suabedissen A, Kiyokage E, Maclachlan C, Toida K, Knott G, Molnár Z. Maturation of Complex Synaptic Connections of Layer 5 Cortical Axons in the Posterior Thalamic Nucleus Requires SNAP25. Cereb Cortex 2021; 31:2625-2638. [PMID: 33367517 PMCID: PMC8023812 DOI: 10.1093/cercor/bhaa379] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 02/06/2023] Open
Abstract
Synapses are able to form in the absence of neuronal activity, but how is their subsequent maturation affected in the absence of regulated vesicular release? We explored this question using 3D electron microscopy and immunoelectron microscopy analyses in the large, complex synapses formed between cortical sensory efferent axons and dendrites in the posterior thalamic nucleus. Using a Synaptosome-associated protein 25 conditional knockout (Snap25 cKO), we found that during the first 2 postnatal weeks the axonal boutons emerge and increase in the size similar to the control animals. However, by P18, when an adult-like architecture should normally be established, axons were significantly smaller with 3D reconstructions, showing that each Snap25 cKO bouton only forms a single synapse with the connecting dendritic shaft. No excrescences from the dendrites were formed, and none of the normally large glomerular axon endings were seen. These results show that activity mediated through regulated vesicular release from the presynaptic terminal is not necessary for the formation of synapses, but it is required for the maturation of the specialized synaptic structures between layer 5 corticothalamic projections in the posterior thalamic nucleus.
Collapse
Affiliation(s)
- Shuichi Hayashi
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom
- Department of Anatomy, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan
| | - Anna Hoerder-Suabedissen
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom
| | - Emi Kiyokage
- Department of Medical Technology, Kawasaki University of Medical Welfare, Kurashiki, Okayama 701-0193, Japan
| | - Catherine Maclachlan
- BioEM Facility, School of Life Sciences, EPFL, Lausanne 1015, Switzerland
- The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Kazunori Toida
- Department of Anatomy, Kawasaki Medical School, Kurashiki, Okayama 701-0192, Japan
- Research Center for Ultra-High Voltage Electron Microscopy, Osaka University, Ibaraki, Osaka 567-0047, Japan
| | - Graham Knott
- BioEM Facility, School of Life Sciences, EPFL, Lausanne 1015, Switzerland
| | - Zoltán Molnár
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3PT, United Kingdom
| |
Collapse
|
40
|
Boolean Networks: A Primer. SYSTEMS MEDICINE 2021. [DOI: 10.1016/b978-0-12-801238-3.11518-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
41
|
Kuo DH, De-Miguel FF, Heath-Heckman EAC, Szczupak L, Todd K, Weisblat DA, Winchell CJ. A tale of two leeches: Toward the understanding of the evolution and development of behavioral neural circuits. Evol Dev 2020; 22:471-493. [PMID: 33226195 DOI: 10.1111/ede.12358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 10/23/2020] [Accepted: 11/02/2020] [Indexed: 11/29/2022]
Abstract
In the animal kingdom, behavioral traits encompass a broad spectrum of biological phenotypes that have critical roles in adaptive evolution, but an EvoDevo approach has not been broadly used to study behavior evolution. Here, we propose that, by integrating two leech model systems, each of which has already attained some success in its respective field, it is possible to take on behavioral traits with an EvoDevo approach. We first identify the developmental changes that may theoretically lead to behavioral evolution and explain why an EvoDevo study of behavior is challenging. Next, we discuss the pros and cons of the two leech model species, Hirudo, a classic model for invertebrate neurobiology, and Helobdella, an emerging model for clitellate developmental biology, as models for behavioral EvoDevo research. Given the limitations of each leech system, neither is particularly strong for behavioral EvoDevo. However, the two leech systems are complementary in their technical accessibilities, and they do exhibit some behavioral similarities and differences. By studying them in parallel and together with additional leech species such as Haementeria, it is possible to explore the different levels of behavioral development and evolution.
Collapse
Affiliation(s)
- Dian-Han Kuo
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Francisco F De-Miguel
- Instituto de Fisiología Celular - Neurociencias, Universidad Nacional Autónoma de México, México City, México
| | | | - Lidia Szczupak
- Departamento de Fisiología Biología Molecular y Celular, Universidad de Buenos Aires, and IFIBYNE UBA-CONICET, Buenos Aires, Argentina
| | - Krista Todd
- Department of Neuroscience, Westminster College, Salt Lake City, Utah, USA
| | - David A Weisblat
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | - Christopher J Winchell
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| |
Collapse
|
42
|
Torres-Berrío A, Hernandez G, Nestler EJ, Flores C. The Netrin-1/DCC Guidance Cue Pathway as a Molecular Target in Depression: Translational Evidence. Biol Psychiatry 2020; 88:611-624. [PMID: 32593422 PMCID: PMC7529861 DOI: 10.1016/j.biopsych.2020.04.025] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/14/2020] [Accepted: 04/28/2020] [Indexed: 12/15/2022]
Abstract
The Netrin-1/DCC guidance cue pathway plays a critical role in guiding growing axons toward the prefrontal cortex during adolescence and in the maturational organization and adult plasticity of prefrontal cortex connectivity. In this review, we put forward the idea that alterations in prefrontal cortex architecture and function, which are intrinsically linked to the development of major depressive disorder, originate in part from the dysregulation of the Netrin-1/DCC pathway by a mechanism that involves microRNA-218. We discuss evidence derived from mouse models of stress and from human postmortem brain and genome-wide association studies indicating an association between the Netrin-1/DCC pathway and major depressive disorder. We propose a potential role of circulating microRNA-218 as a biomarker of stress vulnerability and major depressive disorder.
Collapse
Affiliation(s)
- Angélica Torres-Berrío
- Integrated Program in Neuroscience, Montreal, Quebec, Canada; Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Cecilia Flores
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Douglas Mental Health University Institute, Montreal, Quebec, Canada.
| |
Collapse
|
43
|
Wright EAP, Goltsev AV. Statistical analysis of unidirectional and reciprocal chemical connections in the C. elegans connectome. Eur J Neurosci 2020; 52:4525-4535. [PMID: 33022789 DOI: 10.1111/ejn.14988] [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: 07/06/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 11/29/2022]
Abstract
We analyze unidirectional and reciprocally connected pairs of neurons in the chemical connectomes of the male and hermaphrodite Caenorhabditis elegans, using recently published data. Our analysis reveals that reciprocal connections provide communication between most neurons with chemical synapses, and comprise on average more synapses than both unidirectional connections and the entire connectome. We further reveal that the C. elegans connectome is wired so that afferent connections onto neurons with large numbers of presynaptic neighbors (in-degree) comprise an above-average number of synapses (synaptic multiplicity). Notably, the larger the in-degree of a neuron the larger the synaptic multiplicity of its afferent connections. Finally, we show that the male forms two times fewer reciprocal connections between sex-shared neurons than the hermaphrodite, but a large number of reciprocal connections with male-specific neurons. These observations provide evidence for Hebbian structural plasticity in the C. elegans.
Collapse
Affiliation(s)
- Edgar A P Wright
- Department of Physics & I3N, University of Aveiro, Aveiro, Portugal
| | - Alexander V Goltsev
- Department of Physics & I3N, University of Aveiro, Aveiro, Portugal.,A.F. Ioffe Physico-Technical Institute, St. Petersburg, Russia
| |
Collapse
|
44
|
Synaptic Protein Degradation Controls Sexually Dimorphic Circuits through Regulation of DCC/UNC-40. Curr Biol 2020; 30:4128-4141.e5. [PMID: 32857970 PMCID: PMC7658809 DOI: 10.1016/j.cub.2020.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/13/2020] [Accepted: 08/03/2020] [Indexed: 12/27/2022]
Abstract
Sexually dimorphic circuits underlie behavioral differences between the sexes, yet the molecular mechanisms involved in their formation are poorly understood. We show here that sexually dimorphic connectivity patterns arise in C. elegans through local ubiquitin-mediated protein degradation in selected synapses of one sex but not the other. Specifically, synaptic degradation occurs via binding of the evolutionary conserved E3 ligase SEL-10/FBW7 to a phosphodegron binding site of the netrin receptor UNC-40/DCC (Deleted in Colorectal Cancer), resulting in degradation of UNC-40. In animals carrying an undegradable unc-40 gain-of-function allele, synapses were retained in both sexes, compromising the activity of the circuit without affecting neurite guidance. Thus, by decoupling the synaptic and guidance functions of the netrin pathway, we reveal a critical role for dimorphic protein degradation in controlling neuronal connectivity and activity. Additionally, the interaction between SEL-10 and UNC-40 is necessary not only for sex-specific synapse pruning, but also for other synaptic functions. These findings provide insight into the mechanisms that generate sex-specific differences in neuronal connectivity, activity, and function. Sex-specific synapse pruning during development is regulated by the ubiquitin pathway The E3 ligase SEL-10 targets the UNC-40 netrin receptor via binding to a CPD motif UNC-40 degradation leads to synapse removal only in hermaphrodites, not males CPD mutations disrupt synaptic functions of UNC-40, leaving axon guidance intact
Collapse
|
45
|
Maurin T, Melancia F, Jarjat M, Castro L, Costa L, Delhaye S, Khayachi A, Castagnola S, Mota E, Di Giorgio A, Servadio M, Drozd M, Poupon G, Schiavi S, Sardone L, Azoulay S, Ciranna L, Martin S, Vincent P, Trezza V, Bardoni B. Involvement of Phosphodiesterase 2A Activity in the Pathophysiology of Fragile X Syndrome. Cereb Cortex 2020; 29:3241-3252. [PMID: 30137253 DOI: 10.1093/cercor/bhy192] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 07/12/2018] [Accepted: 07/20/2018] [Indexed: 12/26/2022] Open
Abstract
The fragile X mental retardation protein (FMRP) is an RNA-binding protein involved in translational regulation of mRNAs that play key roles in synaptic morphology and plasticity. The functional absence of FMRP causes the fragile X syndrome (FXS), the most common form of inherited intellectual disability and the most common monogenic cause of autism. No effective treatment is available for FXS. We recently identified the Phosphodiesterase 2A (Pde2a) mRNA as a prominent target of FMRP. PDE2A enzymatic activity is increased in the brain of Fmr1-KO mice, a recognized model of FXS, leading to decreased levels of cAMP and cGMP. Here, we pharmacologically inhibited PDE2A in Fmr1-KO mice and observed a rescue both of the maturity of dendritic spines and of the exaggerated hippocampal mGluR-dependent long-term depression. Remarkably, PDE2A blockade rescued the social and communicative deficits of both mouse and rat Fmr1-KO animals. Importantly, chronic inhibition of PDE2A in newborn Fmr1-KO mice followed by a washout interval, resulted in the rescue of the altered social behavior observed in adolescent mice. Altogether, these results reveal the key role of PDE2A in the physiopathology of FXS and suggest that its pharmacological inhibition represents a novel therapeutic approach for FXS.
Collapse
Affiliation(s)
- Thomas Maurin
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France.,CNRS LIA «Neogenex», Valbonne, France
| | | | - Marielle Jarjat
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France.,CNRS LIA «Neogenex», Valbonne, France
| | - Liliana Castro
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France.,Labex BioPsy, Paris, France
| | - Lara Costa
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Sébastien Delhaye
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France.,CNRS LIA «Neogenex», Valbonne, France
| | | | - Sara Castagnola
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France.,CNRS LIA «Neogenex», Valbonne, France
| | - Elia Mota
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France.,Labex BioPsy, Paris, France
| | - Audrey Di Giorgio
- Université Côte d'Azur, CNRS, Institut de Chimie de Nice, Nice, France
| | | | - Malgorzata Drozd
- Université Côte d'Azur, CNRS, IPMC, Valbonne, France.,CNRS LIA «Neogenex», Valbonne, France
| | | | - Sara Schiavi
- Department of Sciences, Università RomaTre, Roma, Italy
| | - Lara Sardone
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Stéphane Azoulay
- Université Côte d'Azur, CNRS, Institut de Chimie de Nice, Nice, France
| | - Lucia Ciranna
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Stéphane Martin
- Université Côte d'Azur, INSERM, CNRS, IPMC, Valbonne, France
| | - Pierre Vincent
- Sorbonne Université, CNRS, Biological Adaptation and Ageing, Paris, France.,Labex BioPsy, Paris, France
| | | | - Barbara Bardoni
- CNRS LIA «Neogenex», Valbonne, France.,Université Côte d'Azur, INSERM, CNRS, IPMC, Valbonne, France
| |
Collapse
|
46
|
Gamma oryzanol impairs alcohol-induced anxiety-like behavior in mice via upregulation of central monoamines associated with Bdnf and Il-1β signaling. Sci Rep 2020; 10:10677. [PMID: 32606350 PMCID: PMC7326911 DOI: 10.1038/s41598-020-67689-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
Adolescent alcohol exposure may increase anxiety-like behaviors by altering central monoaminergic functions and other important neuronal pathways. The present study was designed to investigate the anxiolytic effect of 0.5% γ-oryzanol (GORZ) and its neurochemical and molecular mechanisms under chronic 10% ethanol consumption. Five-week-old ICR male mice received either control (14% casein, AIN 93 M) or GORZ (14% casein, AIN 93 M + 0.5% GORZ) diets in this study. We showed that GORZ could potentially attenuate alcohol-induced anxiety-like behaviors by significantly improving the main behavioral parameters measured by the elevated plus maze test. Moreover, GORZ treatment significantly restored the alcohol-induced downregulation of 5-hydroxytryptophan and 5-hydroxyindole acetic acid in the hippocampus and improved homovanillic acid levels in the cerebral cortex. Furthermore, a recovery increase in the level of 3-methoxy-4-hydroxyphenylglycol both in the hippocampus and cerebral cortex supported the anxiolytic effect of GORZ. The significant elevation and reduction in the hippocampus of relative mRNA levels of brain-derived neurotrophic factor and interleukin 1β, respectively, also showed the neuroprotective role of GORZ in ethanol-induced anxiety. Altogether, these results suggest that 0.5% GORZ is a promising neuroprotective drug candidate with potential anxiolytic, neurogenic, and anti-neuroinflammatory properties for treating adolescent alcohol exposure.
Collapse
|
47
|
Cuesta S, Nouel D, Reynolds LM, Morgunova A, Torres-Berrío A, White A, Hernandez G, Cooper HM, Flores C. Dopamine Axon Targeting in the Nucleus Accumbens in Adolescence Requires Netrin-1. Front Cell Dev Biol 2020; 8:487. [PMID: 32714924 PMCID: PMC7344302 DOI: 10.3389/fcell.2020.00487] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/25/2020] [Indexed: 12/17/2022] Open
Abstract
The fine arrangement of neuronal connectivity during development involves the coordinated action of guidance cues and their receptors. In adolescence, the dopamine circuitry is still developing, with mesolimbic dopamine axons undergoing target-recognition events in the nucleus accumbens (NAcc), while mesocortical projections continue to grow toward the prefrontal cortex (PFC) until adulthood. This segregation of mesolimbic versus mesocortical dopamine pathways is mediated by the guidance cue receptor DCC, which signals dopamine axons intended to innervate the NAcc to recognize this region as their final target. Whether DCC-dependent mesolimbic dopamine axon targeting in adolescence requires the action of its ligand, Netrin-1, is unknown. Here we combined shRNA strategies, quantitative analysis of pre- and post-synaptic markers of neuronal connectivity, and pharmacological manipulations to address this question. Similar to DCC levels in the ventral tegmental area, Netrin-1 expression in the NAcc is dynamic across postnatal life, transitioning from high to low expression across adolescence. Silencing Netrin-1 in the NAcc in adolescence results in an increase in the expanse of the dopamine input to the PFC in adulthood, with a corresponding increase in the number of presynaptic dopamine sites. This manipulation also results in altered dendritic spine density and morphology of medium spiny neurons in the NAcc in adulthood and in reduced sensitivity to the behavioral activating effects of the stimulant drug of abuse, amphetamine. These cellular and behavioral effects mirror those induced by Dcc haploinsufficiency within dopamine neurons in adolescence. Dopamine targeting in adolescence requires the complementary interaction between DCC receptors in mesolimbic dopamine axons and Netrin-1 in the NAcc. Factors regulating either DCC or Netrin-1 in adolescence can disrupt mesocorticolimbic dopamine development, rendering vulnerability or protection to phenotypes associated with psychiatric disorders.
Collapse
Affiliation(s)
- Santiago Cuesta
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Dominique Nouel
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Lauren M Reynolds
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Alice Morgunova
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Angélica Torres-Berrío
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Amanda White
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Giovanni Hernandez
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| | - Helen M Cooper
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Cecilia Flores
- Department of Psychiatry, Douglas Mental Health University Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
48
|
Biallelic PDE2A variants: a new cause of syndromic paroxysmal dyskinesia. Eur J Hum Genet 2020; 28:1403-1413. [PMID: 32467598 PMCID: PMC7608189 DOI: 10.1038/s41431-020-0641-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 04/01/2020] [Accepted: 04/28/2020] [Indexed: 11/08/2022] Open
Abstract
Cause of complex dyskinesia remains elusive in some patients. A homozygous missense variant leading to drastic decrease of PDE2A enzymatic activity was reported in one patient with childhood-onset choreodystonia preceded by paroxysmal dyskinesia and associated with cognitive impairment and interictal EEG abnormalities. Here, we report three new cases with biallelic PDE2A variants identified by trio whole-exome sequencing. Mitochondria network was analyzed after Mitotracker™ Red staining in control and mutated primary fibroblasts. Analysis of retrospective video of patients' movement disorder and refinement of phenotype was carried out. We identified a homozygous gain of stop codon variant c.1180C>T; p.(Gln394*) in PDE2A in siblings and compound heterozygous variants in young adult: a missense c.446C>T; p.(Pro149Leu) and splice-site variant c.1922+5G>A predicted and shown to produce an out of frame transcript lacking exon 22. All three patients had cognitive impairment or developmental delay. The phenotype of the two oldest patients, aged 9 and 26, was characterized by childhood-onset refractory paroxysmal dyskinesia initially misdiagnosed as epilepsy due to interictal EEG abnormalities. The youngest patient showed a proven epilepsy at the age of 4 months and no paroxysmal dyskinesia at 15 months. Interestingly, analysis of the fibroblasts with the biallelic variants in PDE2A variants revealed mitochondria network morphology changes. Together with previously reported case, our three patients confirm that biallelic PDE2A variants are a cause of childhood-onset refractory paroxysmal dyskinesia with cognitive impairment, sometimes associated with choreodystonia and interictal baseline EEG abnormalities or epilepsy.
Collapse
|
49
|
Sasaki T, Komatsu Y, Yamamori T. Expression patterns of SLIT/ROBO mRNAs reveal a characteristic feature in the entorhinal-hippocampal area of macaque monkeys. BMC Res Notes 2020; 13:262. [PMID: 32460877 PMCID: PMC7251749 DOI: 10.1186/s13104-020-05100-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 05/19/2020] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE SLITs are secreted glycoproteins that bind to Roundabouts (ROBOs) which are a family member of transmembrane receptors. SLIT signaling has well-conserved roles in mediating axon repulsion in a developing nervous system. We previously reported that SLIT1 mRNA is enriched in middle layers of the prefrontal cortex of macaque monkeys in a developmentally regulated manner. Other SLIT (SLIT2 and SLIT3) mRNAs showed preferential expressions in the prefrontal cortex with a distinct laminar pattern. To obtain further clues to the role of SLIT signaling in the organization of the primate brain, we performed ISH analysis of SLIT and ROBO mRNAs using adult macaque brain tissues. RESULTS In this study, we examined the expression patterns of SLITs and ROBOs (ROBO1 and ROBO2) in other brain regions, and found intense and characteristic expression patterns of these genes in the entorhinal-hippocampal area. In situ hybridization analysis revealed that SLIT1 and SLIT2 mRNAs showed marked complementary distribution in the entorhinal cortex. SLIT and ROBO mRNAs were widely expressed in the hippocampus with modest regional preference. These findings suggest that each SLIT gene has a specialized role that is particularly important for prefrontal as well as hippocampal connectivity in the primate cortex.
Collapse
Affiliation(s)
- Tetsuya Sasaki
- Division of Brain Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, 444-8585, Japan.
- Department of Anatomy and Neuroscience, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
- Ph.D Program of Neurosciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8577, Japan.
| | - Yusuke Komatsu
- Division of Brain Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, 444-8585, Japan
- Laboratory of Veterinary Hygiene, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, 060-0816, Japan
| | - Tetsuo Yamamori
- Division of Brain Biology, National Institute for Basic Biology, 38 Nishigonaka Myodaiji, Okazaki, 444-8585, Japan.
- Laboratory for Molecular Analysis of Higher Brain Function, RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan.
| |
Collapse
|
50
|
Mohan V, Sullivan CS, Guo J, Wade SD, Majumder S, Agarwal A, Anton ES, Temple BS, Maness PF. Temporal Regulation of Dendritic Spines Through NrCAM-Semaphorin3F Receptor Signaling in Developing Cortical Pyramidal Neurons. Cereb Cortex 2020; 29:963-977. [PMID: 29415226 DOI: 10.1093/cercor/bhy004] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 01/06/2018] [Indexed: 01/03/2023] Open
Abstract
Neuron-glial related cell adhesion molecule NrCAM is a newly identified negative regulator of spine density that genetically interacts with Semaphorin3F (Sema3F), and is implicated in autism spectrum disorders (ASD). To investigate a role for NrCAM in spine pruning during the critical adolescent period when networks are established, we generated novel conditional, inducible NrCAM mutant mice (Nex1Cre-ERT2: NrCAMflox/flox). We demonstrate that NrCAM functions cell autonomously during adolescence in pyramidal neurons to restrict spine density in the visual (V1) and medial frontal cortex (MFC). Guided by molecular modeling, we found that NrCAM promoted clustering of the Sema3F holoreceptor complex by interfacing with Neuropilin-2 (Npn2) and PDZ scaffold protein SAP102. NrCAM-induced receptor clustering stimulated the Rap-GAP activity of PlexinA3 (PlexA3) within the holoreceptor complex, which in turn, inhibited Rap1-GTPase and inactivated adhesive β1 integrins, essential for Sema3F-induced spine pruning. These results define a developmental function for NrCAM in Sema3F receptor signaling that limits dendritic spine density on cortical pyramidal neurons during adolescence.
Collapse
Affiliation(s)
- Vishwa Mohan
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Chelsea S Sullivan
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Jiami Guo
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Sarah D Wade
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Samarpan Majumder
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Amit Agarwal
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Goettingen, Germany
| | - Eva S Anton
- UNC Neuroscience Center and the Department of Cell Biology and Physiology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Brenda S Temple
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Patricia F Maness
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|