1
|
Gregor A, Distel L, Ekici AB, Kirchner P, Uebe S, Krumbiegel M, Turan S, Winner B, Zweier C. Proteasomal activation ameliorates neuronal phenotypes linked to FBXO11-deficiency. HGG ADVANCES 2025; 6:100425. [PMID: 40114442 PMCID: PMC11999343 DOI: 10.1016/j.xhgg.2025.100425] [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: 11/15/2024] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Haploinsufficiency of FBXO11, encoding a ubiquitin ligase complex subunit, is associated with a variable neurodevelopmental disorder. So far, the underlying nervous system-related pathomechanisms are poorly understood, and specific therapies are lacking. Using a combined approach, we established an FBXO11-deficient human stem cell-based neuronal model using CRISPR-Cas9 and a Drosophila model using tissue-specific knockdown techniques. We performed transcriptomic analyses on iPSC-derived neurons and molecular phenotyping in both models. RNA sequencing revealed disrupted transcriptional networks related to processes important for neuronal development, such as differentiation, migration, and cell signaling. Consistently, we found that loss of FBXO11 leads to neuronal phenotypes such as impaired neuronal migration and abnormal proliferation/differentiation balance in human cultured neurons and impaired dendritic development and behavior in Drosophila. Interestingly, application of three different proteasome-activating substances could alleviate FBXO11-deficiency-associated phenotypes in both human neurons and flies. One of these substances is the long-approved drug Verapamil, opening the possibility of drug repurposing in the future. Our study shows the importance of FBXO11 for neurodevelopment and highlights the reversibility of related phenotypes, opening an avenue for potential development of therapeutic approaches through drug repurposing.
Collapse
Affiliation(s)
- Anne Gregor
- Department of Human Genetics, Inselspital University Hospital Bern, University of Bern, 3010 Bern, Switzerland; Department for Biomedical Research (DBMR), University of Bern, 3010 Bern, Switzerland.
| | - Laila Distel
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Arif B Ekici
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Philipp Kirchner
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; Institute of Tissue Medicine and Pathology, University of Bern, 3010 Bern, Switzerland
| | - Steffen Uebe
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Mandy Krumbiegel
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Soeren Turan
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Beate Winner
- Department of Stem Cell Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany; Centre for Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christiane Zweier
- Department of Human Genetics, Inselspital University Hospital Bern, University of Bern, 3010 Bern, Switzerland; Department for Biomedical Research (DBMR), University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
2
|
Cárdenas-Bedoya J, Torres-Mendoza BM, Martínez-Torres NI. Cerebrolysin Induces Dendritic Tree Plastic Changes and BDNF Increase in the Amygdala of Male Rats with Maternal Deprivation. Neurochem Res 2025; 50:123. [PMID: 40123021 DOI: 10.1007/s11064-025-04376-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Several psychopathologies may be triggered, among other factors, by stress or trauma in childhood. The maternal deprivation model (MD) replicates some of the core factors that may induce the onset of different psychopathologies like structural and plasticity defects. Among other brain regions, the amygdala is susceptible to stress and trauma and plays a key role in integrating social behavior. Several molecules, such as Cerebrolysin® (CBL), have been used to treat different symptoms by reversing the underlying neurobiological plasticity-related changes. In this study, maternal deprivation (MD) was conducted on 9-day-old male Sprague-Dawley rats (n = 28; 7 rats per group: Intact, Intact + CBL, MD, and MD + CBL). At 25 postnatal days (PND), CBL treatment was administered for 10 consecutive days, and social behavior was evaluated in a three-chamber social test at 35 PND. Moreover, Sholl analysis and immunohistochemistry were carried out for dendritic intersection and brain-derived neurotrophic factor (BDNF) presence in the amygdala, respectively. CBL treatment had an augmentative effect on intact animals in terms of social behavior and incidences, but no differences between MD and CBL-treated animals. Moreover, dendritic intersections and BDNF decreased after the MD protocol but increased by CBL treatment in MD animals. Our results show that CBL could be part of the treatment in case of a traumatic or stressful event in neurodevelopment, especially in the youth age. According to this preclinical study, CBL could help reverse symptoms of different psychopathologies caused by stress or trauma, with neurobiological changes underlying its effect.
Collapse
Affiliation(s)
- Jhonathan Cárdenas-Bedoya
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Col. Independencia, Guadalajara, C.P. 44340, Jalisco, México
- Centro Universitario de Ciencias de la Salud, Departamento de Disciplinas Filósofico, Metodológicas e Instrumentales, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Blanca Miriam Torres-Mendoza
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Col. Independencia, Guadalajara, C.P. 44340, Jalisco, México
- Centro Universitario de Ciencias de la Salud, Departamento de Disciplinas Filósofico, Metodológicas e Instrumentales, Universidad de Guadalajara, Guadalajara, Jalisco, México
| | - Nestor Ismael Martínez-Torres
- División de Neurociencias, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Sierra Mojada #800, Col. Independencia, Guadalajara, C.P. 44340, Jalisco, México.
- Departamento de Bienestar y Desarrollo Sustentable, Centro Universitario del Norte, Universidad de Guadalajara, Colotlán, Jalisco, México.
| |
Collapse
|
3
|
Ishchenko Y, Jeng AT, Feng S, Nottoli T, Manriquez-Rodriguez C, Nguyen KK, Carrizales MG, Vitarelli MJ, Corcoran EE, Greer CA, Myers SA, Koleske AJ. Heterozygosity for neurodevelopmental disorder-associated TRIO variants yields distinct deficits in behavior, neuronal development, and synaptic transmission in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.01.05.574442. [PMID: 39131289 PMCID: PMC11312463 DOI: 10.1101/2024.01.05.574442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Genetic variants in TRIO are associated with neurodevelopmental disorders (NDDs) including schizophrenia (SCZ), autism spectrum disorder (ASD) and intellectual disability. TRIO uses its two guanine nucleotide exchange factor (GEF) domains to activate GTPases (GEF1: Rac1 and RhoG; GEF2: RhoA) that control neuronal development and connectivity. It remains unclear how discrete TRIO variants differentially impact these neurodevelopmental events. Here, we investigate how heterozygosity for NDD-associated Trio variants - +/K1431M (ASD), +/K1918X (SCZ), and +/M2145T (bipolar disorder, BPD) - impact mouse behavior, brain development, and synapse structure and function. Heterozygosity for different Trio variants impacts motor, social, and cognitive behaviors in distinct ways that model clinical phenotypes in humans. Trio variants differentially impact head and brain size, with corresponding changes in dendritic arbors of motor cortex layer 5 pyramidal neurons (M1 L5 PNs). Although neuronal structure was only modestly altered in the Trio variant heterozygotes, we observe significant changes in synaptic function and plasticity. We also identified distinct changes in glutamate synaptic release in +/K1431M and +/M2145T cortico-cortical synapses. The TRIO K1431M GEF1 domain has impaired ability to promote GTP exchange on Rac1, but +/K1431M mice exhibit increased Rac1 activity, associated with increased levels of the Rac1 GEF Tiam1. Acute Rac1 inhibition with NSC23766 rescued glutamate release deficits in +/K1431M variant cortex. Our work reveals that discrete NDD-associated Trio variants yield overlapping but distinct phenotypes in mice, demonstrates an essential role for Trio in presynaptic glutamate release, and underscores the importance of studying the impact of variant heterozygosity in vivo.
Collapse
Affiliation(s)
- Yevheniia Ishchenko
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Amanda T Jeng
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
| | - Shufang Feng
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Gerontology, The Third Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Timothy Nottoli
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
| | | | - Khanh K Nguyen
- Laboratory for Immunochemical Circuits, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Melissa G Carrizales
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Matthew J Vitarelli
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Ellen E Corcoran
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
| | - Charles A Greer
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Samuel A Myers
- Laboratory for Immunochemical Circuits, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale School of Medicine, New Haven, CT, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, CT, USA
| |
Collapse
|
4
|
Aksan B, Mauceri D. Beyond vessels: unraveling the impact of VEGFs on neuronal functions and structure. J Biomed Sci 2025; 32:33. [PMID: 40050849 PMCID: PMC11884128 DOI: 10.1186/s12929-025-01128-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 02/21/2025] [Indexed: 03/10/2025] Open
Abstract
Neurons rely on the bloodstream for essential nutrients and oxygen, which is facilitated by an intricate coupling of the neuronal and vascular systems. Central to this neurovascular interaction is the vascular endothelial growth factor (VEGF) family, a group of secreted growth factors traditionally known for their roles in promoting endothelial cell proliferation, migration, and survival in the cardiovascular and lymphatic systems. However, emerging evidence shows that VEGFs also play indispensable roles in the nervous system, extending beyond their canonical angiogenic and lymphangiogenic functions. Over the past two decades, VEGFs have been found to exert direct effects on neurons, influencing key aspects of neuronal function independently of their actions on vascular cells. In particular, it has become increasingly evident that VEGFs also play crucial functions in the development, regulation, and maintenance of neuronal morphology. Understanding the roles of VEGFs in neuronal development is of high scientific and clinical interest because of the significance of precise neuronal morphology for neural connectivity and network function, as well as the association of morphological abnormalities with neurological and neurodegenerative disorders. This review begins with an overview of the VEGF family members, their structural characteristics, receptors, and established roles in vasculature. However, it then highlights and focuses on the exciting variety of neuronal functions of VEGFs, especially their crucial role in the development, regulation, and maintenance of neuronal morphology.
Collapse
Affiliation(s)
- Bahar Aksan
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Daniela Mauceri
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany.
- Institute of Anatomy and Cell Biology, Dept. Molecular and Cellular Neuroscience, University of Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany.
| |
Collapse
|
5
|
Saccenti D, Lauro LJR, Crespi SA, Moro AS, Vergallito A, Grgič RG, Pretti N, Lamanna J, Ferro M. Boosting Psychotherapy With Noninvasive Brain Stimulation: The Whys and Wherefores of Modulating Neural Plasticity to Promote Therapeutic Change. Neural Plast 2024; 2024:7853199. [PMID: 39723244 PMCID: PMC11669434 DOI: 10.1155/np/7853199] [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: 05/03/2024] [Accepted: 12/04/2024] [Indexed: 12/28/2024] Open
Abstract
The phenomenon of neural plasticity pertains to the intrinsic capacity of neurons to undergo structural and functional reconfiguration through learning and experiential interaction with the environment. These changes could manifest themselves not only as a consequence of various life experiences but also following therapeutic interventions, including the application of noninvasive brain stimulation (NIBS) and psychotherapy. As standalone therapies, both NIBS and psychotherapy have demonstrated their efficacy in the amelioration of psychiatric disorders' symptoms, with a certain variability in terms of effect sizes and duration. Consequently, scholars suggested the convenience of integrating the two interventions into a multimodal treatment to boost and prolong the therapeutic outcomes. Such an approach is still in its infancy, and the physiological underpinnings substantiating the effectiveness and utility of combined interventions are still to be clarified. Therefore, this opinion paper aims to provide a theoretical framework consisting of compelling arguments as to why adding NIBS to psychotherapy can promote therapeutic change. Namely, we will discuss the physiological effects of the two interventions, thus providing a rationale to explain the potential advantages of a combined approach.
Collapse
Affiliation(s)
- Daniele Saccenti
- Department of Psychology, Sigmund Freud University, Milan, Italy
| | - Leonor J. Romero Lauro
- Department of Psychology and NeuroMi, University of Milano-Bicocca, Milan, Italy
- Cognitive Studies, Cognitive Psychotherapy School and Research Center, Milan, Italy
| | - Sofia A. Crespi
- Cognitive Studies, Cognitive Psychotherapy School and Research Center, Milan, Italy
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
| | - Andrea S. Moro
- Department of Psychology, Sigmund Freud University, Milan, Italy
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy
| | | | | | - Novella Pretti
- Cognitive Studies, Cognitive Psychotherapy School and Research Center, Milan, Italy
- Clinical Psychology Center, Division of Neurology, Galliera Hospital, Genoa, Italy
| | - Jacopo Lamanna
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy
| | - Mattia Ferro
- Department of Psychology, Sigmund Freud University, Milan, Italy
- Center for Behavioral Neuroscience and Communication (BNC), Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
6
|
Tann JY, Xu F, Kimura M, Wilkes OR, Yoong LF, Skibbe H, Moore AW. Study of Dendrite Differentiation Using Drosophila Dendritic Arborization Neurons. Cold Spring Harb Protoc 2024; 2024:pdb.top108146. [PMID: 38148165 DOI: 10.1101/pdb.top108146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Neurons receive, process, and integrate inputs. These operations are organized by dendrite arbor morphology, and the dendritic arborization (da) neurons of the Drosophila peripheral sensory nervous system are an excellent experimental model for examining the differentiation processes that build and shape the dendrite arbor. Studies in da neurons are enabled by a wealth of fly genetic tools that allow targeted neuron manipulation and labeling of the neuron's cytoskeletal or organellar components. Moreover, as da neuron dendrite arbors cover the body wall, they are highly accessible for live imaging analysis of arbor patterning. Here, we outline the structure and function of different da neuron types and give examples of how they are used to elucidate central mechanisms of dendritic arbor formation.
Collapse
Affiliation(s)
- Jason Y Tann
- Laboratory for Neurodiversity, RIKEN Center for Brain Science, Wako-shi, 351-0106, Japan
| | - Fangke Xu
- Laboratory for Neurodiversity, RIKEN Center for Brain Science, Wako-shi, 351-0106, Japan
| | - Minami Kimura
- Laboratory for Neurodiversity, RIKEN Center for Brain Science, Wako-shi, 351-0106, Japan
| | - Oliver R Wilkes
- Laboratory for Neurodiversity, RIKEN Center for Brain Science, Wako-shi, 351-0106, Japan
- Department of Cellular and Molecular Biology, Institute for Translational Medicine, University of Liverpool, Liverpool L69 3BX, United Kingdom
| | - Li-Foong Yoong
- Laboratory for Neurodiversity, RIKEN Center for Brain Science, Wako-shi, 351-0106, Japan
| | - Henrik Skibbe
- Brain Image Analysis Unit, RIKEN Center for Brain Science, Wako-shi, 351-0106, Japan
| | - Adrian W Moore
- Laboratory for Neurodiversity, RIKEN Center for Brain Science, Wako-shi, 351-0106, Japan
| |
Collapse
|
7
|
Jaholkowski P, Bahrami S, Fominykh V, Hindley GFL, Tesfaye M, Parekh P, Parker N, Filiz TT, Nordengen K, Hagen E, Koch E, Bakken NR, Frei E, Birkenæs V, Rahman Z, Frei O, Haavik J, Djurovic S, Dale AM, Smeland OB, O'Connell KS, Shadrin AA, Andreassen OA. Charting the shared genetic architecture of Alzheimer's disease, cognition, and educational attainment, and associations with brain development. Neurobiol Dis 2024; 203:106750. [PMID: 39608471 DOI: 10.1016/j.nbd.2024.106750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 10/09/2024] [Accepted: 11/23/2024] [Indexed: 11/30/2024] Open
Abstract
The observation that the risk of developing Alzheimer's disease is reduced in individuals with high premorbid cognitive functioning, higher educational attainment, and occupational status has led to the 'cognitive reserve' hypothesis. This hypothesis suggests that individuals with greater cognitive reserve can tolerate a more significant burden of neuropathological changes before the onset of cognitive decline. The underpinnings of cognitive reserve remain poorly understood, although a shared genetic basis between measures of cognitive reserve and Alzheimer's disease has been suggested. Using the largest samples to date and novel statistical tools, we aimed to investigate shared genetic variants between Alzheimer's disease, and measures of cognitive reserve; cognition and educational attainment to identify molecular and neurobiological foundations. We applied the causal mixture model (MiXeR) to estimate the number of trait-influencing variants shared between Alzheimer's disease, cognition, and educational attainment, and condFDR/conjFDR to identify shared loci. To provide biological insights loci were functionally characterized. Subsequently, we constructed a Structural Equation Model (SEM) to determine if the polygenic foundation of cognition has a direct impact on Alzheimer's disease risk, or if its effect is mediated through established risk factors for the disease, using a case-control sample from the UK Biobank. Univariate MiXeR analysis (after excluding chromosome 19) revealed that Alzheimer's disease was substantially less polygenic (450 trait-influencing variants) compared to cognition (11,100 trait-influencing variants), and educational attainment (12,700 trait-influencing variants). Bivariate MiXeR analysis estimated that Alzheimer's disease shared approximately 70 % of trait-influencing variants with cognition, and approximately 40 % with educational attainment, with mixed effect directions. Using condFDR analysis, we identified 18 loci jointly associated with Alzheimer's disease and cognition and 6 loci jointly associated with Alzheimer's disease and educational attainment. Genes mapped to shared loci were associated with neurodevelopment, expressed in early life, and implicated the dendritic tree and phosphatidylinositol phosphate binding mechanisms. Spatiotemporal gene expression analysis of the identified genes showed that mapped genes were highly expressed during the mid-fetal period, further suggesting early neurodevelopmental stages as critical periods for establishing cognitive reserve which affect the risk of Alzheimer's disease in old age. Furthermore, our SEM analysis showed that genetic variants influencing cognition had a direct effect on the risk of developing Alzheimer's disease, providing evidence in support of the neurodevelopmental hypothesis of the disease.
Collapse
Affiliation(s)
- Piotr Jaholkowski
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Shahram Bahrami
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Vera Fominykh
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Guy F L Hindley
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Institute of Psychiatry, Psychology and Neuroscience, King's College London, UK
| | - Markos Tesfaye
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pravesh Parekh
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nadine Parker
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Tahir T Filiz
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kaja Nordengen
- Department of Neurology, Oslo University Hospital, Oslo, Norway
| | - Espen Hagen
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Elise Koch
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nora R Bakken
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Evgeniia Frei
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Viktoria Birkenæs
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Zillur Rahman
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Oleksandr Frei
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Center for Bioinformatics, Department of Informatics, University of Oslo, Oslo, Norway
| | - Jan Haavik
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway; Division of Psychiatry, Haukeland University Hospital, Bergen, Norway
| | - Srdjan Djurovic
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Anders M Dale
- Department of Radiology, University of California, San Diego, La Jolla, CA 92093, USA; Multimodal Imaging Laboratory, University of California San Diego, La Jolla, CA 92093, USA; Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA; Department of Neurosciences, University of California San Diego, La Jolla, CA 92093, USA
| | - Olav B Smeland
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kevin S O'Connell
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Alexey A Shadrin
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Ole A Andreassen
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; KG Jebsen Centre for Neurodevelopmental Disorders, University of Oslo and Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
8
|
Rahman AFMT, Bulbule S, Belayet JB, Benko A, Gottschalk CG, Frick DN, Arnold LA, Hossain MM, Roy A. JRM-28, a Novel HDAC2 Inhibitor, Upregulates Plasticity-Associated Proteins in Hippocampal Neurons and Enhances Morphological Plasticity via Activation of CREB: Implications for Alzheimer's Disease. Cells 2024; 13:1964. [PMID: 39682714 PMCID: PMC11640089 DOI: 10.3390/cells13231964] [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: 09/23/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Enhancement of neuronal plasticity by small-molecule therapeutics protects cognitive skills and also ameliorates progressive neurodegenerative pathologies like Alzheimer's disease (AD) and dementia. One such compound, a novel histone deacetylase 2 (HDAC2) inhibitor named JRM-28, was shown here to enhance dendritic strength, augment spine density, and upregulate post-synaptic neurotransmission in hippocampal neurons. The molecular basis for this effect correlates with JRM-28-induced upregulation of the transcription of cAMP response element-binding protein(CREB), induction of its transcriptional activity, and subsequent stimulation of expressions of CREB-dependent plasticity-associated genes, such as those encoding N-methyl-D-aspartate (NMDA) receptor subunit NR2A and the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor subunit GluR1. Specifically, JRM-28 stimulated the NMDA- and AMPA-receptor-sensitive ionotropic calcium influx in hippocampal neurons. Interestingly, JRM-28 did not induce NMDA- and AMPA-sensitive calcium influx in hippocampal neurons once the expression of CREB was knocked down by creb siRNA, suggesting the critical role of CREB in JRM-28-mediated upregulation of synaptic plasticity. Finally, JRM-28 upregulated CREB mRNA, CREB-dependent plasticity-associated markers, and ionotropic calcium influx in iPSC-derived AD human neurons, indicating its therapeutic implications in the amelioration of AD pathologies.
Collapse
Affiliation(s)
- A. F. M. Towheedur Rahman
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
| | - Sarojini Bulbule
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV 89451, USA
| | - Jawad Bin Belayet
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
| | - Anna Benko
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
- Milwaukee Institute for Drug Discovery, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA
| | - Carl Gunnar Gottschalk
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV 89451, USA
| | - David N. Frick
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
- Milwaukee Institute for Drug Discovery, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA
| | - Leggy A. Arnold
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
- Milwaukee Institute for Drug Discovery, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA
| | - M. Mahmun Hossain
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
- Milwaukee Institute for Drug Discovery, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA
| | - Avik Roy
- Department of Chemistry and Biochemistry, University of Wisconsin-Milwaukee, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA.; (A.F.M.T.R.); (S.B.); (J.B.B.); (A.B.); (C.G.G.); (D.N.F.); (L.A.A.)
- Simmaron Research Institute, 948 Incline Way, Incline Village, NV 89451, USA
- Milwaukee Institute for Drug Discovery, 2000 E Kenwood Blvd, Milwaukee, WI 53211, USA
- Simmaron Research and Development Laboratory, University of Wisconsin-Milwaukee, Chemistry Building, 2000 E Kenwood Blvd, Suite # 320, Milwaukee, WI 53211, USA
| |
Collapse
|
9
|
Kim H, Jang JW, Sim SE, Lee J, Jeong JH, Park S, Lee YK, Ham HJ, Yu NK, Lim CS, Gao FB, Lee JA, Kaang BK. Crucial role of Snf7-3 in synaptic function and cognitive behavior revealed by conventional and conditional knockout mouse models. Neuroscience 2024; 560:347-356. [PMID: 39369944 PMCID: PMC11975422 DOI: 10.1016/j.neuroscience.2024.10.010] [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: 06/04/2024] [Revised: 09/20/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
Snf7-3 is a crucial component of the endosomal sorting complexes required for transport (ESCRT) pathway, playing a vital role in endolysosomal functions. To elucidate the role of Snf7-3 in vivo, we developed conventional-like and conditional Snf7-3 knockout (KO) mouse models using a "Knockout-first" strategy. Conventional-like Snf7-3 KO mice showed significantly reduced Snf7-3 mRNA expression, and older mice (25-40 weeks) exhibited impaired social recognition and increased miniature excitatory postsynaptic currents (mEPSCs). Similarly, conditional KO mice aged 8-24 weeks, with Snf7-3 specifically deleted in forebrain excitatory neurons, displayed impaired object location memory and elevated mEPSC frequency. Consistently, Snf7-3 knockdown in cultured mouse hippocampal neurons led to increased densities of pre- and postsynaptic puncta, supporting the observed increase in mEPSC frequency. In addition, enhanced dendritic complexity was observed in the medial prefrontal cortex of these mice, indicating early synaptic disturbances. Our findings underscore the critical role of Snf7-3 in maintaining normal cognitive functions and social behaviors. The observed synaptic and behavioral deficits in both conventional-like and conditional KO mice highlight the importance of Snf7-3 in specific neuronal populations, suggesting that early synaptic changes could precede more pronounced cognitive impairments.
Collapse
Affiliation(s)
- Hyopil Kim
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Jae-Woo Jang
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon, Korea
| | - Su-Eon Sim
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Jisu Lee
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - June-Hyun Jeong
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Semin Park
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon, Korea
| | - You-Kyung Lee
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon, Korea
| | - Hyun-Ji Ham
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon, Korea
| | - Nam-Kyung Yu
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Chae-Seok Lim
- Department of Pharmacology, Wonkwang University School of Medicine, Jeonbuk 54538, South Korea
| | - Fen-Biao Gao
- RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jin-A Lee
- Department of Biological Sciences and Biotechnology, College of Life Sciences and Nanotechnology, Hannam University, Daejeon, Korea.
| | - Bong-Kiun Kaang
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea; Center for Cognition and Sociality, Life Science Institute, Institute for Basic Science (IBS), Daejeon 34141, South Korea.
| |
Collapse
|
10
|
Salazar CJ, Diaz-Balzac CA, Wang Y, Rahman M, Grant BD, Bülow HE. RABR-1, an atypical Rab-related GTPase, cell-nonautonomously restricts somatosensory dendrite branching. Genetics 2024; 228:iyae113. [PMID: 39028768 PMCID: PMC11457943 DOI: 10.1093/genetics/iyae113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
Neurons are highly polarized cells with dendrites and axons. Dendrites, which receive sensory information or input from other neurons, often display elaborately branched morphologies. While mechanisms that promote dendrite branching have been widely studied, less is known about the mechanisms that restrict branching. Using the nematode Caenorhabditis elegans, we identify rabr-1 (for Rab-related gene 1) as a factor that restricts branching of the elaborately branched dendritic trees of PVD and FLP somatosensory neurons. Animals mutant for rabr-1 show excessively branched dendrites throughout development and into adulthood in areas where the dendrites overlay epidermal tissues. Phylogenetic analyses show that RABR-1 displays similarity to small GTPases of the Rab-type, although based on sequence alone, no clear vertebrate ortholog of RABR-1 can be identified. We find that rabr-1 is expressed and can function in epidermal tissues, suggesting that rabr-1 restricts dendritic branching cell-nonautonomously. Genetic experiments further indicate that for the formation of ectopic branches rabr-1 mutants require the genes of the Menorin pathway, which have been previously shown to mediate dendrite morphogenesis of somatosensory neurons. A translational reporter for RABR-1 reveals a subcellular localization to punctate, perinuclear structures, which correlates with endosomal and autophagosomal markers, but anticorrelates with lysosomal markers suggesting an amphisomal character. Point mutations in rabr-1 analogous to key residues of small GTPases suggest that rabr-1 functions in a GTP-bound form independently of GTPase activity. Taken together, rabr-1 encodes for an atypical small GTPase of the Rab-type that cell-nonautonomously restricts dendritic branching of somatosensory neurons, likely independently of GTPase activity.
Collapse
Affiliation(s)
| | - Carlos A Diaz-Balzac
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Yu Wang
- Department of Molecular Biology and Biochemistry, Rutgers Center for Lipid Research, Rutgers University, Piscataway, NJ 08854, USA
| | - Maisha Rahman
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Barth D Grant
- Department of Molecular Biology and Biochemistry, Rutgers Center for Lipid Research, Rutgers University, Piscataway, NJ 08854, USA
| | - Hannes E Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
11
|
Zhong G, Fang Z, Sun T, Ying M, Wang A, Chen Y, Wang H, Ma C, Wang C, Ge R, Liu G, Guo Y. Ubiquitin ligase RFWD2 promotes dendritic spine and synapse formation by activating the ERK/PEA3/c-Jun pathway in rat cerebral cortical neurons. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167319. [PMID: 38909848 DOI: 10.1016/j.bbadis.2024.167319] [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: 11/23/2023] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
The regulation of protein degradation through the ubiquitin-proteasome system is essential for normal brain development, axon growth, synaptic growth and plasticity. The E3 ubiquitin ligase RFWD2 plays a key role in the onset and development of neurological diseases, including the pathogenesis of Alzheimer's disease (AD), but the mechanisms controlling the homeostasis of neuronal synaptic proteins are still poorly understood. Here, we showed that the expression level of RFWD2 gradually decreased with the age of the rats and was negatively correlated with the development of cerebral cortical neurons and dendrites in vivo. RFWD2 was shown to localize to presynaptic terminals and some postsynaptic sides of both excitatory synapses and inhibitory synapses via colocalization with neuronal synaptic proteins (SYN, PSD95, Vglut1 and GAD67). Overexpression of RFWD2 promoted dendrite development and dendritic spine formation and markedly decreased the expression of synaptophysin and PSD95 by reducing the expression of ETV1, ETV4, ETV5 and c-JUN in vitro. Furthermore, the whole-cell membrane slice clamp results showed that RFWD2 overexpression resulted in greater membrane capacitance in neuronal cells, inadequate cell repolarization, and a longer time course for neurons to emit action potentials with decreased excitability. RFWD2 regulates dendritic development and plasticity, dendritic spine formation and synaptic function in rat cerebral cortex neurons by activating the ERK/PEA3/c-Jun pathway via a posttranslational regulatory mechanism and can be used as an efficient treatment target for neurological diseases.
Collapse
Affiliation(s)
- Guangshang Zhong
- School of Laboratory Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China; School of Life Sciences, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Zhuling Fang
- School of Laboratory Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Tingting Sun
- School of Life Sciences, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Mengjiao Ying
- School of Life Sciences, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Ao Wang
- School of Laboratory Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Ying Chen
- School of Life Sciences, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Haojie Wang
- School of Clinical Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Caiyun Ma
- School of Life Sciences, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Chunjing Wang
- School of Life Sciences, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Rongjing Ge
- School of Clinical Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China; Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu, 233000, China
| | - Gaofeng Liu
- School of Life Sciences, Bengbu Medical University, Bengbu 233000, Anhui, China; Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu, 233000, China.
| | - Yu Guo
- School of Laboratory Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China; Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu, 233000, China.
| |
Collapse
|
12
|
Asad Z, Fakheir Y, Abukhaled Y, Khalil R. Implications of altered pyramidal cell morphology on clinical symptoms of neurodevelopmental disorders. Eur J Neurosci 2024; 60:4877-4892. [PMID: 39054743 DOI: 10.1111/ejn.16484] [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: 02/13/2024] [Revised: 05/26/2024] [Accepted: 07/13/2024] [Indexed: 07/27/2024]
Abstract
The prevalence of pyramidal cells (PCs) in the mammalian cerebral cortex underscore their value as they play a crucial role in various brain functions, ranging from cognition, sensory processing, to motor output. PC morphology significantly influences brain connectivity and plays a critical role in maintaining normal brain function. Pathological alterations to PC morphology are thought to contribute to the aetiology of neurodevelopmental disorders such as autism spectrum disorder (ASD) and schizophrenia. This review explores the relationship between abnormalities in PC morphology in key cortical areas and the clinical manifestations in schizophrenia and ASD. We focus largely on human postmortem studies and provide evidence that dendritic segment length, complexity and spine density are differentially affected in these disorders. These morphological alterations can lead to disruptions in cortical connectivity, potentially contributing to the cognitive and behavioural deficits observed in these disorders. Furthermore, we highlight the importance of investigating the functional and structural characteristics of PCs in these disorders to illuminate the underlying pathogenesis and stimulate further research in this area.
Collapse
Affiliation(s)
- Zummar Asad
- School of Medicine, Royal College of Surgeons in Ireland (RCSI), Dublin, Ireland
| | - Yara Fakheir
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Yara Abukhaled
- College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Reem Khalil
- Department of Biology, Chemistry and Environmental Sciences, American University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
13
|
Aksan B, Kenkel AK, Yan J, Sánchez Romero J, Missirlis D, Mauceri D. VEGFD signaling balances stability and activity-dependent structural plasticity of dendrites. Cell Mol Life Sci 2024; 81:354. [PMID: 39158743 PMCID: PMC11335284 DOI: 10.1007/s00018-024-05357-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 08/20/2024]
Abstract
Mature neurons have stable dendritic architecture, which is essential for the nervous system to operate correctly. The ability to undergo structural plasticity, required to support adaptive processes like memory formation, is still present in mature neurons. It is unclear what molecular and cellular processes control this delicate balance between dendritic structural plasticity and stabilization. Failures in the preservation of optimal dendrite structure due to atrophy or maladaptive plasticity result in abnormal connectivity and are associated with various neurological diseases. Vascular endothelial growth factor D (VEGFD) is critical for the maintenance of mature dendritic trees. Here, we describe how VEGFD affects the neuronal cytoskeleton and demonstrate that VEGFD exerts its effects on dendrite stabilization by influencing the actin cortex and reducing microtubule dynamics. Further, we found that during synaptic activity-induced structural plasticity VEGFD is downregulated. Our findings revealed that VEGFD, acting on its cognate receptor VEGFR3, opposes structural changes by negatively regulating dendrite growth in cultured hippocampal neurons and in vivo in the adult mouse hippocampus with consequences on memory formation. A phosphoproteomic screening identified several regulatory proteins of the cytoskeleton modulated by VEGFD. Among the actin cortex-associated proteins, we found that VEGFD induces dephosphorylation of ezrin at tyrosine 478 via activation of the striatal-enriched protein tyrosine phosphatase (STEP). Activity-triggered structural plasticity of dendrites was impaired by expression of a phospho-deficient mutant ezrin in vitro and in vivo. Thus, VEGFD governs the equilibrium between stabilization and plasticity of dendrites by acting as a molecular brake of structural remodeling.
Collapse
Affiliation(s)
- Bahar Aksan
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Ann-Kristin Kenkel
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Jing Yan
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Javier Sánchez Romero
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany
| | - Dimitris Missirlis
- Department of Cellular Biophysics, Max-Planck-Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany
| | - Daniela Mauceri
- Department of Neurobiology, Interdisciplinary Centre for Neurosciences (IZN), Heidelberg University, INF 366, 69120, Heidelberg, Germany.
- Department Molecular and Cellular Neuroscience, Institute of Anatomy and Cell Biology, University of Marburg, Robert-Koch-Str. 8, 35032, Marburg, Germany.
| |
Collapse
|
14
|
Akhgari A, Michel TM, Vafaee MS. Dendritic spines and their role in the pathogenesis of neurodevelopmental and neurological disorders. Rev Neurosci 2024; 35:489-502. [PMID: 38440811 DOI: 10.1515/revneuro-2023-0151] [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: 12/06/2023] [Accepted: 02/02/2024] [Indexed: 03/06/2024]
Abstract
Since Cajal introduced dendritic spines in the 19th century, they have attained considerable attention, especially in neuropsychiatric and neurologic disorders. Multiple roles of dendritic spine malfunction and pathology in the progression of various diseases have been reported. Thus, it is inevitable to consider these structures as new therapeutic targets for treating neuropsychiatric and neurologic disorders such as autism spectrum disorders, schizophrenia, dementia, Down syndrome, etc. Therefore, we attempted to prepare a narrative review of the literature regarding the role of dendritic spines in the pathogenesis of aforementioned diseases and to shed new light on their pathophysiology.
Collapse
Affiliation(s)
- Aisan Akhgari
- Student Research Committee, Tabriz University of Medical Sciences, Golgasht Street, Tabriz 5166616471, Iran
| | - Tanja Maria Michel
- Research Unit for Psychiatry, Odense University Hospital, J. B. Winsløws Vej 4, Odense 5000, Denmark
- Clinical Institute, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| | - Manouchehr Seyedi Vafaee
- Research Unit for Psychiatry, Odense University Hospital, J. B. Winsløws Vej 4, Odense 5000, Denmark
- Clinical Institute, University of Southern Denmark, Campusvej 55, Odense 5230, Denmark
| |
Collapse
|
15
|
Antipova V, Heimes D, Seidel K, Schulz J, Schmitt O, Holzmann C, Rolfs A, Bidmon HJ, González de San Román Martín E, Huesgen PF, Amunts K, Keiler J, Hammer N, Witt M, Wree A. Differently increased volumes of multiple brain areas in Npc1 mutant mice following various drug treatments. Front Neuroanat 2024; 18:1430790. [PMID: 39081805 PMCID: PMC11286580 DOI: 10.3389/fnana.2024.1430790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Background Niemann-Pick disease type C1 (NPC1, MIM 257220) is a heritable lysosomal storage disease characterized by a progressive neurological degeneration that causes disability and premature death. A murine model of Npc1-/- displays a rapidly progressing form of Npc1 disease, which is characterized by weight loss, ataxia, and increased cholesterol storage. Npc1-/- mice receiving a combined therapy (COMBI) of miglustat (MIGLU), the neurosteroid allopregnanolone (ALLO) and the cyclic oligosaccharide 2-hydroxypropyl-β-cyclodextrin (HPßCD) showed prevention of Purkinje cell loss, improved motor function and reduced intracellular lipid storage. Although therapy of Npc1-/- mice with COMBI, MIGLU or HPßCD resulted in the prevention of body weight loss, reduced total brain weight was not positively influenced. Methods In order to evaluate alterations of different brain areas caused by pharmacotherapy, fresh volumes (volumes calculated from the volumes determined from paraffin embedded brain slices) of various brain structures in sham- and drug-treated wild type and mutant mice were measured using stereological methods. Results In the wild type mice, the volumes of investigated brain areas were not significantly altered by either therapy. Compared with the respective wild types, fresh volumes of specific brain areas, which were significantly reduced in sham-treated Npc1-/- mice, partly increased after the pharmacotherapies in all treatment strategies; most pronounced differences were found in the CA1 area of the hippocampus and in olfactory structures. Discussion Volumes of brain areas of Npc1-/- mice were not specifically changed in terms of functionality after administering COMBI, MIGLU, or HPßCD. Measurements of fresh volumes of brain areas in Npc1-/- mice could monitor region-specific changes and response to drug treatment that correlated, in part, with behavioral improvements in this mouse model.
Collapse
Affiliation(s)
- Veronica Antipova
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
- Division of Macroscopic and Clinical Anatomy, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria
| | - Diana Heimes
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Mainz, Germany
| | - Katharina Seidel
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
- Klinik für Frauenheilkunde und Geburtshilfe, Dietrich-Bonhoeffer-Klinikum, Neubrandenburg, Germany
| | - Jennifer Schulz
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Oliver Schmitt
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
- Department of Anatomy, Medical School Hamburg, University of Applied Sciences and Medical University, Hamburg, Germany
| | - Carsten Holzmann
- Institute of Medical Genetics, Rostock University Medical Center, Rostock, Germany
- Centre of Transdisciplinary Neuroscience Rostock, Rostock, Germany
| | - Arndt Rolfs
- Medical Faculty, University of Rostock, Rostock, Germany
| | - Hans-Jürgen Bidmon
- Institute of Neurosciences and Medicine, Structural and Functional Organisation of the Brain (INM-1), Forschungszentrum Jülich, Jülich, Germany
- Central Institute of Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Jülich, Germany
| | | | - Pitter F. Huesgen
- Central Institute of Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Jülich, Germany
- Institut für Biologie II, AG Funktional Proteomics, Freiburg, Germany
| | - Katrin Amunts
- Institute of Neurosciences and Medicine, Structural and Functional Organisation of the Brain (INM-1), Forschungszentrum Jülich, Jülich, Germany
- C. and O. Vogt Institute for Brain Research, University Hospital Düsseldorf, University Düsseldorf, Düsseldorf, Germany
| | - Jonas Keiler
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Niels Hammer
- Division of Macroscopic and Clinical Anatomy, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria
- Department of Orthopedic and Trauma Surgery, University of Leipzig, Leipzig, Germany
- Division of Biomechatronics, Fraunhofer Institute for Machine Tools and Forming Technology, Dresden, Germany
| | - Martin Witt
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
- Department of Anatomy, Technische Universität Dresden, Dresden, Germany
- Department of Anatomy, Institute of Biostructural Basics of Medical Sciences, Poznan Medical University, Poznan, Poland
| | - Andreas Wree
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
- Centre of Transdisciplinary Neuroscience Rostock, Rostock, Germany
| |
Collapse
|
16
|
Brennand KJ. Aligning Stem Cell Models and Postmortem Studies to Query Striatal Neurodevelopment in Schizophrenia. Am J Psychiatry 2024; 181:465-467. [PMID: 38822585 DOI: 10.1176/appi.ajp.20240245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/03/2024]
Affiliation(s)
- Kristen J Brennand
- Departments of Psychiatry and Genetics, Division of Molecular Psychiatry, Yale University School of Medicine, New Haven, Conn
| |
Collapse
|
17
|
Kim Y, Ma R, Zhang Y, Kang HR, Kim US, Han K. Cell-autonomous reduction of CYFIP2 changes dendrite length, dendritic protrusion morphology, and inhibitory synapse density in the hippocampal CA1 pyramidal neurons of 17-month-old mice. Anim Cells Syst (Seoul) 2024; 28:294-302. [PMID: 38832126 PMCID: PMC11146249 DOI: 10.1080/19768354.2024.2360740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 05/22/2024] [Indexed: 06/05/2024] Open
Abstract
The cytoplasmic FMR1-interacting protein 2 (CYFIP2) have diverse molecular functions in neurons, including the regulation of actin polymerization, mRNA translation, and mitochondrial morphology and function. Mutations in the CYFIP2 gene are associated with early-onset epilepsy and neurodevelopmental disorders, while decreases in its protein levels are linked to Alzheimer's disease (AD). Notably, previous research has revealed AD-like phenotypes, such as dendritic spine loss, in the hippocampal CA1 pyramidal neurons of 12-month-old Cyfip2 heterozygous mice but not of age-matched CA1 pyramidal neuron-specific Cyfip2 conditional knock-out (cKO) mice. This study aims to investigate whether dendritic spine loss in Cyfip2 cKO mice is merely delayed compared to Cyfip2 heterozygous mice, and to explore further neuronal phenotypes regulated by CYFIP2 in aged mice. We characterized dendrite and dendritic protrusion morphologies, along with excitatory/inhibitory synapse densities in CA1 pyramidal neurons of 17-month-old Cyfip2 cKO mice. Overall dendritic branching was normal, with a reduction in the length of basal, not apical, dendrites in CA1 pyramidal neurons of Cyfip2 cKO mice. Furthermore, while dendritic protrusion density remained normal, alterations were observed in the length of mushroom spines and the head volume of stubby spines in basal, not apical, dendrites of Cyfip2 cKO mice. Although excitatory synapse density remained unchanged, inhibitory synapse density increased in apical, not basal, dendrites of Cyfip2 cKO mice. Consequently, a cell-autonomous reduction of CYFIP2 appears insufficient to induce dendritic spine loss in CA1 pyramidal neurons of aged mice. However, CYFIP2 is required to maintain normal dendritic length, dendritic protrusion morphology, and inhibitory synapse density.
Collapse
Affiliation(s)
- Yoonhee Kim
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ruiying Ma
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Yinhua Zhang
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hyae Rim Kang
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - U Suk Kim
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
| | - Kihoon Han
- Department of Neuroscience, Korea University College of Medicine, Seoul, Republic of Korea
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
18
|
Zhang W, Tariq M, Roy B, Shen J, Khan A, Altaf Malik N, He S, Baig SM, Fang X, Zhang J. Whole exome sequencing identified a homozygous novel variant in DOP1A gene in the Pakistan family with neurodevelopmental disabilities: case report and literature review. Front Genet 2024; 15:1351710. [PMID: 38818041 PMCID: PMC11137318 DOI: 10.3389/fgene.2024.1351710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 04/11/2024] [Indexed: 06/01/2024] Open
Abstract
Background Hereditary neurodevelopmental disorders (NDDs) are prevalent in poorly prognostic pediatric diseases, but the pathogenesis of NDDs is still unclear. Irregular myelination could be one of the possible causes of NDDs. Case presentation Here, whole exome sequencing was carried out for a consanguineous Pakistani family with NDDs to identify disease-associated variants. The co-segregation of candidate variants in the family was validated using Sanger sequencing. The potential impact of the gene on NDDs has been supported by conservation analysis, protein prediction, and expression analysis. A novel homozygous variant DOP1A(NM_001385863.1):c.2561A>G was identified. It was concluded that the missense variant might affect the protein-protein binding sites of the critical MEC interaction region of DOP1A, and DOP1A-MON2 may cause stability deficits in Golgi-endosome protein traffic. Proteolipid protein (PLP) and myelin-associate glycoprotein (MAG) could be targets of the DOP1A-MON2 Golgi-endosome traffic complex, especially during the fetal stage and the early developmental stages. This further supports the perspective that disorganized myelinogenesis due to congenital DOP1A deficiency might cause neurodevelopmental disorders (NDDs). Conclusion Our case study revealed the potential pathway of myelinogenesis-relevant NDDs and identified DOP1A as a potential NDDs-relevant gene in humans.
Collapse
Affiliation(s)
- Wei Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- BGI Genomics, Shenzhen, China
| | - Muhammad Tariq
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C PIEAS), Faisalabad, Pakistan
| | - Bhaskar Roy
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Juan Shen
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Ayaz Khan
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C PIEAS), Faisalabad, Pakistan
| | - Naveed Altaf Malik
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C PIEAS), Faisalabad, Pakistan
| | - Sijie He
- Hebei Industrial Technology Research Institute of Genomics in Maternal and Child Health, Shijiazhuang, China
- Clin Lab, BGI Genomics, Shijiazhuang, China
| | - Shahid Mahmood Baig
- National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C PIEAS), Faisalabad, Pakistan
| | - Xiaodong Fang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- China National GeneBank, BGI-Shenzhen, Shenzhen, China
| | - Jianguo Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
19
|
Sahly AN, Sierra-Marquez J, Bungert-Plümke S, Franzen A, Mougharbel L, Berrahmoune S, Dassi C, Poulin C, Srour M, Guzman RE, Myers KA. Genotype-phenotype correlation in CLCN4-related developmental and epileptic encephalopathy. Hum Genet 2024; 143:667-681. [PMID: 38578438 DOI: 10.1007/s00439-024-02668-z] [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: 02/17/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024]
Abstract
CLCN4-related disorder is a rare X-linked neurodevelopmental condition with a pathogenic mechanism yet to be elucidated. CLCN4 encodes the vesicular 2Cl-/H+ exchanger ClC-4, and CLCN4 pathogenic variants frequently result in altered ClC-4 transport activity. The precise cellular and molecular function of ClC-4 remains unknown; however, together with ClC-3, ClC-4 is thought to have a role in the ion homeostasis of endosomes and intracellular trafficking. We reviewed our research database for patients with CLCN4 variants and epilepsy, and performed thorough phenotyping. We examined the functional properties of the variants in mammalian cells using patch-clamp electrophysiology, protein biochemistry, and confocal fluorescence microscopy. Three male patients with developmental and epileptic encephalopathy were identified, with differing phenotypes. Patients #1 and #2 had normal growth parameters and normal-appearing brains on MRI, while patient #3 had microcephaly, microsomia, complete agenesis of the corpus callosum and cerebellar and brainstem hypoplasia. The p.(Gly342Arg) variant of patient #1 significantly impaired ClC-4's heterodimerization capability with ClC-3 and suppressed anion currents. The p.(Ile549Leu) variant of patient #2 and p.(Asp89Asn) variant of patient #3 both shift the voltage dependency of transport activation by 20 mV to more hyperpolarizing potentials, relative to the wild-type, with p.(Asp89Asn) favouring higher transport activity. We concluded that p.(Gly342Arg) carried by patient #1 and the p.(Ile549Leu) expressed by patient #2 impair ClC-4 transport function, while the p.(Asp89Asn) variant results in a gain-of-transport function; all three variants result in epilepsy and global developmental impairment, but with differences in epilepsy presentation, growth parameters, and presence or absence of brain malformations.
Collapse
Affiliation(s)
- Ahmed N Sahly
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC, Canada
- Department of Neurosciences, King Faisal Specialist Hospital & Research Centre, Jeddah, Saudi Arabia
| | - Juan Sierra-Marquez
- Institute of Biological Information Processing; Biological Molecular and Cell Physiology (IBI-1), Molecular and Cell Physiology, Research Center Jülich , GmbH Leo-Brandt-Strasse 1, 52428, Jülich, Germany
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stefanie Bungert-Plümke
- Institute of Biological Information Processing; Biological Molecular and Cell Physiology (IBI-1), Molecular and Cell Physiology, Research Center Jülich , GmbH Leo-Brandt-Strasse 1, 52428, Jülich, Germany
| | - Arne Franzen
- Institute of Biological Information Processing; Biological Molecular and Cell Physiology (IBI-1), Molecular and Cell Physiology, Research Center Jülich , GmbH Leo-Brandt-Strasse 1, 52428, Jülich, Germany
| | - Lina Mougharbel
- Research Institute of the McGill University Medical Centre, Montreal, QC, Canada
| | - Saoussen Berrahmoune
- Research Institute of the McGill University Medical Centre, Montreal, QC, Canada
| | - Christelle Dassi
- Research Institute of the McGill University Medical Centre, Montreal, QC, Canada
| | - Chantal Poulin
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, 1001 Boulevard Décarie, Montreal, PQ, H4A 3J1, Canada
| | - Myriam Srour
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC, Canada
- Research Institute of the McGill University Medical Centre, Montreal, QC, Canada
- Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, 1001 Boulevard Décarie, Montreal, PQ, H4A 3J1, Canada
| | - Raul E Guzman
- Institute of Biological Information Processing; Biological Molecular and Cell Physiology (IBI-1), Molecular and Cell Physiology, Research Center Jülich , GmbH Leo-Brandt-Strasse 1, 52428, Jülich, Germany.
| | - Kenneth A Myers
- Division of Neurology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC, Canada.
- Research Institute of the McGill University Medical Centre, Montreal, QC, Canada.
- Department of Neurology and Neurosurgery, Montreal Children's Hospital, McGill University Health Centre, 1001 Boulevard Décarie, Montreal, PQ, H4A 3J1, Canada.
| |
Collapse
|
20
|
Zeng Y, Wang Y. Complete Neuron Reconstruction Based on Branch Confidence. Brain Sci 2024; 14:396. [PMID: 38672045 PMCID: PMC11047972 DOI: 10.3390/brainsci14040396] [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: 03/04/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
In the past few years, significant advancements in microscopic imaging technology have led to the production of numerous high-resolution images capturing brain neurons at the micrometer scale. The reconstructed structure of neurons from neuronal images can serve as a valuable reference for research in brain diseases and neuroscience. Currently, there lacks an accurate and efficient method for neuron reconstruction. Manual reconstruction remains the primary approach, offering high accuracy but requiring significant time investment. While some automatic reconstruction methods are faster, they often sacrifice accuracy and cannot be directly relied upon. Therefore, the primary goal of this paper is to develop a neuron reconstruction tool that is both efficient and accurate. The tool aids users in reconstructing complete neurons by calculating the confidence of branches during the reconstruction process. The method models the neuron reconstruction as multiple Markov chains, and calculates the confidence of the connections between branches by simulating the reconstruction artifacts in the results. Users iteratively modify low-confidence branches to ensure precise and efficient neuron reconstruction. Experiments on both the publicly accessible BigNeuron dataset and a self-created Whole-Brain dataset demonstrate that the tool achieves high accuracy similar to manual reconstruction, while significantly reducing reconstruction time.
Collapse
Affiliation(s)
- Ying Zeng
- School of Computer Science and Technology, Shanghai University, Shanghai 200444, China;
- Guangdong Institute of Intelligence Science and Technology, Zhuhai 519031, China
| | - Yimin Wang
- Guangdong Institute of Intelligence Science and Technology, Zhuhai 519031, China
| |
Collapse
|
21
|
Qian P, Manubens-Gil L, Jiang S, Peng H. Non-homogenous axonal bouton distribution in whole-brain single-cell neuronal networks. Cell Rep 2024; 43:113871. [PMID: 38451816 DOI: 10.1016/j.celrep.2024.113871] [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: 09/09/2023] [Revised: 01/08/2024] [Accepted: 02/09/2024] [Indexed: 03/09/2024] Open
Abstract
We examined the distribution of pre-synaptic contacts in axons of mouse neurons and constructed whole-brain single-cell neuronal networks using an extensive dataset of 1,891 fully reconstructed neurons. We found that bouton locations were not homogeneous throughout the axon and among brain regions. As our algorithm was able to generate whole-brain single-cell connectivity matrices from full morphology reconstruction datasets, we further found that non-homogeneous bouton locations have a significant impact on network wiring, including degree distribution, triad census, and community structure. By perturbing neuronal morphology, we further explored the link between anatomical details and network topology. In our in silico exploration, we found that dendritic and axonal tree span would have the greatest impact on network wiring, followed by synaptic contact deletion. Our results suggest that neuroanatomical details must be carefully addressed in studies of whole-brain networks at the single-cell level.
Collapse
Affiliation(s)
- Penghao Qian
- New Cornerstone Science Laboratory, SEU-ALLEN Joint Center, State Key Laboratory of Digital Medical Engineering, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu 210096, China; School of Computer Science and Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Linus Manubens-Gil
- New Cornerstone Science Laboratory, SEU-ALLEN Joint Center, State Key Laboratory of Digital Medical Engineering, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu 210096, China.
| | - Shengdian Jiang
- New Cornerstone Science Laboratory, SEU-ALLEN Joint Center, State Key Laboratory of Digital Medical Engineering, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu 210096, China; School of Computer Science and Engineering, Southeast University, Nanjing, Jiangsu 210096, China
| | - Hanchuan Peng
- New Cornerstone Science Laboratory, SEU-ALLEN Joint Center, State Key Laboratory of Digital Medical Engineering, Institute for Brain and Intelligence, Southeast University, Nanjing, Jiangsu 210096, China.
| |
Collapse
|
22
|
Cilingir V, Seven E. Retinal clues for selective neuronal loss in multiple sclerosis. Neurol Sci 2024; 45:1163-1171. [PMID: 37837508 DOI: 10.1007/s10072-023-07110-2] [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: 05/03/2023] [Accepted: 09/30/2023] [Indexed: 10/16/2023]
Abstract
OBJECTIVE The relationship between the cell body layer and the dendritic network layer of the retina and cognitive performance (CP) in MS patients has not been examined separately. The objective of this study is to predict cognitive impairment (CI) in RRMS patients and to examine the relationship between CP and ganglion cell layer (GCL), inner plexiform layer (IPL), and GCL divided by IPL (GCL/IPL). METHODS Ophthalmological evaluation, retinal segmentation, and Symbol Digit Modalities Test (SDMT) were performed on 102 RRMS patients and 54 healthy subjects. The relationships of GCL, IPL, and GCL/IPL with CP in eyes without a history of optic neuritis were investigated using Spearman's correlation. Models were created by accepting 1 standard deviation less of the SDMT mean of the control group as the limit for CI. The cutoff value of the GCL/IPL variable that could predict CI was calculated by ROC analysis, and the ability to accurately predict CI was tested with binary logistic regression. RESULTS No correlation was found between OCT parameters and CP in healthy subjects. Correlation was found between GCL thickness and GCL/IPL variable and CP in RRMS patients (r=0.235, r=0.667 respectively). A GCL/IPL value of 1.255 was able to identify CI with 81.8% sensitivity and 75.9% specificity (AUC=0.844, LR=3.38) and predicted CI with 74.5% accuracy (Nagelkerke R2=0.439). CONCLUSION In RRMS patients, the IPL thickness is unrelated to CP. Therewithal, the GCL/IPL-CP relationship is stronger than the GCL-CP relationship and GCL/IPL can predict CI.
Collapse
Affiliation(s)
- Vedat Cilingir
- Department of Neurology, Van Yuzuncu Yil University, Bardakcı, Tusba, 65300, Van, Turkey.
- Department of Neuroscience Research, Van Yuzuncu Yil University, Tusba, Van, Turkey.
| | - Erbil Seven
- Department of Ophthalmology, Van Yuzuncu Yil University, Tusba, Van, Turkey
| |
Collapse
|
23
|
Izquierdo-Villalba I, Mirra S, Manso Y, Parcerisas A, Rubio J, Del Valle J, Gil-Bea FJ, Ulloa F, Herrero-Lorenzo M, Verdaguer E, Benincá C, Castro-Torres RD, Rebollo E, Marfany G, Auladell C, Navarro X, Enríquez JA, López de Munain A, Soriano E, Aragay AM. A mammalian-specific Alex3/Gα q protein complex regulates mitochondrial trafficking, dendritic complexity, and neuronal survival. Sci Signal 2024; 17:eabq1007. [PMID: 38320000 DOI: 10.1126/scisignal.abq1007] [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: 03/17/2022] [Accepted: 01/16/2024] [Indexed: 02/08/2024]
Abstract
Mitochondrial dynamics and trafficking are essential to provide the energy required for neurotransmission and neural activity. We investigated how G protein-coupled receptors (GPCRs) and G proteins control mitochondrial dynamics and trafficking. The activation of Gαq inhibited mitochondrial trafficking in neurons through a mechanism that was independent of the canonical downstream PLCβ pathway. Mitoproteome analysis revealed that Gαq interacted with the Eutherian-specific mitochondrial protein armadillo repeat-containing X-linked protein 3 (Alex3) and the Miro1/Trak2 complex, which acts as an adaptor for motor proteins involved in mitochondrial trafficking along dendrites and axons. By generating a CNS-specific Alex3 knockout mouse line, we demonstrated that Alex3 was required for the effects of Gαq on mitochondrial trafficking and dendritic growth in neurons. Alex3-deficient mice had altered amounts of ER stress response proteins, increased neuronal death, motor neuron loss, and severe motor deficits. These data revealed a mammalian-specific Alex3/Gαq mitochondrial complex, which enables control of mitochondrial trafficking and neuronal death by GPCRs.
Collapse
Affiliation(s)
| | - Serena Mirra
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBER-CIBERER), ISCIII, Madrid 28031, Spain
- Institut de Biomedicina- Institut de Recerca Sant Joan de Déu (IBUB-IRSJD), Universitat de Barcelona, Barcelona 08028, Spain
| | - Yasmina Manso
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
| | - Antoni Parcerisas
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
- Biosciences Department, Faculty of Sciences, Technology and Engineering, University of Vic, Central University of Catalonia (UVic-UCC); and Tissue Repair and Regeneration Laboratory (TR2Lab), Institut de Recerca i Innovació en Ciències de la Vida i de la Salut a la Catalunya Central (IRIS-CC), 08500 Vic, Spain
| | - Javier Rubio
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona 08028, Spain
| | - Jaume Del Valle
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, Universitat Autonoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - Francisco J Gil-Bea
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián 20014, Spain
| | - Fausto Ulloa
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
| | - Marina Herrero-Lorenzo
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
| | - Ester Verdaguer
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
| | - Cristiane Benincá
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona 08028, Spain
| | - Rubén D Castro-Torres
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
| | - Elena Rebollo
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona 08028, Spain
| | - Gemma Marfany
- Department of Genetics, Microbiology and Statistics, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Raras (CIBER-CIBERER), ISCIII, Madrid 28031, Spain
- Institut de Biomedicina- Institut de Recerca Sant Joan de Déu (IBUB-IRSJD), Universitat de Barcelona, Barcelona 08028, Spain
| | - Carme Auladell
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
| | - Xavier Navarro
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, Universitat Autonoma de Barcelona, Bellaterra, Barcelona 08193, Spain
| | - José A Enríquez
- Fundación Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid 28029, Spain
- Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBER-CIBERFES), Madrid 28031, Spain
| | - Adolfo López de Munain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián 20014, Spain
- Neurology Department, Donostia University Hospital, San Sebastián 20014, Spain
| | - Eduardo Soriano
- Department of Cell Biology, Physiology and Immunology, and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBER-CIBERNED), ISCIII, Madrid 28031, Spain
| | - Anna M Aragay
- Institut de Biologia Molecular de Barcelona (IBMB-CSIC), Barcelona 08028, Spain
| |
Collapse
|
24
|
Diaz JR, Sadowski MJ. Analysis of Neurite and Spine Formation in Neurons In Vitro. Methods Mol Biol 2024; 2831:21-37. [PMID: 39134841 PMCID: PMC11684374 DOI: 10.1007/978-1-0716-3969-6_3] [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: 01/06/2025]
Abstract
Primary neuronal cultures are commonly used to study genetic and exogenous factors influencing neuronal development and maturation. During development, neurons undergo robust morphological changes involving expansion of dendritic arbor, formation of dendritic spines, and expression of synaptic proteins. In this chapter, we will cover methodological approaches allowing quantitative assessment of in vitro cultured neurons. Various quantitative characteristics of dendritic arbor can be derived based on immunostaining against anti-microtubule-associated protein 2 followed by dendrite tracing with the SNT plug-in of the FIJI software package. The number and subtypes of dendritic spines can be assessed by double labeling with DiI and Phalloidin iFluor448 followed by laser scanning confocal microscopy analysis. Finally, expression of presynaptic and postsynaptic proteins can be determined by immunohistochemistry and quantification using several available software packages including FIJI and Imaris, which also allows for 3D rendering and statistical displaying of the expression level of synaptic proteins.
Collapse
Affiliation(s)
- Jenny R Diaz
- Departments of Neurology, New York University Grossman School of Medicine, New York, NY, USA
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, United States
| | - Martin J Sadowski
- Departments of Neurology, New York University Grossman School of Medicine, New York, NY, USA.
- Departments of Psychiatry, New York University Grossman School of Medicine, New York, NY, USA.
- Departments of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
25
|
Liu S, Yan Z, Peng Y, Liu Y, Li Y, Xu D, Gong Y, Cui Z, Wu Y, Zhang Y, Wang D, Pan W, Yang X. Lentinan has a beneficial effect on cognitive deficits induced by chronic Toxoplasma gondii infection in mice. Parasit Vectors 2023; 16:454. [PMID: 38093309 PMCID: PMC10717010 DOI: 10.1186/s13071-023-06023-5] [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: 04/29/2023] [Accepted: 10/19/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Toxoplasma gondii (T. gondii) is increasingly considered a risk factor for neurodegenerative diseases. However, there is only limited information on the development of drugs for T. gondii infection. Lentinan from Lentinula edodes is a bioactive ingredient with the potential to enhance anti-infective immunity. The present study aimed to investigate the neuroprotective effect of lentinan on T. gondii-associated cognitive deficits in mice. METHODS A chronic T. gondii infection mouse model was established by administering 10 cysts of T. gondii by gavage. Lentinan was intraperitoneally administered 2 weeks before infection. Behavioral tests, RNA sequencing, immunofluorescence, transmission electron microscopy and Golgi-Cox staining were performed to assess the effect of lentinan on cognitive deficits and neuropathology in vivo. In vitro, the direct and indirect effects of lentinan on the proliferation of T. gondii tachyzoites were evaluated in the absence and presence of BV-2 cells, respectively. RESULTS Lentinan prevented T. gondii-induced cognitive deficits and altered the transcriptome profile of genes related to neuroinflammation, microglial activation, synaptic function, neural development and cognitive behavior in the hippocampus of infected mice. Moreover, lentinan reduced the infection-induced accumulation of microglia and downregulated the mRNA expression of proinflammatory cytokines. In addition, the neurite and synaptic ultrastructural damage in the hippocampal CA1 region due to infection was ameliorated by lentinan administration. Lentinan decreased the cyst burden in the brains of infected mice, which was correlated with behavioral performance. In line with this finding, lentinan could significantly inhibit the proliferation of T. gondii tachyzoites in the microglial cell line BV2, although lentinan had no direct inhibitory effect on parasite growth. CONCLUSIONS Lentinan prevents cognitive deficits via the improvement of neurite impairment and synaptic loss induced by T. gondii infection, which may be associated with decreased cyst burden in the brain. Overall, our findings indicate that lentinan can ameliorate T. gondii-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Shuxi Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ziyi Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuan Peng
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yunqiu Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yiling Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Daxiang Xu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yuying Gong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Zeyu Cui
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yongshui Wu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yumei Zhang
- Department of Pathogenic Biology, Binzhou Medical University, Binzhou, 256603, Shandong, China
| | - Dahui Wang
- Liangshan College (Li Shui) China, Lishui University, Lishui, 323000, Zhejiang, China.
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Jiangsu International Laboratory of Immunity and Metabolism, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
- National Experimental Demonstration Center for Basic Medicine Education, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
26
|
Di Credico A, Weiss A, Corsini M, Gaggi G, Ghinassi B, Wilbertz JH, Di Baldassarre A. Machine learning identifies phenotypic profile alterations of human dopaminergic neurons exposed to bisphenols and perfluoroalkyls. Sci Rep 2023; 13:21907. [PMID: 38081991 PMCID: PMC10713827 DOI: 10.1038/s41598-023-49364-y] [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: 09/21/2023] [Accepted: 12/07/2023] [Indexed: 12/18/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease and is characterized by the loss of midbrain dopaminergic neurons. Endocrine disrupting chemicals (EDCs) are active substances that interfere with hormonal signaling. Among EDCs, bisphenols (BPs) and perfluoroalkyls (PFs) are chemicals leached from plastics and other household products, and humans are unavoidably exposed to these xenobiotics. Data from animal studies suggest that EDCs exposure may play a role in PD, but data about the effect of BPs and PFs on human models of the nervous system are lacking. Previous studies demonstrated that machine learning (ML) applied to microscopy data can classify different cell phenotypes based on image features. In this study, the effect of BPs and PFs at different concentrations within the real-life exposure range (0.01, 0.1, 1, and 2 µM) on the phenotypic profile of human stem cell-derived midbrain dopaminergic neurons (mDANs) was analyzed. Cells exposed for 72 h to the xenobiotics were stained with neuronal markers and evaluated using high content microscopy yielding 126 different phenotypic features. Three different ML models (LDA, XGBoost and LightGBM) were trained to classify EDC-treated versus control mDANs. EDC treated mDANs were identified with high accuracies (0.88-0.96). Assessment of the phenotypic feature contribution to the classification showed that EDCs induced a significant increase of alpha-synuclein (αSyn) and tyrosine hydroxylase (TH) staining intensity within the neurons. Moreover, microtubule-associated protein 2 (MAP2) neurite length and branching were significantly diminished in treated neurons. Our study shows that human mDANs are adversely impacted by exposure to EDCs, causing their phenotype to shift and exhibit more characteristics of PD. Importantly, ML-supported high-content imaging can identify concrete but subtle subcellular phenotypic changes that can be easily overlooked by visual inspection alone and that define EDCs effects in mDANs, thus enabling further pathological characterization in the future.
Collapse
Affiliation(s)
- Andrea Di Credico
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies, and Technology (CAST), 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UdATech Lab Center (UdATech), 66100, Chieti, Italy
| | | | - Massimo Corsini
- Dipartimento Di Neuroscienze Umane, "Sapienza" University of Rome, Chieti, Italy
| | - Giulia Gaggi
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies, and Technology (CAST), 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UdATech Lab Center (UdATech), 66100, Chieti, Italy
| | - Barbara Ghinassi
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies, and Technology (CAST), 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UdATech Lab Center (UdATech), 66100, Chieti, Italy
| | | | - Angela Di Baldassarre
- Reprogramming and Cell Differentiation Lab, Center for Advanced Studies, and Technology (CAST), 66100, Chieti, Italy
- Department of Medicine and Aging Sciences, "G. D'Annunzio" University of Chieti-Pescara, 66100, Chieti, Italy
- UdATech Lab Center (UdATech), 66100, Chieti, Italy
| |
Collapse
|
27
|
Tat L, Cannizzaro N, Schaaf Z, Racherla S, Bottiglieri T, Green R, Zarbalis KS. Prenatal folic acid and vitamin B 12 imbalance alter neuronal morphology and synaptic density in the mouse neocortex. Commun Biol 2023; 6:1133. [PMID: 37938221 PMCID: PMC10632462 DOI: 10.1038/s42003-023-05492-9] [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: 01/30/2023] [Accepted: 10/19/2023] [Indexed: 11/08/2023] Open
Abstract
Previous reports have provided evidence that insufficient or excessive maternal folic acid (FA) intake during pregnancy can alter neurodevelopment of the offspring by modulating prenatal neurogenesis. Furthermore, our earlier work in a mouse model confirmed long-term structural changes at the cellular level of either deficient or excessive FA supply by comparably reducing dendritic arborization of cortical projection neurons. Here, we report that excessive amounts of FA decrease arborization of deep layer projection neurons, but not upper layer neurons and that reduced complexity of deep layer neurons is not observed when folic acid is replaced by folinic acid, a stable reduced form of folate. In addition, deficiency of B12, a vitamin that critically regulates folate metabolism, causes even more marked decreases in neuronal arborization in both deep and upper layer neurons and particularly in combination with FA excess. Furthermore, both FA excess and B12 deficiency affect synaptic density and morphology. Our findings point to neurodevelopmental risks associated with insufficient amounts of prenatal B12, particularly in association with high levels of FA intake, suggesting that the neurodevelopmental program is sensitive to an imbalance in the status of these interacting micronutrients.
Collapse
Affiliation(s)
- Lyvin Tat
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, 95817, USA
| | - Noemi Cannizzaro
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, 95817, USA
| | - Zachary Schaaf
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, 95817, USA
| | - Shailaja Racherla
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, 95817, USA
| | - Teodoro Bottiglieri
- Baylor Scott & White Research Institute, Center of Metabolomics, 3434 Live Oak, Dallas, TX, 75204, USA
| | - Ralph Green
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, 2425 Stockton Boulevard, Sacramento, CA, 95817, USA.
| | - Konstantinos S Zarbalis
- Department of Pathology and Laboratory Medicine, University of California, Davis, CA, 95817, USA.
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Northern California, 2425 Stockton Boulevard, Sacramento, CA, 95817, USA.
- MIND Institute, University of California, Davis, CA, 95817, USA.
| |
Collapse
|
28
|
Ebrahimi M, Thompson P, Lauer AK, Sivaprasad S, Perry G. The retina-brain axis and diabetic retinopathy. Eur J Ophthalmol 2023; 33:2079-2095. [PMID: 37259525 DOI: 10.1177/11206721231172229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Diabetic retinopathy (DR) is a major contributor to permanent vision loss and blindness. Changes in retinal neurons, glia, and microvasculature have been the focus of intensive study in the quest to better understand DR. However, the impact of diabetes on the rest of the visual system has received less attention. There are reports of associations of changes in the visual system with preclinical and clinical manifestations of diabetes. Simultaneous investigation of the retina and the brain may shed light on the mechanisms underlying neurodegeneration in diabetics. Additionally, investigating the links between DR and other neurodegenerative disorders of the brain including Alzheimer's and Parkinson's disease may reveal shared mechanisms for neurodegeneration and potential therapy options.
Collapse
Affiliation(s)
- Moein Ebrahimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy, and Autoimmunity, Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Paul Thompson
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging and Informatics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Andreas K Lauer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA
| | - Sobha Sivaprasad
- National Institute of Health and Care Research Moorfields Biomedical Research Centre, Moorfields Eye Hospital, London, UK
| | - George Perry
- Department of Neuroscience, Developmental and Regenerative Biology, University of Texas and San Antonio, San Antonio, TX, USA
| |
Collapse
|
29
|
Chen M, Xu L, Wu Y, Soba P, Hu C. The organization and function of the Golgi apparatus in dendrite development and neurological disorders. Genes Dis 2023; 10:2425-2442. [PMID: 37554209 PMCID: PMC10404969 DOI: 10.1016/j.gendis.2022.11.009] [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: 07/04/2022] [Revised: 09/13/2022] [Accepted: 11/05/2022] [Indexed: 12/24/2022] Open
Abstract
Dendrites are specialized neuronal compartments that sense, integrate and transfer information in the neural network. Their development is tightly controlled and abnormal dendrite morphogenesis is strongly linked to neurological disorders. While dendritic morphology ranges from relatively simple to extremely complex for a specified neuron, either requires a functional secretory pathway to continually replenish proteins and lipids to meet dendritic growth demands. The Golgi apparatus occupies the center of the secretory pathway and is regulating posttranslational modifications, sorting, transport, and signal transduction, as well as acting as a non-centrosomal microtubule organization center. The neuronal Golgi apparatus shares common features with Golgi in other eukaryotic cell types but also forms distinct structures known as Golgi outposts that specifically localize in dendrites. However, the organization and function of Golgi in dendrite development and its impact on neurological disorders is just emerging and so far lacks a systematic summary. We describe the organization of the Golgi apparatus in neurons, review the current understanding of Golgi function in dendritic morphogenesis, and discuss the current challenges and future directions.
Collapse
Affiliation(s)
- Meilan Chen
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
- Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510320, China
| | - Lu Xu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
| | - Yi Wu
- Department of Ophthalmology, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510320, China
| | - Peter Soba
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Bonn 53115, Germany
- Institute of Physiology and Pathophysiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Chun Hu
- Key Laboratory of Brain, Cognition and Education Sciences, Ministry of Education Institute for Brain, Science and Rehabilitation, South China Normal University, Guangzhou, Guangdong 510631, China
| |
Collapse
|
30
|
Tello JA, Jiang L, Zohar Y, Restifo LL. Drosophila CASK regulates brain size and neuronal morphogenesis, providing a genetic model of postnatal microcephaly suitable for drug discovery. Neural Dev 2023; 18:6. [PMID: 37805506 PMCID: PMC10559581 DOI: 10.1186/s13064-023-00174-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 09/08/2023] [Indexed: 10/09/2023] Open
Abstract
BACKGROUND CASK-related neurodevelopmental disorders are untreatable. Affected children show variable severity, with microcephaly, intellectual disability (ID), and short stature as common features. X-linked human CASK shows dosage sensitivity with haploinsufficiency in females. CASK protein has multiple domains, binding partners, and proposed functions at synapses and in the nucleus. Human and Drosophila CASK show high amino-acid-sequence similarity in all functional domains. Flies homozygous for a hypomorphic CASK mutation (∆18) have motor and cognitive deficits. A Drosophila genetic model of CASK-related disorders could have great scientific and translational value. METHODS We assessed the effects of CASK loss of function on morphological phenotypes in Drosophila using established genetic, histological, and primary neuronal culture approaches. NeuronMetrics software was used to quantify neurite-arbor morphology. Standard nonparametric statistics methods were supplemented by linear mixed effects modeling in some cases. Microfluidic devices of varied dimensions were fabricated and numerous fluid-flow parameters were used to induce oscillatory stress fields on CNS tissue. Dissociation into viable neurons and neurite outgrowth in vitro were assessed. RESULTS We demonstrated that ∆18 homozygous flies have small brains, small heads, and short bodies. When neurons from developing CASK-mutant CNS were cultured in vitro, they grew small neurite arbors with a distinctive, quantifiable "bushy" morphology that was significantly rescued by transgenic CASK+. As in humans, the bushy phenotype showed dosage-sensitive severity. To overcome the limitations of manual tissue trituration for neuronal culture, we optimized the design and operation of a microfluidic system for standardized, automated dissociation of CNS tissue into individual viable neurons. Neurons from CASK-mutant CNS dissociated in the microfluidic system recapitulate the bushy morphology. Moreover, for any given genotype, device-dissociated neurons grew larger arbors than did manually dissociated neurons. This automated dissociation method is also effective for rodent CNS. CONCLUSIONS These biological and engineering advances set the stage for drug discovery using the Drosophila model of CASK-related disorders. The bushy phenotype provides a cell-based assay for compound screening. Nearly a dozen genes encoding CASK-binding proteins or transcriptional targets also have brain-development mutant phenotypes, including ID. Hence, drugs that improve CASK phenotypes might also benefit children with disorders due to mutant CASK partners.
Collapse
Affiliation(s)
- Judith A Tello
- Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, 85721, USA
- Department of Neurology, University of Arizona Health Sciences, 1501 N. Campbell Ave, Tucson, AZ, 85724-5023, USA
- Present address: Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, 10010, USA
| | - Linan Jiang
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, 85721, USA
| | - Yitshak Zohar
- Department of Aerospace and Mechanical Engineering, University of Arizona, Tucson, AZ, 85721, USA
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, 85721, USA
- BIO5 Interdisciplinary Research Institute, University of Arizona, Tucson, AZ, 85721, USA
| | - Linda L Restifo
- Graduate Interdisciplinary Program in Neuroscience, University of Arizona, Tucson, AZ, 85721, USA.
- Department of Neurology, University of Arizona Health Sciences, 1501 N. Campbell Ave, Tucson, AZ, 85724-5023, USA.
- BIO5 Interdisciplinary Research Institute, University of Arizona, Tucson, AZ, 85721, USA.
- Department of Cellular & Molecular Medicine, University of Arizona Health Sciences, Tucson, AZ, 85724, USA.
| |
Collapse
|
31
|
Lima-Castañeda LÁ, Bringas ME, Aguilar-Hernandez L, Garcés-Ramírez L, Morales-Medina JC, Flores G. The antipsychotic olanzapine reduces memory deficits and neuronal abnormalities in a male rat model of Autism. J Chem Neuroanat 2023; 132:102317. [PMID: 37482145 DOI: 10.1016/j.jchemneu.2023.102317] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023]
Abstract
The prevalence of autism spectrum disorder (ASD), a neurodevelopmental condition that impacts social interaction and sensory processing, is rising. Valproic acid (VPA) exposure during pregnancy causes autistic-like traits in offspring. Olanzapine (OLZ), an atypical antipsychotic, is used to treat ASD. We assessed the impact of OLZ on behavior, neuromorphology, and nitric oxide (NO) levels in the hippocampus using prenatal VPA treatment in rats. It is commonly known that ASD patients exhibit sensory abnormalities. As such, we utilized the tail flick test to validate the ASD model. In the novel object recognition test (NORT), VPA exposure reduces the discrimination index (DI) in the first introduction to the novel object. Moreover, OLZ and vehicle-treated rats perform differently in the second exposition to the DI of the novel object, suggesting that OLZ reverses VPA-induced deficits in recognition memory. The latency to find the hidden platform in the Morris water maze test of memory and learning improves in VPA-exposed rats after OLZ administration, indicating that OLZ improves spatial memory in these rats. Administration of prenatal VPA induces neuronal hypotrophy and reduces spine density in pyramidal neurons of the CA1 region of the hippocampus. Treatment with OLZ corrects the neuromorphological changes brought on by VPA. In the CA1 region of the hippocampus, VPA treatment increases the number of neurons, which normalizes with OLZ treatment. OLZ increases the NO levels in the dorsal hippocampus in control rats. In rats exposed to VPA, the second-generation antipsychotic OLZ reduces memory-related and neuroplastic alterations. The current findings support the use of OLZ in this illness and further validate the use of prenatal VPA as a model of ASD.
Collapse
Affiliation(s)
- Luis Ángel Lima-Castañeda
- Lab. Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, 14 Sur 6301, San Manuel 72570, Puebla, Mexico; Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - María Elena Bringas
- Lab. Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, 14 Sur 6301, San Manuel 72570, Puebla, Mexico
| | - Leonardo Aguilar-Hernandez
- Lab. Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, 14 Sur 6301, San Manuel 72570, Puebla, Mexico
| | - Linda Garcés-Ramírez
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Julio César Morales-Medina
- Centro de Investigación en Reproducción Animal, CINVESTAV-Universidad Autónoma de Tlaxcala, AP 62, CP 90000 Tlaxcala, Mexico.
| | - Gonzalo Flores
- Lab. Neuropsiquiatría, Instituto de Fisiología, Benemérita Universidad Autónoma de Puebla, 14 Sur 6301, San Manuel 72570, Puebla, Mexico.
| |
Collapse
|
32
|
Velásquez MM, Lattig MC, Chitiva LC, Costa GM, Sutachan JJ, Albarracin SL. Dendritogenic Potential of the Ethanol Extract from Lippia alba Leaves in Rat Cortical Neurons. Molecules 2023; 28:6666. [PMID: 37764442 PMCID: PMC10537230 DOI: 10.3390/molecules28186666] [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: 07/13/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
A reduced dendritic complexity, especially in regions such as the hippocampus and the prefrontal cortex, has been linked to the pathophysiology of some neuropsychiatric disorders, in which synaptic plasticity and functions such as emotional and cognitive processing are compromised. For this reason, the identification of new therapeutic strategies would be enriched by the search for metabolites that promote structural plasticity. The present study evaluated the dendritogenic potential of the ethanol extract of Lippia alba, an aromatic plant rich in flavonoids and terpenes, which has been widely used in traditional medicine for its presumed analgesic, anxiolytic, and antidepressant potential. An in vitro model of rat cortical neurons was used to determine the kinetics of the plant's effect at different time intervals. Changes in morphological parameters of the neurons were determined, as well as the dendritic complexity, by Sholl analysis. The extract promotes the outgrowth of dendritic branching in a rapid and sustained fashion, without being cytotoxic to the cells. We found that this effect could be mediated by the phosphatidylinositol 3-kinase pathway, which is involved in mechanisms of neuronal plasticity, differentiation, and survival. The evidence presented in this study provides a basis for further research that, through in vivo models, can delve into the plant's therapeutic potential.
Collapse
Affiliation(s)
- María Marcela Velásquez
- Instituto de Genética Humana, Facultad de Medicina, Pontifica Universidad Javeriana, Bogotá 110911, Colombia
| | | | - Luis Carlos Chitiva
- Departamento de Química, Pontificia Universidad Javeriana, Bogotá 110911, Colombia
| | - Geison M. Costa
- Departamento de Química, Pontificia Universidad Javeriana, Bogotá 110911, Colombia
| | - Jhon Jairo Sutachan
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá 110911, Colombia
| | - Sonia Luz Albarracin
- Departamento de Nutrición y Bioquímica, Pontificia Universidad Javeriana, Bogotá 110911, Colombia
| |
Collapse
|
33
|
Kerstein PC, Agreda YS, Curran BM, Ma L, Wright KM. Gbx2 controls amacrine cell dendrite stratification through Robo1/2 receptors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.03.551861. [PMID: 37577554 PMCID: PMC10418232 DOI: 10.1101/2023.08.03.551861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Within the neuronal classes of the retina, amacrine cells (ACs) exhibit the greatest neuronal diversity in morphology and function. We show that the selective expression of the transcription factor Gbx2 is required for cell fate specification and dendritic stratification of an individual AC subtype in the mouse retina. We identify Robo1 and Robo2 as downstream effectors that when deleted, phenocopy the dendritic misprojections seen in Gbx2 mutants. Slit1 and Slit2, the ligands of Robo receptors, are localized to the OFF layers of the inner plexiform layer where we observe the dendritic misprojections in both Gbx2 and Robo1/2 mutants. We show that Robo receptors also are required for the proper dendritic stratification of additional AC subtypes, such as Vglut3+ ACs. These results show both that Gbx2 functions as a terminal selector in a single AC subtype and identify Slit-Robo signaling as a developmental mechanism for ON-OFF pathway segregation in the retina.
Collapse
|
34
|
Van Dyck A, Masin L, Bergmans S, Schevenels G, Beckers A, Vanhollebeke B, Moons L. A new microfluidic model to study dendritic remodeling and mitochondrial dynamics during axonal regeneration of adult zebrafish retinal neurons. Front Mol Neurosci 2023; 16:1196504. [PMID: 37396787 PMCID: PMC10307971 DOI: 10.3389/fnmol.2023.1196504] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 05/10/2023] [Indexed: 07/04/2023] Open
Abstract
Unlike mammals, adult zebrafish are able to fully regenerate axons and functionally recover from neuronal damage in the mature central nervous system (CNS). Decades of research have tried to identify the mechanisms behind their spontaneous regenerative capacity, but the exact underlying pathways and molecular drivers remain to be fully elucidated. By studying optic nerve injury-induced axonal regrowth of adult zebrafish retinal ganglion cells (RGCs), we previously reported transient dendritic shrinkage and changes in the distribution and morphology of mitochondria in the different neuronal compartments throughout the regenerative process. These data suggest that dendrite remodeling and temporary changes in mitochondrial dynamics contribute to effective axonal and dendritic repair upon optic nerve injury. To further elucidate these interactions, we here present a novel adult zebrafish microfluidic model in which we can demonstrate compartment-specific alterations in resource allocation in real-time at single neuron level. First, we developed a pioneering method that enables to isolate and culture adult zebrafish retinal neurons in a microfluidic setup. Notably, with this protocol, we report on a long-term adult primary neuronal culture with a high number of surviving and spontaneously outgrowing mature neurons, which was thus far only very limitedly described in literature. By performing time-lapse live cell imaging and kymographic analyses in this setup, we can explore changes in dendritic remodeling and mitochondrial motility during spontaneous axonal regeneration. This innovative model system will enable to discover how redirecting intraneuronal energy resources supports successful regeneration in the adult zebrafish CNS, and might facilitate the discovery of new therapeutic targets to promote neuronal repair in humans.
Collapse
Affiliation(s)
- Annelies Van Dyck
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Luca Masin
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Steven Bergmans
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Giel Schevenels
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies, Belgium
| | - An Beckers
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, Department of Molecular Biology, ULB Neuroscience Institute, Université libre de Bruxelles, Gosselies, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Animal Physiology and Neurobiology Division, Department of Biology, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
35
|
Kalotay E, Klugmann M, Housley GD, Fröhlich D. Recessive aminoacyl-tRNA synthetase disorders: lessons learned from in vivo disease models. Front Neurosci 2023; 17:1182874. [PMID: 37274208 PMCID: PMC10234152 DOI: 10.3389/fnins.2023.1182874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/17/2023] [Indexed: 06/06/2023] Open
Abstract
Protein synthesis is a fundamental process that underpins almost every aspect of cellular functioning. Intriguingly, despite their common function, recessive mutations in aminoacyl-tRNA synthetases (ARSs), the family of enzymes that pair tRNA molecules with amino acids prior to translation on the ribosome, cause a diverse range of multi-system disorders that affect specific groups of tissues. Neurological development is impaired in most ARS-associated disorders. In addition to central nervous system defects, diseases caused by recessive mutations in cytosolic ARSs commonly affect the liver and lungs. Patients with biallelic mutations in mitochondrial ARSs often present with encephalopathies, with variable involvement of peripheral systems. Many of these disorders cause severe disability, and as understanding of their pathogenesis is currently limited, there are no effective treatments available. To address this, accurate in vivo models for most of the recessive ARS diseases are urgently needed. Here, we discuss approaches that have been taken to model recessive ARS diseases in vivo, highlighting some of the challenges that have arisen in this process, as well as key results obtained from these models. Further development and refinement of animal models is essential to facilitate a better understanding of the pathophysiology underlying recessive ARS diseases, and ultimately to enable development and testing of effective therapies.
Collapse
Affiliation(s)
- Elizabeth Kalotay
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Matthias Klugmann
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
- Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Gary D. Housley
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Dominik Fröhlich
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
36
|
Allen AR, Jones A'V, LoBianco FV, Krager KJ, Aykin-Burns N. Effect of Sirt3 on hippocampal MnSOD activity, mitochondrial function, physiology, and cognition in an aged murine model. Behav Brain Res 2023; 444:114335. [PMID: 36804441 PMCID: PMC10081808 DOI: 10.1016/j.bbr.2023.114335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023]
Abstract
The NAD(+)-dependent deacetylase SIRT3 is a proven mitochondrial metabolic stress sensor. It has been linked to the regulation of the mitochondrial acetylome and activation of several metabolic enzymes (e.g., manganese superoxide dismutase [MnSOD]) to protect mitochondrial function and redox homeostasis, which are vital for survival, excitability, and synaptic signaling of neurons mediating short- and long-term memory formation as well as retention. Eighteen-month-old male and female wild-type (WT) and Sirt3-/- mice were behaviorally tested for hippocampus-dependent cognitive performance in a Morris water maze paradigm. Cognitive impairment was displayed during the probe trial by female and male Sirt3-/- mice but not WT mice. Upon sacrifice, brains were fixed, and morphological assessments were conducted on hippocampal tissues. Both female and male Sirt3-/- mice demonstrated impaired spatial memory retention implying that SIRT3 plays a role in long-term memory function. Golgi-staining studies revealed decreased dendritic arborization and dendritic length in the hippocampi of male Sirt3-/- compared to WT animals. Sirt3 deletion significantly increased NR1, NR2A, and NR2B expression in the hippocampus of female mice only. Enzymatic activity of MnSOD, a major mitochondrial deacetylation target of SIRT3, was significantly decreased in both female and male Sirt3-/- mice. Similarly, both female and male Sirt3-/- mice demonstrated a significant decrease in their respiratory control ratio during Complex I-driven respiration, which was apparent only in female Sirt3-/- mice during Complex II-driven respiration.
Collapse
Affiliation(s)
- Antiño R Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - A 'Vonte Jones
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Francesca V LoBianco
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Kimberly J Krager
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Nukhet Aykin-Burns
- Division of Radiation Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States.
| |
Collapse
|
37
|
Shin HS, Lee SH, Moon HJ, So YH, Lee HR, Lee EH, Jung EM. Exposure to polystyrene particles causes anxiety-, depression-like behavior and abnormal social behavior in mice. JOURNAL OF HAZARDOUS MATERIALS 2023; 454:131465. [PMID: 37130475 DOI: 10.1016/j.jhazmat.2023.131465] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/04/2023] [Accepted: 04/20/2023] [Indexed: 05/04/2023]
Abstract
In the era of plastic use, organisms are constantly exposed to polystyrene particles (PS-Ps). PS-Ps accumulated in living organisms exert negative effects on the body, although studies evaluating their effects on brain development are scarce. In this study, the effects of PS-Ps on nervous system development were investigated using cultured primary cortical neurons and mice exposed to PS-Ps at different stages of brain development. The gene expression associated with brain development was downregulated in embryonic brains following PS-Ps exposure, and Gabra2 expression decreased in the embryonic and adult mice exposed to PS-Ps. Additionally, offspring of PS-Ps-treated dams exhibited signs of anxiety- and depression-like behavior, and abnormal social behavior. We propose that PS-Ps accumulation in the brain disrupts brain development and behavior in mice. This study provides novel information regarding PS-Ps toxicity and its harmful effects on neural development and behavior in mammals.
Collapse
Affiliation(s)
- Hyun Seung Shin
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea
| | - Seung Hyun Lee
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea
| | - Ha Jung Moon
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea
| | - Yun Hee So
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea
| | - Ha Ram Lee
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea
| | - Eun-Hee Lee
- Department of Microbiology, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea
| | - Eui-Man Jung
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
38
|
Donta MS, Srivastava Y, Di Mauro CM, Paulucci-Holthauzen A, Waxham MN, McCrea PD. p120-catenin subfamily members have distinct as well as shared effects on dendrite morphology during neuron development in vitro. Front Cell Neurosci 2023; 17:1151249. [PMID: 37082208 PMCID: PMC10112520 DOI: 10.3389/fncel.2023.1151249] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/21/2023] [Indexed: 04/22/2023] Open
Abstract
Dendritic arborization is essential for proper neuronal connectivity and function. Conversely, abnormal dendrite morphology is associated with several neurological pathologies like Alzheimer's disease and schizophrenia. Among major intrinsic mechanisms that determine the extent of the dendritic arbor is cytoskeletal remodeling. Here, we characterize and compare the impact of the four proteins involved in cytoskeletal remodeling-vertebrate members of the p120-catenin subfamily-on neuronal dendrite morphology. In relation to each of their own distributions, we find that p120-catenin and delta-catenin are expressed at relatively higher proportions in growth cones compared to ARVCF-catenin and p0071-catenin; ARVCF-catenin is expressed at relatively high proportions in the nucleus; and all catenins are expressed in dendritic processes and the soma. Through altering the expression of each p120-subfamily catenin in neurons, we find that exogenous expression of either p120-catenin or delta-catenin correlates with increased dendritic length and branching, whereas their respective depletion decreases dendritic length and branching. While increasing ARVCF-catenin expression also increases dendritic length and branching, decreasing expression has no grossly observable morphological effect. Finally, increasing p0071-catenin expression increases dendritic branching, but not length, while decreasing expression decreases dendritic length and branching. These distinct localization patterns and morphological effects during neuron development suggest that these catenins have both shared and distinct roles in the context of dendrite morphogenesis.
Collapse
Affiliation(s)
- Maxsam S. Donta
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Program in Genetics and Epigenetics, Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yogesh Srivastava
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Christina M. Di Mauro
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | | | - M. Neal Waxham
- Department of Neurobiology and Anatomy, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Program in Neuroscience, Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Pierre D. McCrea
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Program in Genetics and Epigenetics, Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, United States
- Program in Neuroscience, Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
39
|
Marques DM, Almeida AS, Oliveira CBA, Machado ACL, Lara MVS, Porciúncula LO. Delayed Outgrowth in Response to the BDNF and Altered Synaptic Proteins in Neurons From SHR Rats. Neurochem Res 2023:10.1007/s11064-023-03917-9. [PMID: 36995561 DOI: 10.1007/s11064-023-03917-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/23/2023] [Accepted: 03/21/2023] [Indexed: 03/31/2023]
Abstract
Attention deficit hyperactivity disorder (ADHD) is a neurodevelopmental disorder characterized by inattention, hyperactivity, and impulsivity symptoms. Neuroimaging studies have revealed a delayed cortical and subcortical development pattern in children diagnosed with ADHD. This study followed up on the development in vitro of frontal cortical neurons from Spontaneously hypertensive rats (SHR), an ADHD rat model, and Wistar-Kyoto rats (WKY), control strain, over their time in culture, and in response to BDNF treatment at two different days in vitro (DIV). These neurons were also evaluated for synaptic proteins, brain-derived neurotrophic factor (BDNF), and related protein levels. Frontal cortical neurons from the ADHD rat model exhibited shorter dendrites and less dendritic branching over their time in culture. While pro- and mature BDNF levels were not altered, the cAMP-response element-binding (CREB) decreased at 1 DIV and SNAP-25 decreased at 5 DIV. Different from control cultures, exogenous BDNF promoted less dendritic branching in neurons from the ADHD model. Our data revealed that neurons from the ADHD model showed decreased levels of an important transcription factor at the beginning of their development, and their delayed outgrowth and maturation had consequences in the levels of SNAP-25 and may be associated with less response to BDNF. These findings provide an alternative tool for studies on synaptic dysfunctions in ADHD. They may also offer a valuable tool for investigating drug effects and new treatment opportunities.
Collapse
Affiliation(s)
- Daniela M Marques
- Departamento de Bioquímica, Programa de Pós-Graduação Em Ciências Biológicas-Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, RS, 90035-003, Brasil
| | - Amanda S Almeida
- Departamento de Bioquímica, Programa de Pós-Graduação Em Ciências Biológicas-Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, RS, 90035-003, Brasil
| | - Catiane B A Oliveira
- Departamento de Bioquímica, Programa de Pós-Graduação Em Ciências Biológicas-Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, RS, 90035-003, Brasil
| | - Ana Carolina L Machado
- Departamento de Bioquímica, Programa de Pós-Graduação Em Ciências Biológicas-Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, RS, 90035-003, Brasil
| | - Marcus Vinícius S Lara
- Departamento de Bioquímica, Programa de Pós-Graduação Em Ciências Biológicas-Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, RS, 90035-003, Brasil
| | - Lisiane O Porciúncula
- Departamento de Bioquímica, Programa de Pós-Graduação Em Ciências Biológicas-Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-anexo, Porto Alegre, RS, 90035-003, Brasil.
| |
Collapse
|
40
|
Radler MR, Liu X, Peng M, Doyle B, Toyo-Oka K, Spiliotis ET. Pyramidal neuron morphogenesis requires a septin network that stabilizes filopodia and suppresses lamellipodia during neurite initiation. Curr Biol 2023; 33:434-448.e8. [PMID: 36538929 PMCID: PMC9905282 DOI: 10.1016/j.cub.2022.11.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 10/31/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022]
Abstract
Pyramidal neurons are a major cell type of the forebrain, consisting of a pyramidally shaped soma with axonal and apicobasal dendritic processes. It is poorly understood how the neuronal soma develops its pyramidal morphology, while generating neurites of the proper shape and orientation. Here, we discovered that the spherical somata of immature neurite-less neurons possess a circumferential wreath-like network of septin filaments, which promotes neuritogenesis by balancing the protrusive activity of lamellipodia and filopodia. In embryonic rat hippocampal and mouse cortical neurons, the septin wreath network consists of curvilinear filaments that contain septins 5, 7, and 11 (Sept5/7/11). The Sept5/7/11 wreath network demarcates a zone of myosin II enrichment and Arp2/3 diminution at the base of filopodial actin bundles. In Sept7-depleted neurons, cell bodies are enlarged with hyperextended lamellae and abnormally shaped neurites that originate from lamellipodia. This phenotype is accompanied by diminished myosin II and filopodia lifetimes and increased Arp2/3 and lamellipodial activity. Inhibition of Arp2/3 rescues soma and neurite phenotypes, indicating that the septin wreath network suppresses the extension of lamellipodia, facilitating the formation of neurites from the filopodia of a consolidated soma. We show that this septin function is critical for developing a pyramidally shaped soma with properly distributed and oriented dendrites in cultured rat hippocampal neurons and in vivo in mouse perinatal cortical neurons. Therefore, the somatic septin cytoskeleton provides a key morphogenetic mechanism for neuritogenesis and the development of pyramidal neurons.
Collapse
Affiliation(s)
- Megan R Radler
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Xiaonan Liu
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Megan Peng
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Brenna Doyle
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA
| | - Kazuhito Toyo-Oka
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA 19129, USA
| | - Elias T Spiliotis
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
41
|
Quach TT, Stratton HJ, Khanna R, Mackey-Alfonso S, Deems N, Honnorat J, Meyer K, Duchemin AM. Neurodegenerative Diseases: From Dysproteostasis, Altered Calcium Signalosome to Selective Neuronal Vulnerability to AAV-Mediated Gene Therapy. Int J Mol Sci 2022; 23:ijms232214188. [PMID: 36430666 PMCID: PMC9694178 DOI: 10.3390/ijms232214188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/01/2022] [Accepted: 11/04/2022] [Indexed: 11/18/2022] Open
Abstract
Despite intense research into the multifaceted etiology of neurodegenerative diseases (ND), they remain incurable. Here we provide a brief overview of several major ND and explore novel therapeutic approaches. Although the cause (s) of ND are not fully understood, the accumulation of misfolded/aggregated proteins in the brain is a common pathological feature. This aggregation may initiate disruption of Ca++ signaling, which is an early pathological event leading to altered dendritic structure, neuronal dysfunction, and cell death. Presently, ND gene therapies remain unidimensional, elusive, and limited to modifying one pathological feature while ignoring others. Considering the complexity of signaling cascades in ND, we discuss emerging therapeutic concepts and suggest that deciphering the molecular mechanisms involved in dendritic pathology may broaden the phenotypic spectrum of ND treatment. An innovative multiplexed gene transfer strategy that employs silencing and/or over-expressing multiple effectors could preserve vulnerable neurons before they are lost. Such therapeutic approaches may extend brain health span and ameliorate burdensome chronic disease states.
Collapse
Affiliation(s)
- Tam T. Quach
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
- INSERM U1217/CNRS UMR5310, Université de Lyon, Université Claude Bernard Lyon 1, 69677 Lyon, France
| | | | - Rajesh Khanna
- Department of Molecular Pathobiology, New York University, New York, NY 10010, USA
| | - Sabrina Mackey-Alfonso
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Nicolas Deems
- Institute for Behavioral Medicine Research, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jérome Honnorat
- INSERM U1217/CNRS UMR5310, Université de Lyon, Université Claude Bernard Lyon 1, 69677 Lyon, France
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, 69677 Lyon, France
- SynatAc Team, Institut NeuroMyoGène, 69677 Lyon, France
| | - Kathrin Meyer
- The Research Institute of Nationwide Children Hospital, Columbus, OH 43205, USA
- Department of Pediatric, The Ohio State University, Columbus, OH 43210, USA
| | - Anne-Marie Duchemin
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH 43210, USA
- Correspondence: ; Tel.: +1-614-293-5517; Fax: +1-614-293-7599
| |
Collapse
|
42
|
Chau DDL, Li W, Chan WWR, Sun JKL, Zhai Y, Chow HM, Lau KF. Insulin stimulates atypical protein kinase C-mediated phosphorylation of the neuronal adaptor FE65 to potentiate neurite outgrowth by activating ARF6-Rac1 signaling. FASEB J 2022; 36:e22594. [PMID: 36250347 DOI: 10.1096/fj.202200757r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/21/2022] [Accepted: 09/26/2022] [Indexed: 11/11/2022]
Abstract
Neurite outgrowth is a fundamental process in neurons that produces extensions and, consequently, neural connectivity. Neurite damage and atrophy are observed in various brain injuries and disorders. Understanding the intrinsic pathways of neurite outgrowth is essential for developing strategies to stimulate neurite regeneration. Insulin is a pivotal hormone in the regulation of glucose homeostasis. There is increasing evidence for the neurotrophic functions of insulin, including the induction of neurite outgrowth. However, the associated mechanism remains elusive. Here, we demonstrate that insulin potentiates neurite outgrowth mediated by the small GTPases ADP-ribosylation factor 6 (ARF6) and Ras-related C3 botulinum toxin substrate 1 (Rac1) through the neuronal adaptor FE65. Moreover, insulin enhances atypical protein kinase Cι/λ (PKCι/λ) activation and FE65 phosphorylation at serine 459 (S459) in neurons and mouse brains. In vitro and cellular assays show that PKCι/λ phosphorylated FE65 at S459. Consistently, insulin potentiates FE65 S459 phosphorylation only in the presence of PKCι/λ. Phosphomimetic studies show that an FE65 S459E mutant potently activates ARF6, Rac1, and neurite outgrowth. Notably, this phosphomimetic mutation enhances the FE65-ARF6 interaction, a process that promotes ARF6-Rac1-mediated neurite outgrowth. Likewise, insulin treatment and PKCι/λ overexpression potentiate the FE65-ARF6 interaction. Conversely, PKCι/λ knockdown suppresses the stimulatory effect of FE65 on ARF6-Rac1-mediated neurite outgrowth. The effect of insulin on neurite outgrowth is also markedly attenuated in PKCι/λ knockdown neurons, in the presence and absence of FE65. Our findings reveal a novel mechanism linking insulin with ARF6-Rac1-dependent neurite extension through the PKCι/λ-mediated phosphorylation of FE65.
Collapse
Affiliation(s)
- Dennis Dik-Long Chau
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Wen Li
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China.,Research Laboratory for Biomedical Optics and Molecular Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wai Wa Ray Chan
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Jacquelyne Ka-Li Sun
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuqi Zhai
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Hei-Man Chow
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwok-Fai Lau
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
43
|
Gelon PA, Dutchak PA, Sephton CF. Synaptic dysfunction in ALS and FTD: anatomical and molecular changes provide insights into mechanisms of disease. Front Mol Neurosci 2022; 15:1000183. [PMID: 36263379 PMCID: PMC9575515 DOI: 10.3389/fnmol.2022.1000183] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022] Open
Abstract
Synaptic loss is a pathological feature of all neurodegenerative diseases including amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). ALS is a disease of the cortical and spinal motor neurons resulting in fatal paralysis due to denervation of muscles. FTD is a form of dementia that primarily affects brain regions controlling cognition, language and behavior. Once classified as two distinct diseases, ALS and FTD are now considered as part of a common disease spectrum based on overlapping clinical, pathological and genetic evidence. At the cellular level, aggregation of common proteins and overlapping gene susceptibilities are shared in both ALS and FTD. Despite the convergence of these two fields of research, the underlying disease mechanisms remain elusive. However, recent discovers from ALS and FTD patient studies and models of ALS/FTD strongly suggests that synaptic dysfunction is an early event in the disease process and a unifying hallmark of these diseases. This review provides a summary of the reported anatomical and cellular changes that occur in cortical and spinal motor neurons in ALS and FTD tissues and models of disease. We also highlight studies that identify changes in the proteome and transcriptome of ALS and FTD models and provide a conceptual overview of the processes that contribute to synaptic dysfunction in these diseases. Due to space limitations and the vast number of publications in the ALS and FTD fields, many articles have not been discussed in this review. As such, this review focuses on the three most common shared mutations in ALS and FTD, the hexanucleuotide repeat expansion within intron 1 of chromosome 9 open reading frame 72 (C9ORF72), transactive response DNA binding protein 43 (TARDBP or TDP-43) and fused in sarcoma (FUS), with the intention of highlighting common pathways that promote synaptic dysfunction in the ALS-FTD disease spectrum.
Collapse
|
44
|
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
|
45
|
A photo-switchable assay system for dendrite degeneration and repair in Drosophila melanogaster. Proc Natl Acad Sci U S A 2022; 119:e2204577119. [PMID: 35969739 PMCID: PMC9407391 DOI: 10.1073/pnas.2204577119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neurodegeneration arising from aging, injury, or diseases has devastating health consequences. Whereas neuronal survival and axon degeneration have been studied extensively, much less is known about how neurodegeneration affects dendrites, in part due to the limited assay systems available. To develop an assay for dendrite degeneration and repair, we used photo-switchable caspase-3 (caspase-Light-Oxygen-Voltage-sensing [caspase-LOV]) in peripheral class 4 dendrite arborization (c4da) neurons to induce graded neurodegeneration by adjusting illumination duration during development and adulthood in Drosophila melanogaster. We found that both developing and mature c4da neurons were able to survive while sustaining mild neurodegeneration induced by moderate caspase-LOV activation. Further, we observed active dendrite addition and dendrite regeneration in developing and mature c4da neurons, respectively. Using this assay, we found that the mouse Wallerian degeneration slow (WldS) protein can protect c4da neurons from caspase-LOV-induced dendrite degeneration and cell death. Furthermore, our data show that WldS can reduce dendrite elimination without affecting dendrite addition. In summary, we successfully established a photo-switchable assay system in both developing and mature neurons and used WldS as a test case to study the mechanisms underlying dendrite regeneration and repair.
Collapse
|
46
|
Post-Synapses in the Brain: Role of Dendritic and Spine Structures. Biomedicines 2022; 10:biomedicines10081859. [PMID: 36009405 PMCID: PMC9405724 DOI: 10.3390/biomedicines10081859] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/26/2022] [Accepted: 07/22/2022] [Indexed: 02/07/2023] Open
Abstract
Brain synapses are neuronal structures of the greatest interest. For a long time, however, the knowledge about them was variable, and interest was mostly focused on their pre-synaptic portions, especially neurotransmitter release from axon terminals. In the present review interest is focused on post-synapses, the structures receiving and converting pre-synaptic messages. Upon further modulation, such messages are transferred to dendritic fibers. Dendrites are profoundly different from axons; they are shorter and of variable thickness. Their post-synapses are of two types. Those called flat/intended/aspines, integrated into dendritic fibers, are very frequent in inhibitory neurons. The spines, small and stemming protrusions, connected to dendritic fibers by their necks, are present in almost all excitatory neurons. Several structures and functions including the post-synaptic densities and associated proteins, the nanoscale mechanisms of compartmentalization, the cytoskeletons of actin and microtubules, are analogous in the two post-synaptic forms. However other properties, such as plasticity and its functions of learning and memory, are largely distinct. Several properties of spines, including emersion from dendritic fibers, growth, change in shape and decreases in size up to disappearance, are specific. Spinal heads correspond to largely independent signaling compartments. They are motile, their local signaling is fast, however transport through their thin necks is slow. When single spines are activated separately, their dendritic effects are often lacking; when multiple spines are activated concomitantly, their effects take place. Defects of post-synaptic responses, especially those of spines, take place in various brain diseases. Here alterations affecting symptoms and future therapy are shown to occur in neurodegenerative diseases and autism spectrum disorders.
Collapse
|
47
|
The role of ciliopathy-associated type 3 adenylyl cyclase in infanticidal behavior in virgin adult male mice. iScience 2022; 25:104534. [PMID: 35754726 PMCID: PMC9218507 DOI: 10.1016/j.isci.2022.104534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/11/2022] [Accepted: 06/01/2022] [Indexed: 12/04/2022] Open
Abstract
Virgin adult male mice often display killing of alien newborns, defined as infanticide, and this behavior is dependent on olfactory signaling. Olfactory perception is achieved by the main olfactory system (MOS) or vomeronasal system (VNS). Although it has been established that the VNS is crucial for infanticide in male mice, the role of the MOS in infanticide remains unknown. Herein, by producing lesions via ZnSO4 perfusion and N-methyl-D-aspartic acid stereotactic injection, we demonstrated that the main olfactory epithelium (MOE), anterior olfactory nucleus (AON), or ventromedial hypothalamus (VMH) is crucial for infanticide in adult males. By using CRISPR-Cas9 coupled with adeno-associated viruses to induce specific knockdown of type 3 adenylyl cyclase (AC3) in these tissues, we further demonstrated that AC3, a ciliopathy-associated protein, in the MOE and the expression of related proteins in the AON or VMH are necessary for infanticidal behavior in virgin adult male mice. MOE lesions and knockdown of AC3 in the MOE result in abnormal infanticidal behavior The infanticidal behavior of male mice is impaired by lesioning of the AON or VMH AC3 knockdown in the AON or VMH affects the infanticidal behavior of male mice
Collapse
|
48
|
Shree S, Sutradhar S, Trottier O, Tu Y, Liang X, Howard J. Dynamic instability of dendrite tips generates the highly branched morphologies of sensory neurons. SCIENCE ADVANCES 2022; 8:eabn0080. [PMID: 35767611 PMCID: PMC9242452 DOI: 10.1126/sciadv.abn0080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 05/12/2022] [Indexed: 06/15/2023]
Abstract
The highly ramified arbors of neuronal dendrites provide the substrate for the high connectivity and computational power of the brain. Altered dendritic morphology is associated with neuronal diseases. Many molecules have been shown to play crucial roles in shaping and maintaining dendrite morphology. However, the underlying principles by which molecular interactions generate branched morphologies are not understood. To elucidate these principles, we visualized the growth of dendrites throughout larval development of Drosophila sensory neurons and found that the tips of dendrites undergo dynamic instability, transitioning rapidly and stochastically between growing, shrinking, and paused states. By incorporating these measured dynamics into an agent-based computational model, we showed that the complex and highly variable dendritic morphologies of these cells are a consequence of the stochastic dynamics of their dendrite tips. These principles may generalize to branching of other neuronal cell types, as well as to branching at the subcellular and tissue levels.
Collapse
Affiliation(s)
- Sonal Shree
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Sabyasachi Sutradhar
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
| | - Olivier Trottier
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Department of Physics, Yale University, New Haven, CT 06511, USA
| | - Yuhai Tu
- IBM T.J. Watson Research Center, Yorktown Heights, NY 10598, USA
| | - Xin Liang
- Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, 100084 Beijing, China
| | - Jonathon Howard
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Department of Physics, Yale University, New Haven, CT 06511, USA
- Quantitative Biology Institute, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
49
|
Rowland C, Harland B, Smith JH, Moslehi S, Dalrymple-Alford J, Taylor RP. Investigating Fractal Analysis as a Diagnostic Tool That Probes the Connectivity of Hippocampal Neurons. Front Physiol 2022; 13:932598. [PMID: 35812343 PMCID: PMC9260144 DOI: 10.3389/fphys.2022.932598] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 05/31/2022] [Indexed: 11/30/2022] Open
Abstract
Many of nature’s fractal objects benefit from the favorable functionality that results from their pattern repetition at multiple scales. Our recent research focused on the importance of fractal scaling in establishing connectivity between neurons. Fractal dimension DA of the neuron arbors was shown to relate to the optimization of competing functional constraints—the ability of dendrites to connect to other neurons versus the costs associated with building the dendrites. Here, we consider whether pathological states of neurons might affect this fractal optimization and if changes in DA might therefore be used as a diagnostic tool in parallel with traditional measures like Sholl analyses. We use confocal microscopy to obtain images of CA1 pyramidal neurons in the coronal plane of the dorsal rat hippocampus and construct 3-dimensional models of the dendritic arbors using Neurolucida software. We examine six rodent groups which vary in brain condition (whether they had lesions in the anterior thalamic nuclei, ATN) and experience (their housing environment and experience in a spatial task). Previously, we showed ATN lesions reduced spine density in hippocampal CA1 neurons, whereas enriched housing increased spine density in both ATN lesion and sham rats. Here, we investigate whether ATN lesions and experience also effect the complexity and connectivity of CA1 dendritic arbors. We show that sham rats exposed to enriched housing and spatial memory training exhibited higher complexity (as measured by DA) and connectivity compared to other groups. When we categorize the rodent groups into those with or without lesions, we find that both categories achieve an optimal balance of connectivity with respect to material cost. However, the DA value used to achieve this optimization does not change between these two categories, suggesting any morphological differences induced by the lesions are too small to influence the optimization process. Accordingly, we highlight considerations associated with applying our technique to publicly accessible repositories of neuron images with a broader range of pathological conditions.
Collapse
Affiliation(s)
- Conor Rowland
- Physics Department, University of Oregon, Eugene, OR, United States
| | - Bruce Harland
- School of Pharmacy, University of Auckland, Auckland, New Zealand
| | - Julian H. Smith
- Physics Department, University of Oregon, Eugene, OR, United States
| | - Saba Moslehi
- Physics Department, University of Oregon, Eugene, OR, United States
| | - John Dalrymple-Alford
- School of Psychology, Speech and Hearing, University of Canterbury, Christchurch, New Zealand
- New Zealand Brain Research Institute, Christchurch, New Zealand
| | - Richard P. Taylor
- Physics Department, University of Oregon, Eugene, OR, United States
- *Correspondence: Richard P. Taylor,
| |
Collapse
|
50
|
Li H, Gavis ER. The Drosophila fragile X mental retardation protein modulates the neuronal cytoskeleton to limit dendritic arborization. Development 2022; 149:275257. [DOI: 10.1242/dev.200379] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 04/21/2022] [Indexed: 01/02/2023]
Abstract
ABSTRACT
Dendritic arbor development is a complex, highly regulated process. Post-transcriptional regulation mediated by RNA-binding proteins plays an important role in neuronal dendrite morphogenesis by delivering on-site, on-demand protein synthesis. Here, we show how the Drosophila fragile X mental retardation protein (FMRP), a conserved RNA-binding protein, limits dendrite branching to ensure proper neuronal function during larval sensory neuron development. FMRP knockdown causes increased dendritic terminal branch growth and a resulting overelaboration defect due, in part, to altered microtubule stability and dynamics. FMRP also controls dendrite outgrowth by regulating the Drosophila profilin homolog chickadee (chic). FMRP colocalizes with chic mRNA in dendritic granules and regulates its dendritic localization and protein expression. Whereas RNA-binding domains KH1 and KH2 are both crucial for FMRP-mediated dendritic regulation, KH2 specifically is required for FMRP granule formation and chic mRNA association, suggesting a link between dendritic FMRP granules and FMRP function in dendrite elaboration. Our studies implicate FMRP-mediated modulation of both the neuronal microtubule and actin cytoskeletons in multidendritic neuronal architecture, and provide molecular insight into FMRP granule formation and its relevance to FMRP function in dendritic patterning.
Collapse
Affiliation(s)
- Hui Li
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Elizabeth R. Gavis
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|