1
|
Zuo Y, Formoli N, Libster A, Sun D, Turner A, Iemolo A, Telese F. Single-Nucleus Transcriptomics Identifies Neuroblast Migration Programs Sensitive to Reelin and Cannabis in the Adolescent Nucleus Accumbens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.03.646846. [PMID: 40236084 PMCID: PMC11996521 DOI: 10.1101/2025.04.03.646846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The interplay between cannabis exposure during adolescence and genetic predisposition has been linked to increased vulnerability to psychiatric disorders. To investigate the molecular underpinnings of this interaction, we performed single-nucleus RNA sequencing of the nucleus accumbens (NAc) in a mouse model of Reln haploinsufficiency, a genetic risk factor for psychiatric disorders, following adolescent exposure to tetrahydrocannabinol (THC), the primary psychoactive component of cannabis. We identified a gene co-expression network influenced by both Reln genotype and THC, enriched in genes associated with human psychiatric disorders and predominantly expressed in a GABAergic neuroblast subpopulation. We showed that neuroblasts actively migrated in the adolescent NAc, but declined with age. Cell-to-cell communication analysis further revealed that these neuroblasts receive migratory cues from cholecystokinin interneurons, which express high levels of cannabinoid receptors. Together, these findings provide mechanistic insights into how adolescent THC exposure and genetic risk factors may impair GABAergic circuit maturation.
Collapse
|
2
|
Sajukumar K, Yadav P, Lee GH. Dab1 expression level controls Reelin-induced PI3K-Akt activation in early GABAergic neurons. Biochem Biophys Res Commun 2025; 751:151444. [PMID: 39919390 DOI: 10.1016/j.bbrc.2025.151444] [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: 11/17/2024] [Revised: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 02/09/2025]
Abstract
Disabled-1 (Dab1) is a key regulator of the Reelin signaling cascades and controls many neurodevelopmental processes, including pyramidal neuron migration, dendrite growth, and spine formation. Dab1 is phosphorylated upon the binding of Reelin to Very low density lipoprotein receptor (VLDLR) and Apolipoprotein E receptor 2 (ApoER2) receptors, resulting in activation of a series of downstream signaling pathways, including Phosphoinositide 3-kinase (PI3K)/Akt, Lissencephaly 1 (Lis1), Crks/C3G, and Extracellular signal-regulated kinase 1/2 (Erk1/2). Dab1 is then rapidly degraded via the proteasome pathway. In humans, REELIN and DAB1 are genetically associated with several psychiatric disorders, such as schizophrenia and autism spectrum disorder. Although a subset of GABAergic neurons express Reelin and are continuously exposed to Reelin from early developmental stages through adulthood, most studies have only investigated the role of Reelin in the development and function of pyramidal neurons; as such the role of Reelin in GABAergic neurons remains poorly understood. In this study, we isolated primary neurons from mouse medial ganglionic eminence (MGE) at embryonic day 14.5 that 98-99 % were composed of GABAergic neurons. Using MGE-isolated GABAergic neurons, we studied the quantitative differences in Reelin signaling and expression of related genes in these neurons for the first time. Reelin supplementation did not activate PI3K-Akt signaling in most GABAergic neurons, but it did activate the signaling pathway in Somatostatin-positive GABAergic neurons. Dab1 was transcriptionally repressed in early GABAergic neurons, demonstrating the selective activation of Reelin signaling between subsets of neurons. This study provides quantitative evidence and contributes insights into the molecular mechanisms underlying the limited effects of Reelin on Dab1-related developmental activities in the majority of GABAergic neurons during brain development.
Collapse
Affiliation(s)
- Kavitha Sajukumar
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Prabhakar Yadav
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea
| | - Gum Hwa Lee
- College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea; Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chosun University, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
3
|
Silva-Hurtado TJ, Giua G, Lassalle O, Makrini-Maleville L, Strauss B, Wager-Miller J, Freyermuth JM, Mackie K, Valjent E, Manzoni OJ, Chavis P. Reelin Deficiency and Synaptic Impairment in the Adolescent Prefrontal Cortex Following Initial Synthetic Cannabinoid Exposure. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100426. [PMID: 39926699 PMCID: PMC11804564 DOI: 10.1016/j.bpsgos.2024.100426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 02/11/2025] Open
Abstract
Background Adolescent cannabinoid exposure can have long-lasting effects on the brain, particularly in the prefrontal cortex, where the reelin protein plays a crucial role in neural organization. Chronic cannabinoid exposure leads to reelin deficiency and behavioral abnormalities, but the underlying mechanisms remain unclear. With the increasing use of synthetic cannabinoids (SCs) among young people, understanding these effects is crucial. Methods We examined the cellular and synaptic consequences of initial SC exposure in adolescent male mice 1 day after a single in vivo exposure to WIN 55,212-2. Our approach combined immunohistochemistry, Western blots, conditional CB1 receptor (CB1R) knockout mouse lines, quantitative polymerase chain reaction, and ex vivo electrophysiology to investigate the effects of SC on reelin expression and synaptic plasticity. Additionally, single-molecule fluorescent in situ hybridization profiling was used to identify cellular coexpression patterns of reelin and CB1Rs. Results Our findings indicate that a single exposure to SC decreased reelin expression in specific prefrontal cortex layers accompanied by disrupted proteolytic fragmentation but not changes in messenger RNA expression. Single-molecule fluorescent in situ hybridization profiling revealed a strong coexpression of CB1R and reelin. Furthermore, our pharmacological and genetic approaches demonstrated that CB1Rs in GABAergic (gamma-aminobutyric acidergic) neurons mediate the SC-induced decrease in reelin. This decrease in reelin results in a reduction in long-term potentiation, phenocopying reelin haploinsufficient mice. Notably, we restored long-term potentiation by infusing reelin bilaterally, establishing a functional link between reelin depletion and synaptic deficits. Conclusions These findings provide new insights into the neural consequences of adolescent cannabinoid consumption and highlight the critical role of reelin in the cellular mechanisms associated with SC initiation during adolescence.
Collapse
Affiliation(s)
- Thenzing J. Silva-Hurtado
- Aix-Marseille University, INSERM, INMED, Marseille, France
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
| | - Gabriele Giua
- Aix-Marseille University, INSERM, INMED, Marseille, France
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
| | - Olivier Lassalle
- Aix-Marseille University, INSERM, INMED, Marseille, France
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
| | - Leila Makrini-Maleville
- Institute of Functional Genomics, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Benjamin Strauss
- Aix-Marseille University, INSERM, INMED, Marseille, France
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
| | - Jim Wager-Miller
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
- The Gill Institute for Neuroscience and Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana
| | | | - Ken Mackie
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
- The Gill Institute for Neuroscience and Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana
| | - Emmanuel Valjent
- Institute of Functional Genomics, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Olivier J.J. Manzoni
- Aix-Marseille University, INSERM, INMED, Marseille, France
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
| | - Pascale Chavis
- Aix-Marseille University, INSERM, INMED, Marseille, France
- Cannalab Cannabinoids Neuroscience Research International Associated Laboratory, INSERM, Aix-Marseille University, Marseille, France and Indiana University, Bloomington, Indiana
| |
Collapse
|
4
|
Tavitian A, Lax E, Song W, Szyf M, Schipper HM. Hippocampal reelin and GAD67 gene expression and methylation in the GFAP.HMOX1 mouse model of schizophrenia. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119899. [PMID: 39798610 DOI: 10.1016/j.bbamcr.2025.119899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 12/17/2024] [Accepted: 12/31/2024] [Indexed: 01/15/2025]
Abstract
Schizophrenia is a complex neuropsychiatric disorder featuring enhanced brain oxidative stress and deficient reelin protein. GFAP.HMOX10-12m mice that overexpress heme oxygenase-1 (HO-1) in astrocytes manifest a schizophrenia-like neurochemical, neuropathological and behavioral phenotype including brain oxidative stress and reelin downregulation. We used RT-PCR, targeted bisulfite next-generation sequencing, immunohistochemistry and in situ hybridization on hippocampal tissue of GFAP.HMOX10-12m mice to delineate a possible molecular mechanism for the downregulation of reelin and to identify the neuronal and non-neuronal (glial) cell types expressing reelin in our model. We found reduced reelin and increased DNMT1 and TET1 mRNA expression in the hippocampus of male GFAP.HMOX10-12m mice and reduced GAD67 mRNA expression in females. These mRNA changes were accompanied by sexually dimorphic alterations in DNA methylation levels of Reln and Gad1 genes. Reelin protein was expressed by oligodendrocytes and GABAergic interneurons, but not by astrocytes or microglia in GFAP.HMOX10-12m and wild-type brains of both sexes. Reelin mRNA was also observed in oligodendrocytes. Moreover, a significant downregulation of reelin-expressing oligodendrocytes was detected in the hippocampal dentate gyrus of male GFAP.HMOX10-12m mice. These results suggest a novel mechanism for brain reelin depletion in schizophrenia. Containment of the astrocytic HO-1 cascade by pharmacological or other means may protect against stress-induced brain reelin depletion in schizophrenia and other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Ayda Tavitian
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Elad Lax
- Department of Molecular Biology, Ariel University, Ariel, Israel; Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada
| | - Wei Song
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Moshe Szyf
- Department of Pharmacology & Therapeutics, McGill University, Montreal, Quebec, Canada.
| | - Hyman M Schipper
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada.
| |
Collapse
|
5
|
Gupta T, Kaur M, Gupta M, Singla N, Kharbanda PS, Bansal YS, Radotra BD, Gupta SK. Analysis of distribution and localization of proteins of the reelin signalling pathway in mesial temporal lobe epilepsy. Int J Neurosci 2025; 135:188-202. [PMID: 38060511 DOI: 10.1080/00207454.2023.2292957] [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: 06/07/2022] [Revised: 11/15/2023] [Accepted: 12/02/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION Granule cell dispersion (GCD) is pathognomonic of hippocampal sclerosis seen in the mesial temporal lobe epilepsy (MTLE). Current animal studies indicate deficiency of Reelin is associated with abnormal granule cell migration leading to GCD. The present study aimed to evaluate complete Reelin signalling pathway to assess whether Reelin deficiency is related to MTLE. MATERIALS AND METHODS Hippocampal sclerosis was confirmed by H and E stain. To explore the amount and cellular location of the Reelin cascade molecules, the hippocampal tissues from MTLE surgery and controls (n = 15 each) were studied using Immuno-histochemistry (IHC). Additionally, confocal imaging was used to validate the IHC findings by co-localization of different proteins. Quantification of IHC images was performed using histo-score and confocal images by Image J software. RESULTS Immune expression of active Reelin was significantly reduced in patients. Reelin receptors were deranged, apolipoprotein E receptor 2 was increased while very low-density lipoprotein receptor was reduced. Disabled-1, a downstream molecule was significantly reduced in MTLE. Its ultimate target, cofilin was thus disinhibited and expressed more in MTLE. Reelin cleaving protease, matrix metalloprotease-9 (MMP-9) and MMP-9 inhibitor, tissue inhibitor of matrix protease-1, showed reduced expression in extracellular matrix. Semi-quantification of immunohistochemistry was done using Histo (H) score. H score of Reelin in diseased patients was 15 against 125 for control patients. These results were validated by confocal fluorescence microscopy. CONCLUSIONS Reelin signalling cascade was deranged in chronic MTLE. Pharmacological manipulation of Reelin cascade can be done at various levels and it may provide novel treatment options for MTLE.
Collapse
Affiliation(s)
- Tulika Gupta
- Department of Anatomy, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Mandeep Kaur
- Department of Anatomy, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Mili Gupta
- Department of Biochemistry, Dr. Harvansh Singh Judge Institute of Dental Sciences and Hospital, Panjab University, Chandigarh, India
| | - Navneet Singla
- Department of Neurosurgery, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Parampreet S Kharbanda
- Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Yogender S Bansal
- Department of Forensic Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - B D Radotra
- Department of Histopathology Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - S K Gupta
- Department of Neurosurgery, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
6
|
Hattori M. Regulatory mechanism of Reelin activity: a platform for exploiting Reelin as a therapeutic agent. Front Mol Neurosci 2025; 18:1546083. [PMID: 39931643 PMCID: PMC11808024 DOI: 10.3389/fnmol.2025.1546083] [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: 12/16/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
Reelin is a secreted glycoprotein that was initially investigated in the field of neuronal development. However, in recent decades, its role in the adult brain has become increasingly important, and it is now clear that diminished Reelin function is involved in the pathogenesis and progression of neuropsychiatric and neurodegenerative disorders, including schizophrenia and Alzheimer's disease (AD). Reelin activity is regulated at multiple steps, including synthesis, posttranslational modification, secretion, oligomerization, proteolytic processing, and interactions with extracellular molecules. Moreover, the differential use of two canonical receptors and the presence of non-canonical receptors and co-receptors add to the functional diversity of Reelin. In this review, I summarize recent findings on the molecular mechanisms of Reelin activity. I also discuss possible strategies to enhance Reelin's function. A complete understanding of Reelin function and its regulatory mechanisms in the adult central nervous system could help ameliorate neuropsychiatric and neurodegenerative disorders.
Collapse
Affiliation(s)
- Mitsuharu Hattori
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| |
Collapse
|
7
|
Kobro-Flatmoen A, Omholt SW. Intraneuronal binding of amyloid beta with reelin-Implications for the onset of Alzheimer's disease. PLoS Comput Biol 2025; 21:e1012709. [PMID: 39775030 PMCID: PMC11741591 DOI: 10.1371/journal.pcbi.1012709] [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: 06/18/2024] [Revised: 01/17/2025] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Numerous studies of the human brain supported by experimental results from rodent and cell models point to a central role for intracellular amyloid beta (Aβ) in the onset of Alzheimer's disease (AD). In a rat model used to study AD, it was recently shown that in layer II neurons of the anteriolateral entorhinal cortex expressing high levels of the glycoprotein reelin (Re+alECLII neurons), reelin and Aβ engage in a direct protein-protein interaction. If reelin functions as a sink for intracellular Aβ and if the binding to reelin makes Aβ physiologically inert, it implies that reelin can prevent the neuron from being exposed to the harmful effects typically associated with increased levels of oligomeric Aβ. Considering that reelin expression is extraordinarily high in Re+alECLII neurons compared to most other cortical neurons, such a protective role appears to be very difficult to reconcile with the fact that this subset of ECLII neurons is clearly a major cradle for the onset of AD. Here, we show that this conundrum can be resolved if Re+alECLII neurons have a higher maximum production capacity of Aβ than neurons expressing low levels of reelin, and we provide a rationale for why this difference has evolved.
Collapse
Affiliation(s)
- Asgeir Kobro-Flatmoen
- Kavli Institute for Systems Neuroscience, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- K. G. Jebsen Centre for Alzheimer’s Disease, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Stig W. Omholt
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
8
|
Hu R, Boshans LL, Zhu B, Cai P, Tao Y, Youssef M, Girrbach GI, Song Y, Wang X, Tsankov A, Buxbaum JD, Ma S, Yang N. Expanding GABAergic Neuronal Diversity in iPSC-Derived Disease Models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626438. [PMID: 39677822 PMCID: PMC11642846 DOI: 10.1101/2024.12.03.626438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
GABAergic interneurons play a critical role in maintaining neural circuit function, and their dysfunction is implicated in various neurodevelopmental and psychiatric disorders. Traditional approaches for differentiating human pluripotent stem cells (PSCs) into neuronal cells often face challenges such as incomplete neural differentiation, prolonged culture periods, and variability across PSC lines. To address these limitations, we developed a new strategy that integrates overexpression of transcription factors ASCL1 and DLX2 with dual-SMAD and WNT inhibition, efficiently driving the differentiation of human PSCs into diverse, region-specific GABAergic neuronal types. Using single-cell sequencing, we characterized the cellular heterogeneity of GABAergic induced neurons (iNs) generated with the patterning factors (patterned iNs) and those derived solely with transcription factors (PSC-derived iNs), uncovering the regulatory mechanisms that govern their fate specification. Patterned iNs exhibited gene expression features corresponding to multiple brain regions, particularly ganglionic eminence (GE) and neocortex, while GABAergic PSC-derived iNs predominantly resembled hypothalamic and thalamic neurons. Both iN types were enriched for genes relevant to neurodevelopmental and psychiatric disorders, with patterned iNs more specifically linked to neural lineage genes, highlighting their utility for disease modeling. We further applied this protocol to investigate the impact of an ADNP syndrome-associated mutation (p.Tyr719* variant) on GABAergic neuron differentiation, revealing that this mutation disrupts GABAergic fate specification and synaptic transmission. Overall, this study expands the toolkit for disease modeling by demonstrating the complementary advantages of GABAergic PSC-derived iNs and patterned iNs in representing distinct GABAergic neuron subtypes, brain regions, and disease contexts. These approaches offer a powerful platform for elucidating the molecular mechanisms underlying various neurodevelopmental and psychiatric disorders.
Collapse
Affiliation(s)
- Ruiqi Hu
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alper Center for Neurodevelopment and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- These authors contributed equally
| | - Linda L Boshans
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alper Center for Neurodevelopment and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- These authors contributed equally
| | - Bohan Zhu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Peiwen Cai
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yiran Tao
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alper Center for Neurodevelopment and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mark Youssef
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alper Center for Neurodevelopment and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gizem Inak Girrbach
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alper Center for Neurodevelopment and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yingnan Song
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xuran Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexander Tsankov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joseph D Buxbaum
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sai Ma
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nan Yang
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alper Center for Neurodevelopment and Regeneration, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Lead contact
| |
Collapse
|
9
|
Katsuyama Y, Hattori M. REELIN ameliorates Alzheimer's disease, but how? Neurosci Res 2024; 208:8-14. [PMID: 39094979 DOI: 10.1016/j.neures.2024.07.004] [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: 07/19/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
Alzheimer's disease (AD) is the most prevalent type of dementia; therefore, there is a high demand for therapeutic medication targeting it. In this context, extensive research has been conducted to identify molecular targets for drugs. AD manifests through two primary pathological signs: senile plaques and neurofibrillary tangles, caused by accumulations of amyloid-beta (Aβ) and phosphorylated tau, respectively. Thus, studies concerning the molecular mechanisms underlying AD etiology have primarily focused on Aβ generation and tau phosphorylation, with the anticipation of uncovering a signaling pathway impacting these molecular processes. Over the past two decades, studies using not only experimental model systems but also examining human brains have accumulated fragmentary evidences suggesting that REELIN signaling pathway is deeply involved in AD. Here, we explore REELIN signaling pathway and its involvement in memory function within the brain and review studies investigating molecular connections between REELIN signaling pathway and AD etiology. This review aims to understand how the manipulation (activation) of this pathway might ameliorate the disease's etiology.
Collapse
Affiliation(s)
- Yu Katsuyama
- Division of Neuroanatomy, Department of Anatomy, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan.
| | - Mitsuharu Hattori
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi 467-8603, Japan
| |
Collapse
|
10
|
Zou X, Tang Q, Ojiro R, Ozawa S, Shobudani M, Sakamaki Y, Ebizuka Y, Jin M, Yoshida T, Shibutani M. Increased spontaneous activity and progressive suppression of adult neurogenesis in the hippocampus of rat offspring after maternal exposure to imidacloprid. Chem Biol Interact 2024; 399:111145. [PMID: 39002876 DOI: 10.1016/j.cbi.2024.111145] [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: 05/13/2024] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Imidacloprid (IMI) is a widely used neonicotinoid insecticide that poses risks for developmental neurotoxicity in mammals. The present study investigated the effects of maternal exposure to IMI on behaviors and adult neurogenesis in the hippocampal dentate gyrus (DG) of rat offspring. Dams were exposed to IMI via diet (83, 250, or 750 ppm in diet) from gestational day 6 until day 21 post-delivery on weaning, and offspring were maintained until adulthood on postnatal day 77. In the neurogenic niche, 750-ppm IMI decreased numbers of late-stage neural progenitor cells (NPCs) and post-mitotic immature granule cells by suppressing NPC proliferation and ERK1/2-FOS-mediated synaptic plasticity of granule cells on weaning. Suppressed reelin signaling might be responsible for the observed reductions of neurogenesis and synaptic plasticity. In adulthood, IMI at ≥ 250 ppm decreased neural stem cells by suppressing their proliferation and increasing apoptosis, and mature granule cells were reduced due to suppressed NPC differentiation. Behavioral tests revealed increased spontaneous activity in adulthood at 750 ppm. IMI decreased hippocampal acetylcholinesterase activity and Chrnb2 transcript levels in the DG on weaning and in adulthood. IMI increased numbers of astrocytes and M1-type microglia in the DG hilus, and upregulated neuroinflammation and oxidative stress-related genes on weaning. In adulthood, IMI increased malondialdehyde level and number of M1-type microglia, and downregulated neuroinflammation and oxidative stress-related genes. These results suggest that IMI persistently affected cholinergic signaling, induced neuroinflammation and oxidative stress during exposure, and increased sensitivity to oxidative stress after exposure in the hippocampus, causing hyperactivity and progressive suppression of neurogenesis in adulthood. The no-observed-adverse-effect level of IMI for offspring behaviors and hippocampal neurogenesis was determined to be 83 ppm (5.5-14.1 mg/kg body weight/day).
Collapse
Affiliation(s)
- Xinyu Zou
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Qian Tang
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Ryota Ojiro
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Shunsuke Ozawa
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Momoka Shobudani
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Yuri Sakamaki
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Yuri Ebizuka
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Meilan Jin
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Southwest University, No. 2 Tiansheng Road, BeiBei District, Chongqing, 400715, PR China.
| | - Toshinori Yoshida
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Division of Animal Life Science, Institute of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan; Cooperative Division of Veterinary Sciences, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo, 183-8509, Japan.
| |
Collapse
|
11
|
Hamad MIK, Daoud S, Petrova P, Rabaya O, Jbara A, Al Houqani S, BaniYas S, Alblooshi M, Almheiri A, Nakhal MM, Ali BR, Shehab S, Allouh MZ, Emerald BS, Schneider-Lódi M, Bataineh MF, Herz J, Förster E. Reelin differentially shapes dendrite morphology of medial entorhinal cortical ocean and island cells. Development 2024; 151:dev202449. [PMID: 38856043 PMCID: PMC11234379 DOI: 10.1242/dev.202449] [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: 10/18/2023] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
The function of medial entorhinal cortex layer II (MECII) excitatory neurons has been recently explored. MECII dysfunction underlies deficits in spatial navigation and working memory. MECII neurons comprise two major excitatory neuronal populations, pyramidal island and stellate ocean cells, in addition to the inhibitory interneurons. Ocean cells express reelin and surround clusters of island cells that lack reelin expression. The influence of reelin expression by ocean cells and interneurons on their own morphological differentiation and that of MECII island cells has remained unknown. To address this, we used a conditional reelin knockout (RelncKO) mouse to induce reelin deficiency postnatally in vitro and in vivo. Reelin deficiency caused dendritic hypertrophy of ocean cells, interneurons and only proximal dendritic compartments of island cells. Ca2+ recording showed that both cell types exhibited an elevation of calcium frequencies in RelncKO, indicating that the hypertrophic effect is related to excessive Ca2+ signalling. Moreover, pharmacological receptor blockade in RelncKO mouse revealed malfunctioning of GABAB, NMDA and AMPA receptors. Collectively, this study emphasizes the significance of reelin in neuronal growth, and its absence results in dendrite hypertrophy of MECII neurons.
Collapse
Affiliation(s)
- Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates
| | - Solieman Daoud
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum 44801, Germany
| | - Petya Petrova
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum 44801, Germany
| | - Obada Rabaya
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum 44801, Germany
| | - Abdalrahim Jbara
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum 44801, Germany
| | - Shaikha Al Houqani
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates
| | - Shamsa BaniYas
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates
| | - Meera Alblooshi
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates
| | - Ayesha Almheiri
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates
| | - Mohammed M. Nakhal
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates
| | - Bassam R. Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates
| | - Safa Shehab
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates
| | - Mohammed Z. Allouh
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates
| | - Mária Schneider-Lódi
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum 44801, Germany
| | - Mo'ath F. Bataineh
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates
| | - Joachim Herz
- Departments of Molecular Genetics, Neuroscience, Neurology and Neurotherapeutics; Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum 44801, Germany
| |
Collapse
|
12
|
Mota B, Brás AR, Araújo-Andrade L, Silva A, Pereira PA, Madeira MD, Cardoso A. High-Caloric Diets in Adolescence Impair Specific GABAergic Subpopulations, Neurogenesis, and Alter Astrocyte Morphology. Int J Mol Sci 2024; 25:5524. [PMID: 38791562 PMCID: PMC11122083 DOI: 10.3390/ijms25105524] [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: 04/10/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
We compared the effects of two different high-caloric diets administered to 4-week-old rats for 12 weeks: a diet rich in sugar (30% sucrose) and a cafeteria diet rich in sugar and high-fat foods. We focused on the hippocampus, particularly on the gamma-aminobutyric acid (GABA)ergic system, including the Ca2+-binding proteins parvalbumin (PV), calretinin (CR), calbindin (CB), and the neuropeptides somatostatin (SST) and neuropeptide Y (NPY). We also analyzed the density of cholinergic varicosities, brain-derived neurotrophic factor (BDNF), reelin (RELN), and cyclin-dependent kinase-5 (CDK-5) mRNA levels, and glial fibrillary acidic protein (GFAP) expression. The cafeteria diet reduced PV-positive neurons in the granular layer, hilus, and CA1, as well as NPY-positive neurons in the hilus, without altering other GABAergic populations or overall GABA levels. The high-sugar diet induced a decrease in the number of PV-positive cells in CA3 and an increase in CB-positive cells in the hilus and CA1. No alterations were observed in the cholinergic varicosities. The cafeteria diet also reduced the relative mRNA expression of RELN without significant changes in BDNF and CDK5 levels. The cafeteria diet increased the number but reduced the length of the astrocyte processes. These data highlight the significance of determining the mechanisms mediating the observed effects of these diets and imply that the cognitive impairments previously found might be related to both the neuroinflammation process and the reduction in PV, NPY, and RELN expression in the hippocampal formation.
Collapse
Affiliation(s)
- Bárbara Mota
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ana Rita Brás
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
| | - Leonardo Araújo-Andrade
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Ana Silva
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Pedro A. Pereira
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - M. Dulce Madeira
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Armando Cardoso
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; (B.M.)
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
13
|
Hamad MIK, Rabaya O, Jbara A, Daoud S, Petrova P, Ali BR, Allouh MZ, Herz J, Förster E. Reelin Regulates Developmental Desynchronization Transition of Neocortical Network Activity. Biomolecules 2024; 14:593. [PMID: 38786001 PMCID: PMC11118507 DOI: 10.3390/biom14050593] [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: 03/12/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/25/2024] Open
Abstract
During the first and second stages of postnatal development, neocortical neurons exhibit a wide range of spontaneous synchronous activity (SSA). Towards the end of the second postnatal week, the SSA is replaced by a more sparse and desynchronized firing pattern. The developmental desynchronization of neocortical spontaneous neuronal activity is thought to be intrinsically generated, since sensory deprivation from the periphery does not affect the time course of this transition. The extracellular protein reelin controls various aspects of neuronal development through multimodular signaling. However, so far it is unclear whether reelin contributes to the developmental desynchronization transition of neocortical neurons. The present study aims to investigate the role of reelin in postnatal cortical developmental desynchronization using a conditional reelin knockout (RelncKO) mouse model. Conditional reelin deficiency was induced during early postnatal development, and Ca2+ recordings were conducted from organotypic cultures (OTCs) of the somatosensory cortex. Our results show that both wild type (wt) and RelncKO exhibited an SSA pattern during the early postnatal week. However, at the end of the second postnatal week, wt OTCs underwent a transition to a desynchronized network activity pattern, while RelncKO activity remained synchronous. This changing activity pattern suggests that reelin is involved in regulating the developmental desynchronization of cortical neuronal network activity. Moreover, the developmental desynchronization impairment observed in RelncKO was rescued when RelncKO OTCs were co-cultured with wt OTCs. Finally, we show that the developmental transition to a desynchronized state at the end of the second postnatal week is not dependent on glutamatergic signaling. Instead, the transition is dependent on GABAAR and GABABR signaling. The results suggest that reelin controls developmental desynchronization through GABAAR and GABABR signaling.
Collapse
Affiliation(s)
- Mohammad I. K. Hamad
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates;
| | - Obada Rabaya
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany; (O.R.); (S.D.); (P.P.); (E.F.)
| | - Abdalrahim Jbara
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany; (O.R.); (S.D.); (P.P.); (E.F.)
| | - Solieman Daoud
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany; (O.R.); (S.D.); (P.P.); (E.F.)
| | - Petya Petrova
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany; (O.R.); (S.D.); (P.P.); (E.F.)
| | - Bassam R. Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates;
| | - Mohammed Z. Allouh
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain 17666, United Arab Emirates;
| | - Joachim Herz
- Departments of Molecular Genetics, Neuroscience, Neurology and Neurotherapeutics, Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 5323, USA
| | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, 44801 Bochum, Germany; (O.R.); (S.D.); (P.P.); (E.F.)
| |
Collapse
|
14
|
Calvier L, Alexander A, Marckx AT, Kounnas MZ, Durakoglugil M, Herz J. Safety of Anti-Reelin Therapeutic Approaches for Chronic Inflammatory Diseases. Cells 2024; 13:583. [PMID: 38607022 PMCID: PMC11011630 DOI: 10.3390/cells13070583] [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: 02/23/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024] Open
Abstract
Reelin, a large extracellular glycoprotein, plays critical roles in neuronal development and synaptic plasticity in the central nervous system (CNS). Recent studies have revealed non-neuronal functions of plasma Reelin in inflammation by promoting endothelial-leukocyte adhesion through its canonical pathway in endothelial cells (via ApoER2 acting on NF-κB), as well as in vascular tone regulation and thrombosis. In this study, we have investigated the safety and efficacy of selectively depleting plasma Reelin as a potential therapeutic strategy for chronic inflammatory diseases. We found that Reelin expression remains stable throughout adulthood and that peripheral anti-Reelin antibody treatment with CR-50 efficiently depletes plasma Reelin without affecting its levels or functionality within the CNS. Notably, this approach preserves essential neuronal functions and synaptic plasticity. Furthermore, in mice induced with experimental autoimmune encephalomyelitis (EAE), selective modulation of endothelial responses by anti-Reelin antibodies reduces pathological leukocyte infiltration without completely abolishing diapedesis. Finally, long-term Reelin depletion under metabolic stress induced by a Western diet did not negatively impact the heart, kidney, or liver, suggesting a favorable safety profile. These findings underscore the promising role of peripheral anti-Reelin therapeutic strategies for autoimmune diseases and conditions where endothelial function is compromised, offering a novel approach that may avoid the immunosuppressive side effects associated with conventional anti-inflammatory therapies.
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anna Alexander
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Austin T. Marckx
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Murat Durakoglugil
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
15
|
Caracci MO, Pizarro H, Alarcón-Godoy C, Fuentealba LM, Farfán P, De Pace R, Santibañez N, Cavieres VA, Pástor TP, Bonifacino JS, Mardones GA, Marzolo MP. The Reelin receptor ApoER2 is a cargo for the adaptor protein complex AP-4: Implications for Hereditary Spastic Paraplegia. Prog Neurobiol 2024; 234:102575. [PMID: 38281682 PMCID: PMC10979513 DOI: 10.1016/j.pneurobio.2024.102575] [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: 07/03/2023] [Revised: 01/11/2024] [Accepted: 01/23/2024] [Indexed: 01/30/2024]
Abstract
Adaptor protein complex 4 (AP-4) is a heterotetrameric complex that promotes export of selected cargo proteins from the trans-Golgi network. Mutations in each of the AP-4 subunits cause a complicated form of Hereditary Spastic Paraplegia (HSP). Herein, we report that ApoER2, a receptor in the Reelin signaling pathway, is a cargo of the AP-4 complex. We identify the motif ISSF/Y within the ApoER2 cytosolic domain as necessary for interaction with the canonical signal-binding pocket of the µ4 (AP4M1) subunit of AP-4. AP4E1- knock-out (KO) HeLa cells and hippocampal neurons from Ap4e1-KO mice display increased co-localization of ApoER2 with Golgi markers. Furthermore, hippocampal neurons from Ap4e1-KO mice and AP4M1-KO human iPSC-derived cortical i3Neurons exhibit reduced ApoER2 protein expression. Analyses of biosynthetic transport of ApoER2 reveal differential post-Golgi trafficking of the receptor, with lower axonal distribution in KO compared to wild-type neurons, indicating a role of AP-4 and the ISSF/Y motif in the axonal localization of ApoER2. Finally, analyses of Reelin signaling in mouse hippocampal and human cortical KO neurons show that AP4 deficiency causes no changes in Reelin-dependent activation of the AKT pathway and only mild changes in Reelin-induced dendritic arborization, but reduces Reelin-induced ERK phosphorylation, CREB activation, and Golgi deployment. This work thus establishes ApoER2 as a novel cargo of the AP-4 complex, suggesting that defects in the trafficking of this receptor and in the Reelin signaling pathway could contribute to the pathogenesis of HSP caused by mutations in AP-4 subunits.
Collapse
Affiliation(s)
- Mario O Caracci
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Héctor Pizarro
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Carlos Alarcón-Godoy
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Luz M Fuentealba
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Pamela Farfán
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile
| | - Raffaella De Pace
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Natacha Santibañez
- Instituto de Patología Animal, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Viviana A Cavieres
- Departamento de Ciencias Biológicas y Químicas, Fac. Med y Ciencia, USS, Santiago, Chile
| | - Tammy P Pástor
- Escuela de Medicina, Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile
| | - Juan S Bonifacino
- Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Gonzalo A Mardones
- Escuela de Medicina, Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile
| | - María-Paz Marzolo
- Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, P. Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
16
|
Clasadonte J, Deprez T, Stephens GS, Mairet-Coello G, Cortin PY, Boutier M, Frey A, Chin J, Rajman M. ΔFosB is part of a homeostatic mechanism that protects the epileptic brain from further deterioration. Front Mol Neurosci 2024; 16:1324922. [PMID: 38283700 PMCID: PMC10810990 DOI: 10.3389/fnmol.2023.1324922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/13/2023] [Indexed: 01/30/2024] Open
Abstract
Activity induced transcription factor ΔFosB plays a key role in different CNS disorders including epilepsy, Alzheimer's disease, and addiction. Recent findings suggest that ΔFosB drives cognitive deficits in epilepsy and together with the emergence of small molecule inhibitors of ΔFosB activity makes it an interesting therapeutic target. However, whether ΔFosB contributes to pathophysiology or provides protection in drug-resistant epilepsy is still unclear. In this study, ΔFosB was specifically downregulated by delivering AAV-shRNA into the hippocampus of chronically epileptic mice using the drug-resistant pilocarpine model of mesial temporal epilepsy (mTLE). Immunohistochemistry analyses showed that prolonged downregulation of ΔFosB led to exacerbation of neuroinflammatory markers of astrogliosis and microgliosis, loss of mossy fibers, and hippocampal granule cell dispersion. Furthermore, prolonged inhibition of ΔFosB using a ΔJunD construct to block ΔFosB signaling in a mouse model of Alzheimer's disease, that exhibits spontaneous recurrent seizures, led to similar findings, with increased neuroinflammation and decreased NPY expression in mossy fibers. Together, these data suggest that seizure-induced ΔFosB, regardless of seizure-etiology, is part of a homeostatic mechanism that protects the epileptic brain from further deterioration.
Collapse
Affiliation(s)
- Jerome Clasadonte
- Epilepsy Discovery Research, UCB Biopharma SRL, Braine-l’Alleud, Belgium
| | - Tania Deprez
- Epilepsy Discovery Research, UCB Biopharma SRL, Braine-l’Alleud, Belgium
| | | | | | - Pierre-Yves Cortin
- Epilepsy Discovery Research, UCB Biopharma SRL, Braine-l’Alleud, Belgium
| | - Maxime Boutier
- Epilepsy Discovery Research, UCB Biopharma SRL, Braine-l’Alleud, Belgium
| | - Aurore Frey
- Epilepsy Discovery Research, UCB Biopharma SRL, Braine-l’Alleud, Belgium
| | - Jeannie Chin
- Baylor College of Medicine, Houston, TX, United States
| | - Marek Rajman
- Epilepsy Discovery Research, UCB Biopharma SRL, Braine-l’Alleud, Belgium
| |
Collapse
|
17
|
Halvorson CS, Sánchez-Lafuente CL, Johnston JN, Kalynchuk LE, Caruncho HJ. Molecular Mechanisms of Reelin in the Enteric Nervous System and the Microbiota-Gut-Brain Axis: Implications for Depression and Antidepressant Therapy. Int J Mol Sci 2024; 25:814. [PMID: 38255890 PMCID: PMC10815176 DOI: 10.3390/ijms25020814] [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: 11/29/2023] [Revised: 12/30/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Current pharmacological treatments for depression fail to produce adequate remission in a significant proportion of patients. Increasingly, other systems, such as the microbiome-gut-brain axis, are being looked at as putative novel avenues for depression treatment. Dysbiosis and dysregulation along this axis are highly comorbid with the severity of depression symptoms. The endogenous extracellular matrix protein reelin is present in all intestinal layers as well as in myenteric and submucosal ganglia, and its receptors are also present in the gut. Reelin secretion from subepithelial myofibroblasts regulates cellular migration along the crypt-villus axis in the small intestine and colon. Reelin brain expression is downregulated in mood and psychotic disorders, and reelin injections have fast antidepressant-like effects in animal models of depression. This review seeks to discuss the roles of reelin in the gastrointestinal system and propose a putative role for reelin actions in the microbiota-gut-brain axis in the pathogenesis and treatment of depression, primarily reflecting on alterations in gut epithelial cell renewal and in the clustering of serotonin transporters.
Collapse
Affiliation(s)
- Ciara S. Halvorson
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Rd., Victoria, BC V8P 5C2, Canada; (C.S.H.); (C.L.S.-L.); (L.E.K.)
| | - Carla Liria Sánchez-Lafuente
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Rd., Victoria, BC V8P 5C2, Canada; (C.S.H.); (C.L.S.-L.); (L.E.K.)
| | - Jenessa N. Johnston
- Section on the Neurobiology and Treatment of Mood Disorders, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Lisa E. Kalynchuk
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Rd., Victoria, BC V8P 5C2, Canada; (C.S.H.); (C.L.S.-L.); (L.E.K.)
| | - Hector J. Caruncho
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Rd., Victoria, BC V8P 5C2, Canada; (C.S.H.); (C.L.S.-L.); (L.E.K.)
| |
Collapse
|
18
|
Caracci MO, Pizarro H, Alarcón-Godoy C, Fuentealba LM, Farfán P, Pace RD, Santibañez N, Cavieres VA, Pástor TP, Bonifacino JS, Mardones GA, Marzolo MP. The Reelin Receptor ApoER2 is a Cargo for the Adaptor Protein Complex AP-4: Implications for Hereditary Spastic Paraplegia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.21.572896. [PMID: 38187774 PMCID: PMC10769347 DOI: 10.1101/2023.12.21.572896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Adaptor protein complex 4 (AP-4) is a heterotetrameric complex that promotes protein export from the trans -Golgi network. Mutations in each of the AP-4 subunits cause a complicated form of Hereditary Spastic Paraplegia (HSP). Herein, we report that ApoER2, a receptor in the Reelin signaling pathway, is a cargo of the AP-4 complex. We identify the motif ISSF/Y within the ApoER2 cytosolic domain as necessary for interaction with the canonical signal-binding pocket of the µ4 (AP4M1) subunit of AP-4. AP4E1 -knock-out (KO) HeLa cells and hippocampal neurons from Ap4e1 -KO mice display increased Golgi localization of ApoER2. Furthermore, hippocampal neurons from Ap4e1 -KO mice and AP4M1 -KO human iPSC-derived cortical i3Neurons exhibit reduced ApoER2 protein expression. Analyses of biosynthetic transport of ApoER2 reveal differential post-Golgi trafficking of the receptor, with lower axonal distribution in KO compared to wild-type neurons, indicating a role of AP-4 and the ISSF/Y motif in the axonal localization of ApoER2. Finally, analyses of Reelin signaling in mouse hippocampal and human cortical KO neurons show that AP4 deficiency causes no changes in Reelin-dependent activation of the AKT pathway and only mild changes in Reelin-induced dendritic arborization, but reduces Reelin-induced ERK phosphorylation, CREB activation, and Golgi deployment. Altogether, this work establishes ApoER2 as a novel cargo of the AP-4 complex, suggesting that defects in the trafficking of this receptor and in the Reelin signaling pathway could contribute to the pathogenesis of HSP caused by mutations in AP-4 subunits.
Collapse
|
19
|
Joly-Amado A, Kulkarni N, Nash KR. Reelin Signaling in Neurodevelopmental Disorders and Neurodegenerative Diseases. Brain Sci 2023; 13:1479. [PMID: 37891846 PMCID: PMC10605156 DOI: 10.3390/brainsci13101479] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Reelin is an extracellular matrix glycoprotein involved in neuronal migration during embryonic brain development and synaptic plasticity in the adult brain. The role of Reelin in the developing central nervous system has been extensively characterized. Indeed, a loss of Reelin or a disruption in its signaling cascade leads to neurodevelopmental defects and is associated with ataxia, intellectual disability, autism, and several psychiatric disorders. In the adult brain, Reelin is critically involved in neurogenesis and synaptic plasticity. Reelin's signaling potentiates glutamatergic and GABAergic neurotransmission, induces synaptic maturation, and increases AMPA and NMDA receptor subunits' expression and activity. As a result, there is a growing literature reporting that a loss of function and/or reduction of Reelin is implicated in numerous neurodegenerative diseases. The present review summarizes the current state of the literature regarding the implication of Reelin and Reelin-mediated signaling during aging and neurodegenerative disorders, highlighting Reelin as a possible target in the prevention or treatment of progressive neurodegeneration.
Collapse
Affiliation(s)
- Aurelie Joly-Amado
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA; (N.K.); (K.R.N.)
| | | | | |
Collapse
|
20
|
Alexander A, Herz J, Calvier L. Reelin through the years: From brain development to inflammation. Cell Rep 2023; 42:112669. [PMID: 37339050 PMCID: PMC10592530 DOI: 10.1016/j.celrep.2023.112669] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/09/2023] [Accepted: 06/02/2023] [Indexed: 06/22/2023] Open
Abstract
Reelin was originally identified as a regulator of neuronal migration and synaptic function, but its non-neuronal functions have received far less attention. Reelin participates in organ development and physiological functions in various tissues, but it is also dysregulated in some diseases. In the cardiovascular system, Reelin is abundant in the blood, where it contributes to platelet adhesion and coagulation, as well as vascular adhesion and permeability of leukocytes. It is a pro-inflammatory and pro-thrombotic factor with important implications for autoinflammatory and autoimmune diseases such as multiple sclerosis, Alzheimer's disease, arthritis, atherosclerosis, or cancer. Mechanistically, Reelin is a large secreted glycoprotein that binds to several membrane receptors, including ApoER2, VLDLR, integrins, and ephrins. Reelin signaling depends on the cell type but mostly involves phosphorylation of NF-κB, PI3K, AKT, or JAK/STAT. This review focuses on non-neuronal functions and the therapeutic potential of Reelin, while highlighting secretion, signaling, and functional similarities between cell types.
Collapse
Affiliation(s)
- Anna Alexander
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA; Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA; Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Laurent Calvier
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
21
|
Jaggar M, Ghosh S, Janakiraman B, Chatterjee A, Maheshwari M, Dewan V, Hare B, Deb S, Figueiredo D, Duman RS, Vaidya VA. Influence of Chronic Electroconvulsive Seizures on Plasticity-Associated Gene Expression and Perineuronal Nets Within the Hippocampi of Young Adult and Middle-Aged Sprague-Dawley Rats. Int J Neuropsychopharmacol 2023; 26:294-306. [PMID: 36879414 PMCID: PMC10109107 DOI: 10.1093/ijnp/pyad008] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
BACKGROUND Electroconvulsive seizure therapy is often used in both treatment-resistant and geriatric depression. However, preclinical studies identifying targets of chronic electroconvulsive seizure (ECS) are predominantly focused on animal models in young adulthood. Given that putative transcriptional, neurogenic, and neuroplastic mechanisms implicated in the behavioral effects of chronic ECS themselves exhibit age-dependent modulation, it remains unknown whether the molecular and cellular targets of chronic ECS vary with age. METHODS We subjected young adult (2-3 months) and middle-aged (12-13 months), male Sprague Dawley rats to sham or chronic ECS and assessed for despair-like behavior, hippocampal gene expression, hippocampal neurogenesis, and neuroplastic changes in the extracellular matrix, reelin, and perineuronal net numbers. RESULTS Chronic ECS reduced despair-like behavior at both ages, accompanied by overlapping and unique changes in activity-dependent and trophic factor gene expression. Although chronic ECS had a similar impact on quiescent neural progenitor numbers at both ages, the eventual increase in hippocampal progenitor proliferation was substantially higher in young adulthood. We noted a decline in reelin⁺ cell numbers following chronic ECS only in young adulthood. In contrast, an age-invariant, robust dissolution of perineuronal net numbers that encapsulate parvalbumin⁺ neurons in the hippocampus were observed following chronic ECS. CONCLUSION Our findings indicate that age is a key variable in determining the nature of chronic ECS-evoked molecular and cellular changes in the hippocampus. This raises the intriguing possibility that chronic ECS may recruit distinct, as well as overlapping, mechanisms to drive antidepressant-like behavioral changes in an age-dependent manner.
Collapse
Affiliation(s)
- Minal Jaggar
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Shreya Ghosh
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Balaganesh Janakiraman
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Ashmita Chatterjee
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Megha Maheshwari
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Vani Dewan
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Brendan Hare
- Division of Molecular Psychiatry, Department of Psychiatry and Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sukrita Deb
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Dwight Figueiredo
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Ronald S Duman
- Division of Molecular Psychiatry, Department of Psychiatry and Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Vidita A Vaidya
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| |
Collapse
|
22
|
Ishii K, Kohno T, Sakai K, Hattori M. Reelin regulates the migration of late-born hippocampal CA1 neurons via cofilin phosphorylation. Mol Cell Neurosci 2023; 124:103794. [PMID: 36435394 DOI: 10.1016/j.mcn.2022.103794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 10/04/2022] [Accepted: 11/10/2022] [Indexed: 11/27/2022] Open
Abstract
Reelin, a large secreted glycoprotein, plays an important role in neuronal migration during brain development. The C-terminal region (CTR) of Reelin is involved in the efficient activation of downstream signaling and its loss leads to abnormal hippocampal layer formation. However, the molecular mechanism by which Reelin CTR regulates hippocampal development remains unknown. Here, we showed that the migration of late-born, but not early-born, neurons is impaired in the knock-in mice in which Reelin CTR is deleted (ΔC-KI mice). The phosphorylation of cofilin, an actin-depolymerizing protein, was remarkably decreased in the hippocampus of the ΔC-KI mice. Exogenous expression of pseudo-phosphorylated cofilin rescued the ectopic positioning of neurons in the hippocampus of ΔC-KI mice. These results suggest that Reelin CTR is required for the migration of late-born neurons in the hippocampus and that this event involves appropriate phosphorylation of cofilin.
Collapse
Affiliation(s)
- Keisuke Ishii
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Takao Kohno
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan.
| | - Kaori Sakai
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan
| | - Mitsuharu Hattori
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603, Japan.
| |
Collapse
|
23
|
Johnson KO, Harel L, Triplett JW. Postsynaptic NMDA Receptor Expression Is Required for Visual Corticocollicular Projection Refinement in the Mouse Superior Colliculus. J Neurosci 2023; 43:1310-1320. [PMID: 36717228 PMCID: PMC9987568 DOI: 10.1523/jneurosci.1473-22.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/09/2022] [Accepted: 11/13/2022] [Indexed: 01/31/2023] Open
Abstract
Efficient sensory processing of spatial information is facilitated through the organization of neuronal connections into topographic maps of space. In integrative sensory centers, converging topographic maps must be aligned to merge spatially congruent information. The superior colliculus (SC) receives topographically ordered visual inputs from retinal ganglion cells (RGCs) in the eye and layer 5 neurons in the primary visual cortex (L5-V1). Previous studies suggest that RGCs instruct the alignment of later-arriving L5-V1 inputs in an activity-dependent manner. However, the molecular mechanisms underlying this remain unclear. Here, we explored the role of NMDA receptors in visual map alignment in the SC using a conditional genetic knockout approach. We leveraged a novel knock-in mouse line that expresses tamoxifen-inducible Cre recombinase under the control of the Tal1 gene (Tal1CreERT2 ), which we show allows for specific recombination in the superficial layers of the SC. We used Tal1CreERT2 mice of either sex to conditionally delete the obligate GluN1 subunit of the NMDA receptor (SC-cKO) during the period of visual map alignment. We observed a significant disruption of L5-V1 axon terminal organization in the SC of SC-cKO mice. Importantly, retinocollicular topography was unaffected in this context, suggesting that alignment is also disrupted. Time-course experiments suggest that NMDA receptors may play a critical role in the refinement of L5-V1 inputs in the SC. Together, these data implicate NMDA receptors as critical mediators of activity-dependent visual map alignment in the SC.SIGNIFICANCE STATEMENT Alignment of topographic inputs is critical for integration of spatially congruent sensory information; however, little is known about the mechanisms underlying this complex process. Here, we took a conditional genetic approach to explore the role of NMDA receptors in the alignment of retinal and cortical visual inputs in the superior colliculus. We characterize a novel mouse line providing spatial and temporal control of recombination in the superior colliculus and reveal a critical role for NMDA expression in visual map alignment. These data support a role for neuronal activity in visual map alignment and provide mechanistic insight into this complex developmental process.
Collapse
Affiliation(s)
- Kristy O Johnson
- Center for Neuroscience Research, Children's National Research Institute, Washington, DC 20010
- Institute for Biomedical Sciences, George Washington University School of Medicine, Washington, DC 20037
| | - Leeor Harel
- Center for Neuroscience Research, Children's National Research Institute, Washington, DC 20010
| | - Jason W Triplett
- Center for Neuroscience Research, Children's National Research Institute, Washington, DC 20010
- Institute for Biomedical Sciences, George Washington University School of Medicine, Washington, DC 20037
- Department of Pediatrics, George Washington University School of Medicine, Washington, DC 20037
- Department of Pharmacology and Physiology, George Washington University School of Medicine, Washington, DC 20037
| |
Collapse
|
24
|
Extracellular matrix and synapse formation. Biosci Rep 2023; 43:232259. [PMID: 36503961 PMCID: PMC9829651 DOI: 10.1042/bsr20212411] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 11/08/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
The extracellular matrix (ECM) is a complex molecular network distributed throughout the extracellular space of different tissues as well as the neuronal system. Previous studies have identified various ECM components that play important roles in neuronal maturation and signal transduction. ECM components are reported to be involved in neurogenesis, neuronal migration, and axonal growth by interacting or binding to specific receptors. In addition, the ECM is found to regulate synapse formation, the stability of the synaptic structure, and synaptic plasticity. Here, we mainly reviewed the effects of various ECM components on synapse formation and briefly described the related diseases caused by the abnormality of several ECM components.
Collapse
|
25
|
Abo Aoun M, Meek BP, Clair L, Wikstrom S, Prasad B, Modirrousta M. Prognostic factors in major depressive disorder: comparing responders and non-responders to Repetitive Transcranial Magnetic Stimulation (rTMS), a naturalistic retrospective chart review. Psychiatry Clin Neurosci 2023; 77:38-47. [PMID: 36207801 DOI: 10.1111/pcn.13488] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/18/2022] [Accepted: 10/04/2022] [Indexed: 01/06/2023]
Abstract
AIM Repetitive transcranial magnetic stimulation (rTMS) is widely utilized as an effective treatment for major depressive disorder (MDD) with varying response rates. Factors associated with better treatment outcome remain scarce. This naturalistic retrospective chart review hopes to shed light on easily obtainable and measurable predictive factors for patients referred to rTMS. METHODS Protocol parameters, medication, rated scales, rTMS protocols, and treatment outcomes were reviewed for 196 patients with MDD who received rTMS at Saint Boniface Hospital between 2013 and 2019. Logistic regression and marginal effects were used to assess the different predictor variables for response (50% reduction or more on the Hamilton Depression Rating Scale (Ham-D)) and remission (Ham-D of ≤7 by the last session). RESULTS HamD at 10 sessions was predictive of remission, and Sheehan Disability Scale (SDS) at 10 sessions was predictive of response to rTMS. Ham-D, SDS, and Beck Anxiety Inventory were predictive of remission and response by Beck Anxiety Inventory 20 sessions. High frequency rTMS had a similar response and remission rate to low frequency, but higher response rate to intermittent Theta Burst Stimulation with no difference in remission rate. Positive predictive factors of response were lower age and bupropion use. Negative predictive factors were antipsychotics, anticonvulsants, or benzodiazepine use. For remission, antipsychotics or anticonvulsants use were negative predictors; bupropion use and higher resting motor threshold were positive predictors. Severity of depression as measured by baseline HamD was not associated with different probabilities of treatment success.
Collapse
Affiliation(s)
| | - Benjamin P Meek
- Department of Clinical Health Psychology, University of Manitoba, Winnipeg, Canada
| | - Luc Clair
- Department of Economics, University of Winnipeg, Winnipeg, Canada.,Canadian Centre for Agri-Food Research in Health and Medicine, Saint Boniface Research Hospital, Winnipeg, Canada
| | - Sara Wikstrom
- Saint Boniface Hospital, Psychiatry, Winnipeg, Canada
| | | | - Mandana Modirrousta
- BrainWave Clinic, Winnipeg, Canada.,Department of Psychiatry, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
26
|
Sánchez-Lafuente CL, Romay-Tallon R, Allen J, Johnston JN, Kalynchuk LE, Caruncho HJ. Sex differences in basal reelin levels in the paraventricular hypothalamus and in response to chronic stress induced by repeated corticosterone in rats. Horm Behav 2022; 146:105267. [PMID: 36274499 DOI: 10.1016/j.yhbeh.2022.105267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/21/2022] [Accepted: 09/24/2022] [Indexed: 11/04/2022]
Abstract
Repeated exposure to the stress hormone corticosterone results in depressive-like behaviours paralleled by the downregulation of hippocampal reelin expression. Reelin is expressed in key neural populations involved in the stress response, but whether its hypothalamic expression is sex-specific or involved in sex-specific vulnerability to stress is unknown. Female and male rats were treated with either daily vehicle or corticosterone injections (40 mg/kg) for 21 days. Thereafter, they were subjected to several behavioural tasks before being sacrificed to allow the analysis of reelin expression in hypothalamic nuclei. The basal density of reelin-positive cells in males was significantly higher in the paraventricular nucleus (19 %) and in the medial preoptic area (51 %) compared to females. Chronic corticosterone injections increased the immobility time in the forced swim test in males (107 %) and females (108 %) and decreased the exploration of the elevated plus maze in males (34 %). Corticosterone also caused a significant decrease in the density of reelin-positive cells in males, in both ventrodorsal (37 %) and ventrolateral (32 %) subdivisions of the paraventricular nucleus, while not affecting females. Moreover, in the paraventricular nucleus of males, 30 % of the basal reelin-positive cells co-expressed oxytocin while only 17.5 % did in females, showing a positive correlation between reelin and oxytocin levels. Chronic corticosterone did not significantly affect co-localization levels. For the first time, this study shows that there is a sexually dimorphic subpopulation of reelin-positive neurons in the paraventricular nucleus that can be differentially affected by chronic stress.
Collapse
Affiliation(s)
| | | | - Josh Allen
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Jenessa N Johnston
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Lisa E Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
27
|
Jin K, Zhang S, Jiang C, Liu R, Chen B, Zhao H, Zhang Q, Shen Z, Xu P, Hu X, Jiao J, Lu J, Huang M. The role of reelin in the pathological mechanism of depression from clinical to rodents. Psychiatry Res 2022; 317:114838. [PMID: 36103758 DOI: 10.1016/j.psychres.2022.114838] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/13/2022] [Accepted: 09/04/2022] [Indexed: 01/04/2023]
Abstract
Major depressive disorder (MDD) is a devastating mental illness and the leading cause of disability worldwide. Previous studies have suggested that synaptic plasticity in the hippocampus plays an important role in depression pathogenesis. Reelin is expressed mainly in the frontal lobe and hippocampus, and is closely associated with neurodevelopment and synaptic plasticity. However, few studies have investigated its role in MDD combining clinical trials and animal experiments. We show that in a clinical trial, plasma reelin levels decreased in patients with first-episode drug-naïve MDD and increased after treatment; further, plasma reelin levels allowed to distinguish drug-naïve patients with first-episode MDD from healthy individuals. In rats, chronic mild and unpredictable stress led to a decrease in both reelin mRNA and protein levels in the hippocampus, which could be reversed by vortioxetine. Subsequent experiments confirmed that the reelin-ApoER2-NR2A /NR2B pathway regulates hippocampal synaptic plasticity and may be involved in depression or antidepressant responses. Our work contributes to a deeper understanding of MDD pathogenesis and provides new evidence that reelin should be considered a potential therapeutic target for MDD.
Collapse
Affiliation(s)
- Kangyu Jin
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Shiyi Zhang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Chaonan Jiang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Ripeng Liu
- College of First Clinical College, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China
| | - Bing Chen
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Haoyang Zhao
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Qin Zhang
- College of First Clinical College, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou 310053, China
| | - Zhe Shen
- Department of Child Psychology, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Pengfeng Xu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Xiaohan Hu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Jianping Jiao
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China
| | - Jing Lu
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China.
| | - Manli Huang
- Department of Psychiatry, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; The Key Laboratory of Mental Disorder Management in Zhejiang Province, Hangzhou 310003, China.
| |
Collapse
|
28
|
Arshad MN, Oppenheimer S, Jeong J, Buyukdemirtas B, Naegele JR. Hippocampal transplants of fetal GABAergic progenitors regulate adult neurogenesis in mice with temporal lobe epilepsy. Neurobiol Dis 2022; 174:105879. [PMID: 36183946 DOI: 10.1016/j.nbd.2022.105879] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 09/21/2022] [Accepted: 09/28/2022] [Indexed: 11/20/2022] Open
Abstract
GABAergic interneurons play a role in regulating adult neurogenesis within the dentate gyrus (DG) of the hippocampus. Neurogenesis occurs within a stem cell niche in the subgranular zone (SGZ) of the DG. In this niche, populations of neural progenitors give rise to granule cells that migrate radially into the granule cell layer of the DG. Altered neurogenesis in temporal lobe epilepsy (TLE) is linked to a transient increase in the proliferation of new neurons and the abnormal inversion of Type 1 progenitors, resulting in ectopic migration of Type 3 progenitors into the hilus of the DG. These ectopic cells mature into granule cells in the hilus that become hyperexcitable and contribute to the development of spontaneous recurrent seizures. To test whether grafts of GABAergic cells in the DG restore synaptic inhibition, prior work focused on transplanting GABAergic progenitors into the hilus of the DG. This cell-based therapeutic approach was shown to alter the disease phenotype by ameliorating spontaneous seizures in mice with pilocarpine-induced TLE. Prior optogenetic and immunohistochemical studies demonstrated that the transplanted GABAergic interneurons increased levels of synaptic inhibition by establishing inhibitory synaptic contacts with adult-born granule cells, consistent with the observed suppression of seizures. Whether GABAergic progenitor transplantation into the DG ameliorates underlying abnormalities in adult neurogenesis caused by TLE is not known. As a first step to address this question, we compared the effects of GABAergic progenitor transplantation on Type 1, Type 2, and Type 3 progenitors in the stem cell niche using cell type-specific molecular markers in naïve, non-epileptic mice. The progenitor transplantation increased GABAergic interneurons in the DG and led to a significant reduction in Type 2 progenitors and a concomitant increase in Type 3 progenitors. Next, we compared the effects of GABAergic interneuron transplantation in epileptic mice. Transplantation of GABAergic progenitors resulted in reductions in inverted Type 1, Type 2, and hilar ectopic Type 3 cells, concomitant with an increase in the radial migration of Type 3 progenitors into the GCL (Granule Cell Layer). Thus, in mice with Pilocarpine induced TLE, hilar transplants of GABA interneurons may reverse abnormal patterns of adult neurogenesis, an outcome that may ameliorate seizures.
Collapse
Affiliation(s)
- Muhammad N Arshad
- Hall-Atwater Laboratory, Wesleyan University, Department of Biology, Program in Neuroscience and Behavior, Middletown, CT 06459-0170, USA.
| | - Simon Oppenheimer
- Hall-Atwater Laboratory, Wesleyan University, Department of Biology, Program in Neuroscience and Behavior, Middletown, CT 06459-0170, USA.
| | - Jaye Jeong
- Hall-Atwater Laboratory, Wesleyan University, Department of Biology, Program in Neuroscience and Behavior, Middletown, CT 06459-0170, USA.
| | - Bilge Buyukdemirtas
- Hall-Atwater Laboratory, Wesleyan University, Department of Biology, Program in Neuroscience and Behavior, Middletown, CT 06459-0170, USA.
| | - Janice R Naegele
- Hall-Atwater Laboratory, Wesleyan University, Department of Biology, Program in Neuroscience and Behavior, Middletown, CT 06459-0170, USA.
| |
Collapse
|
29
|
de Guglielmo G, Iemolo A, Nur A, Turner A, Montilla-Perez P, Martinez A, Crook C, Roberts A, Telese F. Reelin deficiency exacerbates cocaine-induced hyperlocomotion by enhancing neuronal activity in the dorsomedial striatum. GENES, BRAIN, AND BEHAVIOR 2022; 21:e12828. [PMID: 35906757 PMCID: PMC9744517 DOI: 10.1111/gbb.12828] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022]
Abstract
The Reln gene encodes for the extracellular glycoprotein Reelin, which regulates several brain functions from development to adulthood, including neuronal migration, dendritic growth and branching and synapse formation and plasticity. Human studies have implicated Reelin signaling in several neurodevelopmental and psychiatric disorders. Mouse studies using the heterozygous Reeler (HR) mice have shown that reduced levels of Reln expression are associated with deficits in learning and memory and increased disinhibition. Although these traits are relevant to substance use disorders, the role of Reelin in cellular and behavioral responses to addictive drugs remains largely unknown. Here, we compared HR mice to wild-type (WT) littermate controls to investigate whether Reelin signaling contributes to the hyperlocomotor and rewarding effects of cocaine. After a single or repeated cocaine injections, HR mice showed enhanced cocaine-induced locomotor activity compared with WT controls. This effect persisted after withdrawal. In contrast, Reelin deficiency did not induce cocaine sensitization, and did not affect the rewarding effects of cocaine measured in the conditioned place preference assay. The elevated cocaine-induced hyperlocomotion in HR mice was associated with increased protein Fos expression in the dorsal medial striatum (DMS) compared with WT. Lastly, we performed an RNA fluorescent in situ hybridization experiment and found that Reln was highly co-expressed with the Drd1 gene, which encodes for the dopamine receptor D1, in the DMS. These findings show that Reelin signaling contributes to the locomotor effects of cocaine and improve our understanding of the neurobiological mechanisms underlying the cellular and behavioral effects of cocaine.
Collapse
Affiliation(s)
- Giordano de Guglielmo
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Attilio Iemolo
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Aisha Nur
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Andrew Turner
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | | | - Angelica Martinez
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Caitlin Crook
- Department of Psychiatry, University of California, San Diego, La Jolla, California, USA
| | - Amanda Roberts
- Animal Models Core Facility, The Scripps Research Institute, La Jolla, California, USA
| | - Francesca Telese
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
30
|
Bergmann T, Liu Y, Skov J, Mogus L, Lee J, Pfisterer U, Handfield LF, Asenjo-Martinez A, Lisa-Vargas I, Seemann SE, Lee JTH, Patikas N, Kornum BR, Denham M, Hyttel P, Witter MP, Gorodkin J, Pers TH, Hemberg M, Khodosevich K, Hall VJ. Production of human entorhinal stellate cell-like cells by forward programming shows an important role of Foxp1 in reprogramming. Front Cell Dev Biol 2022; 10:976549. [PMID: 36046338 PMCID: PMC9420913 DOI: 10.3389/fcell.2022.976549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Stellate cells are principal neurons in the entorhinal cortex that contribute to spatial processing. They also play a role in the context of Alzheimer's disease as they accumulate Amyloid beta early in the disease. Producing human stellate cells from pluripotent stem cells would allow researchers to study early mechanisms of Alzheimer's disease, however, no protocols currently exist for producing such cells. In order to develop novel stem cell protocols, we characterize at high resolution the development of the porcine medial entorhinal cortex by tracing neuronal and glial subtypes from mid-gestation to the adult brain to identify the transcriptomic profile of progenitor and adult stellate cells. Importantly, we could confirm the robustness of our data by extracting developmental factors from the identified intermediate stellate cell cluster and implemented these factors to generate putative intermediate stellate cells from human induced pluripotent stem cells. Six transcription factors identified from the stellate cell cluster including RUNX1T1, SOX5, FOXP1, MEF2C, TCF4, EYA2 were overexpressed using a forward programming approach to produce neurons expressing a unique combination of RELN, SATB2, LEF1 and BCL11B observed in stellate cells. Further analyses of the individual transcription factors led to the discovery that FOXP1 is critical in the reprogramming process and omission of RUNX1T1 and EYA2 enhances neuron conversion. Our findings contribute not only to the profiling of cell types within the developing and adult brain's medial entorhinal cortex but also provides proof-of-concept for using scRNAseq data to produce entorhinal intermediate stellate cells from human pluripotent stem cells in-vitro.
Collapse
Affiliation(s)
- Tobias Bergmann
- Group of Brain Development and Disease, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Yong Liu
- Group of Brain Development and Disease, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Jonathan Skov
- Group of Brain Development and Disease, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Leo Mogus
- Group of Brain Development and Disease, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Julie Lee
- Novo Nordisk Foundation Center for Stem Cell Research, DanStem University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulrich Pfisterer
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Andrea Asenjo-Martinez
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Irene Lisa-Vargas
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stefan E. Seemann
- Center for non-coding RNA in Technology and Health, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Jimmy Tsz Hang Lee
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Nikolaos Patikas
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Birgitte Rahbek Kornum
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mark Denham
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Poul Hyttel
- Disease, Stem Cells and Embryology, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Menno P. Witter
- Kavli Institute for Systems Neuroscience, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jan Gorodkin
- Center for non-coding RNA in Technology and Health, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Tune H. Pers
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Martin Hemberg
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Konstantin Khodosevich
- Biotech Research and Innovation Centre (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vanessa Jane Hall
- Group of Brain Development and Disease, Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
31
|
Frankowski JC, Tierno A, Pavani S, Cao Q, Lyon DC, Hunt RF. Brain-wide reconstruction of inhibitory circuits after traumatic brain injury. Nat Commun 2022; 13:3417. [PMID: 35701434 PMCID: PMC9197933 DOI: 10.1038/s41467-022-31072-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 05/31/2022] [Indexed: 11/09/2022] Open
Abstract
Despite the fundamental importance of understanding the brain's wiring diagram, our knowledge of how neuronal connectivity is rewired by traumatic brain injury remains remarkably incomplete. Here we use cellular resolution whole-brain imaging to generate brain-wide maps of the input to inhibitory neurons in a mouse model of traumatic brain injury. We find that somatostatin interneurons are converted into hyperconnected hubs in multiple brain regions, with rich local network connections but diminished long-range inputs, even at areas not directly damaged. The loss of long-range input does not correlate with cell loss in distant brain regions. Interneurons transplanted into the injury site receive orthotopic local and long-range input, suggesting the machinery for establishing distant connections remains intact even after a severe injury. Our results uncover a potential strategy to sustain and optimize inhibition after traumatic brain injury that involves spatial reorganization of the direct inputs to inhibitory neurons across the brain.
Collapse
Affiliation(s)
- Jan C Frankowski
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Alexa Tierno
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, 92697, USA.
| | - Shreya Pavani
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Quincy Cao
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, 92697, USA
| | - David C Lyon
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, 92697, USA
| | - Robert F Hunt
- Department of Anatomy & Neurobiology, University of California, Irvine, CA, 92697, USA. .,Epilepsy Research Center, University of California, Irvine, CA, 92697, USA. .,Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, CA, 92697, USA. .,Center for the Neurobiology of Learning and Memory, University of California, Irvine, Irvine, CA, 92697, USA. .,Center for Neural Circuit Mapping, University of California, Irvine, Irvine, CA, 92697, USA.
| |
Collapse
|
32
|
New Strategies for the Treatment of Neuropsychiatric Disorders Based on Reelin Dysfunction. Int J Mol Sci 2022; 23:ijms23031829. [PMID: 35163751 PMCID: PMC8836358 DOI: 10.3390/ijms23031829] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/31/2022] [Accepted: 02/03/2022] [Indexed: 12/16/2022] Open
Abstract
Reelin is an extracellular matrix protein that is mainly produced in Cajal-Retzius cells and controls neuronal migration, which is important for the proper formation of cortical layers in the developmental stage of the brain. In the adult brain, Reelin plays a crucial role in the regulation of N-methyl-D-aspartate receptor-dependent synaptic function, and its expression decreases postnatally. Clinical studies showed reductions in Reelin protein and mRNA expression levels in patients with psychiatric disorders; however, the causal relationship remains unclear. Reelin-deficient mice exhibit an abnormal neuronal morphology and behavior, while Reelin supplementation ameliorates learning deficits, synaptic dysfunctions, and spine loss in animal models with Reelin deficiency. These findings suggest that the neuronal deficits and brain dysfunctions associated with the down-regulated expression of Reelin are attenuated by enhancements in its expression and functions in the brain. In this review, we summarize findings on the role of Reelin in neuropsychiatric disorders and discuss potential therapeutic approaches for neuropsychiatric disorders associated with Reelin dysfunctions.
Collapse
|
33
|
Blankvoort S, Olsen LC, Kentros CG. Single Cell Transcriptomic and Chromatin Profiles Suggest Layer Vb Is the Only Layer With Shared Excitatory Cell Types in the Medial and Lateral Entorhinal Cortex. Front Neural Circuits 2022; 15:806154. [PMID: 35153682 PMCID: PMC8826650 DOI: 10.3389/fncir.2021.806154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 12/06/2021] [Indexed: 11/21/2022] Open
Abstract
All brain functionality arises from the activity in neural circuits in different anatomical regions. These regions contain different circuits comprising unique cell types. An integral part to understanding neural circuits is a full census of the constituent parts, i.e., the neural cell types. This census can be based on different characteristics. Previously combinations of morphology and physiology, gene expression, and chromatin accessibility have been used in various cortical and subcortical regions. This has given an extensive yet incomplete overview of neural cell types. However, these techniques have not been applied to all brain regions. Here we apply single cell analysis of accessible chromatin on two similar but different cortical regions, the medial and the lateral entorhinal cortices. Even though these two regions are anatomically similar, their intrinsic and extrinsic connectivity are different. In 4,136 cells we identify 20 different clusters representing different cell types. As expected, excitatory cells show regionally specific clusters, whereas inhibitory neurons are shared between regions. We find that several deep layer excitatory neuronal cell types as defined by chromatin profile are also shared between the two different regions. Integration with a larger scRNA-seq dataset maintains this shared characteristic for cells in Layer Vb. Interestingly, this layer contains three clusters, two specific to either subregion and one shared between the two. These clusters can be putatively associated with particular functional and anatomical cell types found in this layer. This information is a step forwards into elucidating the cell types within the entorhinal circuit and by extension its functional underpinnings.
Collapse
Affiliation(s)
- Stefan Blankvoort
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim, Norway
- *Correspondence: Stefan Blankvoort
| | - Lene Christin Olsen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- BioCore Bioinformatics Core Facility, NTNU, Trondheim, Norway
- Department of Neurology, St. Olavs Hospital, Trondheim, Norway
| | - Clifford G. Kentros
- Kavli Institute for Systems Neuroscience and Centre for Neural Computation, NTNU, Trondheim, Norway
| |
Collapse
|
34
|
Ogawa B, Nakanishi Y, Wakamatsu M, Takahashi Y, Shibutani M. Repeated administration of acrylamide for 28 days reduces late-stage progenitor cells and immature granule cells accompanying impaired neurite outgrowth in the adult hippocampal neurogenesis in young-adult rats. J Toxicol Sci 2022; 47:467-482. [DOI: 10.2131/jts.47.467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Bunichiro Ogawa
- Drug Safety and Pharmacokinetics Laboratories, Taisho Pharmaceutical Co., Ltd
| | - Yutaka Nakanishi
- Drug Safety and Pharmacokinetics Laboratories, Taisho Pharmaceutical Co., Ltd
| | - Masaki Wakamatsu
- Drug Safety and Pharmacokinetics Laboratories, Taisho Pharmaceutical Co., Ltd
| | - Yasunori Takahashi
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology
| | - Makoto Shibutani
- Institute of Global Innovation Research, Tokyo University of Agriculture and Technology
| |
Collapse
|
35
|
Timmerman BM, Mooney-Leber SM, Brummelte S. The effects of neonatal procedural pain and maternal isolation on hippocampal cell proliferation and reelin concentration in neonatal and adult male and female rats. Dev Psychobiol 2021; 63:e22212. [PMID: 34813104 DOI: 10.1002/dev.22212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 10/04/2021] [Accepted: 10/13/2021] [Indexed: 11/11/2022]
Abstract
Preterm births accounted for over 10% of all U.S. live births in 2019 and the rate is rising. Neonatal stressors, especially procedural pain, experienced by preterm infants in the neonatal intensive care unit (NICU) have been associated with neurodevelopmental impairments. Parental care can alleviate stress during stressful or painful procedures; however, infants in the NICU often receive reduced parental care compared with their peers. Animal studies suggest that decreased maternal care similarly impairs neurodevelopment but also influences the effects of neonatal pain. It is important to mimic both stressors in animal models of neonatal stress exposure. In this study, researchers investigated the individual and combined impact of neonatal pain and maternal isolation on reelin protein levels and cellular proliferation in the hippocampal dentate gyrus of 8 days old and adult rats. Exposure to either stressor individually, but not both, increased reelin levels in the dentate gyrus of adult females without significantly altering reelin levels in adult males. However, cell proliferation levels at either age were unaffected by the early-life stressors. These results suggest that each early-life stressor has a unique effect on markers of brain development and more research is needed to further investigate their distinct influences.
Collapse
Affiliation(s)
- Brian M Timmerman
- Department of Psychology, Wayne State University, Detroit, Michigan, USA
| | - Sean M Mooney-Leber
- Department of Psychology, University of Wisconsin-Stevens Points, Stevens Point, Wisconsin, USA
| | - Susanne Brummelte
- Department of Psychology, Wayne State University, Detroit, Michigan, USA.,Translational Neuroscience Program, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
36
|
Jiménez S, Moreno N. Analysis of the Expression Pattern of Cajal-Retzius Cell Markers in the Xenopus laevis Forebrain. BRAIN, BEHAVIOR AND EVOLUTION 2021; 96:263-282. [PMID: 34614492 DOI: 10.1159/000519025] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 08/09/2021] [Indexed: 01/26/2023]
Abstract
Cajal-Retzius cells are essential for cortical development in mammals, and their involvement in the evolution of this structure has been widely postulated, but very little is known about their progenitor domains in non-mammalian vertebrates. Using in situhybridization and immunofluorescence techniques we analyzed the expression of some of the main Cajal-Retzius cell markers such as Dbx1, Ebf3, ER81, Lhx1, Lhx5, p73, Reelin, Wnt3a, Zic1, and Zic2 in the forebrain of the anuran Xenopus laevis, because amphibians are the only class of anamniote tetrapods and show a tetrapartite evaginated pallium, but no layered or nuclear organization. Our results suggested that the Cajal-Retzius cell progenitor domains were comparable to those previously described in amniotes. Thus, at dorsomedial telencephalic portions a region comparable to the cortical hem was defined in Xenopus based on the expression of Wnt3a, p73, Reelin, Zic1, and Zic2. In the septum, two different domains were observed: a periventricular dorsal septum, at the limit between the pallium and the subpallium, expressing Reelin, Zic1, and Zic2, and a related septal domain, expressing Ebf3, Zic1, and Zic2. In the lateral telencephalon, the ventral pallium next to the pallio-subpallial boundary, the lack of Dbx1 and the unique expression of Reelin during development defined this territory as the most divergent with respect to mammals. Finally, we also analyzed the expression of these markers at the prethalamic eminence region, suggested as Cajal-Retzius progenitor domain in amniotes, observing there Zic1, Zic2, ER81, and Lhx1 expression. Our data show that in anurans there are different subtypes and progenitor domains of Cajal-Retzius cells, which probably contribute to the cortical regional specification and territory-specific properties. This supports the notion that the basic organization of pallial derivatives in vertebrates follows a comparable fundamental arrangement, even in those that do not have a sophisticated stratified cortical structure like the mammalian cerebral cortex.
Collapse
Affiliation(s)
- Sara Jiménez
- Department of Cell Biology, Faculty of Biology, University Complutense, Madrid, Spain
| | - Nerea Moreno
- Department of Cell Biology, Faculty of Biology, University Complutense, Madrid, Spain
| |
Collapse
|
37
|
Orcinha C, Kilias A, Paschen E, Follo M, Haas CA. Reelin Is Required for Maintenance of Granule Cell Lamination in the Healthy and Epileptic Hippocampus. Front Mol Neurosci 2021; 14:730811. [PMID: 34483838 PMCID: PMC8414139 DOI: 10.3389/fnmol.2021.730811] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/26/2021] [Indexed: 11/21/2022] Open
Abstract
One characteristic feature of mesial temporal lobe epilepsy is granule cell dispersion (GCD), a pathological widening of the granule cell layer in the dentate gyrus. The loss of the extracellular matrix protein Reelin, an important positional cue for neurons, correlates with GCD formation in MTLE patients and in rodent epilepsy models. Here, we used organotypic hippocampal slice cultures (OHSC) from transgenic mice expressing enhanced green fluorescent protein (eGFP) in differentiated granule cells (GCs) to monitor GCD formation dynamically by live cell video microscopy and to investigate the role of Reelin in this process. We present evidence that following treatment with the glutamate receptor agonist kainate (KA), eGFP-positive GCs migrated mainly toward the hilar region. In the hilus, Reelin-producing neurons were rapidly lost following KA treatment as shown in a detailed time series. Addition of recombinant Reelin fragments to the medium effectively prevented the KA-triggered movement of eGFP-positive GCs. Placement of Reelin-coated beads into the hilus of KA-treated cultures stopped the migration of GCs in a distance-dependent manner. In addition, quantitative Western blot analysis revealed that KA treatment affects the Reelin signal transduction pathway by increasing intracellular adaptor protein Disabled-1 synthesis and reducing the phosphorylation of cofilin, a downstream target of the Reelin pathway. Both events were normalized by addition of recombinant Reelin fragments. Finally, following neutralization of Reelin in healthy OHSC by incubation with the function-blocking CR-50 Reelin antibody, GCs started to migrate without any direction preference. Together, our findings demonstrate that normotopic position of Reelin is essential for the maintenance of GC lamination in the dentate gyrus and that GCD is the result of a local Reelin deficiency.
Collapse
Affiliation(s)
- Catarina Orcinha
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Antje Kilias
- Biomicrotechnology, Department of Microsystems Engineering (IMTEK), University of Freiburg, Freiburg im Breisgau, Germany
| | - Enya Paschen
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany.,Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Marie Follo
- Lighthouse Core Facility, Department of Internal Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Carola A Haas
- Experimental Epilepsy Research, Department of Neurosurgery, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany.,Center for Basics in NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
38
|
Hamad MIK, Petrova P, Daoud S, Rabaya O, Jbara A, Melliti N, Leifeld J, Jakovčevski I, Reiss G, Herz J, Förster E. Reelin restricts dendritic growth of interneurons in the neocortex. Development 2021; 148:272055. [PMID: 34414407 DOI: 10.1242/dev.199718] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/12/2021] [Indexed: 11/20/2022]
Abstract
Reelin is a large secreted glycoprotein that regulates neuronal migration, lamination and establishment of dendritic architecture in the embryonic brain. Reelin expression switches postnatally from Cajal-Retzius cells to interneurons. However, reelin function in interneuron development is still poorly understood. Here, we have investigated the role of reelin in interneuron development in the postnatal neocortex. To preclude early cortical migration defects caused by reelin deficiency, we employed a conditional reelin knockout (RelncKO) mouse to induce postnatal reelin deficiency. Induced reelin deficiency caused dendritic hypertrophy in distal dendritic segments of neuropeptide Y-positive (NPY+) and calretinin-positive (Calr+) interneurons, and in proximal dendritic segments of parvalbumin-positive (Parv+) interneurons. Chronic recombinant Reelin treatment rescued dendritic hypertrophy in Relncko interneurons. Moreover, we provide evidence that RelncKO interneuron hypertrophy is due to presynaptic GABABR dysfunction. Thus, GABABRs in RelncKO interneurons were unable to block N-type (Cav2.2) Ca2+ channels that control neurotransmitter release. Consequently, the excessive Ca2+ influx through AMPA receptors, but not NMDA receptors, caused interneuron dendritic hypertrophy. These findings suggest that reelin acts as a 'stop-growth-signal' for postnatal interneuron maturation.
Collapse
Affiliation(s)
- Mohammad I K Hamad
- Institute for Anatomy and Clinical Morphology, School of Medicine, Faculty of Health, Witten/Herdecke University, Witten 58455, Germany.,Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum 44801, Germany
| | - Petya Petrova
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum 44801, Germany
| | - Solieman Daoud
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum 44801, Germany
| | - Obada Rabaya
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum 44801, Germany
| | - Abdalrahim Jbara
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum 44801, Germany
| | - Nesrine Melliti
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum 44801, Germany
| | - Jennifer Leifeld
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum 44801, Germany
| | - Igor Jakovčevski
- Institute for Anatomy and Clinical Morphology, School of Medicine, Faculty of Health, Witten/Herdecke University, Witten 58455, Germany
| | - Gebhard Reiss
- Institute for Anatomy and Clinical Morphology, School of Medicine, Faculty of Health, Witten/Herdecke University, Witten 58455, Germany
| | - Joachim Herz
- Departments of Molecular Genetics, Neuroscience, Neurology and Neurotherapeutics; Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Eckart Förster
- Department of Neuroanatomy and Molecular Brain Research, Medical Faculty, Ruhr University Bochum, Bochum 44801, Germany
| |
Collapse
|
39
|
Nishibe M, Toyoda H, Hiraga SI, Yamashita T, Katsuyama Y. Synaptic and Genetic Bases of Impaired Motor Learning Associated with Modified Experience-Dependent Cortical Plasticity in Heterozygous Reeler Mutants. Cereb Cortex 2021; 32:504-519. [PMID: 34339488 DOI: 10.1093/cercor/bhab227] [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: 01/18/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 11/12/2022] Open
Abstract
Patients with neurodevelopmental disorders show impaired motor skill learning. It is unclear how the effect of genetic variation on synaptic function and transcriptome profile may underlie experience-dependent cortical plasticity, which supports the development of fine motor skills. RELN (reelin) is one of the genes implicated in neurodevelopmental psychiatric vulnerability. Heterozygous reeler mutant (HRM) mice displayed impairments in reach-to-grasp learning, accompanied by less extensive cortical map reorganization compared with wild-type mice, examined after 10 days of training by intracortical microstimulation. Assessed by patch-clamp recordings after 3 days of training, the training induced synaptic potentiation and increased glutamatergic-transmission of cortical layer III pyramidal neurons in wild-type mice. In contrast, the basal excitatory and inhibitory synaptic functions were depressed, affected both by presynaptic and postsynaptic impairments in HRM mice; and thus, no further training-induced synaptic plasticity occurred. HRM exhibited downregulations of cortical synaptophysin, immediate-early gene expressions, and gene enrichment, in response to 3 days of training compared with trained wild-type mice, shown using quantitative reverse transcription polymerase chain reaction, immunohistochemisty, and RNA-sequencing. We demonstrated that motor learning impairments associated with modified experience-dependent cortical plasticity are at least partially attributed by the basal synaptic alternation as well as the aberrant early experience-induced gene enrichment in HRM.
Collapse
Affiliation(s)
- Mariko Nishibe
- Office of Strategic Innovative Dentistry, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan.,Department of Anatomy, Shiga University of Medical Science, Shiga 520-2192, Japan
| | - Hiroki Toyoda
- Department of Oral Physiology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Shin-Ichiro Hiraga
- Department of Neuromedical Science, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Toshihide Yamashita
- Department of Neuromedical Science, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.,Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.,Department of Molecular Neuroscience, WPI Immunology Frontier Research Center, Osaka 565-0871, Japan.,Graduate School of Frontier Biosciences, Osaka University, Osaka 565-0871, Japan
| | - Yu Katsuyama
- Department of Anatomy, Shiga University of Medical Science, Shiga 520-2192, Japan
| |
Collapse
|
40
|
Reelin changes hippocampal learning in aging and Alzheimer's disease. Behav Brain Res 2021; 414:113482. [PMID: 34333070 DOI: 10.1016/j.bbr.2021.113482] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 11/23/2022]
Abstract
The hippocampal formation (HF) is a neuroanatomical region essential for learning and memory. As one of the earliest regions to display the histopathological hallmarks of Alzheimer's disease (AD), determining the specific mechanisms of the HF's vulnerability is of capital importance. Reelin, a glycoprotein crucial in cortical lamination during embryonic neurogenesis, has an uncommon expression pattern within the HF and has been implicated in both learning and AD pathogenesis. We hypothesized that Reelin deficiency would expedite behavioral impairments which accompany normal aging. Additionally, we hypothesized that Reelin deficiency in the presence of mutated human microtubule associated protein tau (MAPT) would further impair hippocampal function. To test our hypothesis, we utilized cohorts of aged mice, aged mice with Reelin conditional knockout (RcKO), and adult mice with both RcKO and MAPT in the Barnes maze and Trace fear conditioning. Consistent with prior literature, increased age in wild-type mice was sufficient to reduce spatial searching in the Barnes maze. Increased age both exacerbated spatial impairments and altered context learning in RcKO mice. Lastly, adult mice with both RcKO and the MAPT transgene displayed both the lowest age-of-onset and most severe spatial learning deficits. In conclusion, Reelin deficiency when combined with AD risk-factors produced consistent impairments in spatial memory tasks. Furthermore, our results further implicate Reelin's importance in both HF homeostasis and AD pathogenesis.
Collapse
|
41
|
Liu Y, Wang Y, Yuan W, Dong F, Zhen F, Liu J, Yang L, Qu X, Yao R. Reelin promotes oligodendrocyte precursors migration via the Wnt/β-catenin signaling pathway. Neurol Res 2021; 43:543-552. [PMID: 33616025 DOI: 10.1080/01616412.2021.1888604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 02/07/2021] [Indexed: 10/22/2022]
Abstract
Objectives: The extracellular matrix glycoprotein Reelin plays an important role in the development of the central nervous system and is involved in neurogenesis, neuronal polarization and migration. Although it has been reported that Reelin and its receptor are expressed in oligodendrocyte precursors (OPCs), the main functions and possible mechanism of Reelin in OPCs remain unclear.Methods: In this study, immunofluorescence staining was used to detect the expressions of A2B5, PDGFRα, Reelin, VLDLR and Dab1 in OPCs. The expression of p-Dab1 in OPCs which was treated with Reelin at different concentrations was assayed by western blot. Effects of Reelin on the proliferation of OPCs was measured by EdU and CCK-8. Annexin V-FITC/PI assayed the effect of Reelin on the apoptosis of OPCs. Effects of Reelin on the migration ability of OPCs were detected by the scratch test and transwell experiments. Immunoblotting was used to measure the activation of Wnt/β-catenin signaling with Reelin, while transwell experiments were performed to verify the migration of OPCs under the activation of Wnt/β-catenin signaling.Results: Results showed that the receptor of Reelin, very-low-density lipoprotein receptor (VLDLR), and its adaptor protein, Dab1, are highly expressed in A2B5/PDGFRα double-positive OPCs. Recombinant Reelin protein promoted OPCs migration in vitro but had no obvious effects on proliferation or apoptosis. Reelin also promoted the phosphorylation of Dab1 and increased the expression of β-catenin in OPCs. WIKI4, an inhibitor of Wnt/β-catenin signaling, suppressed the migration of OPCs induced by Reelin.Conclusion: The present study indicated that Reelin promotes OPCs migration via the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Yaping Liu
- Laboratory of National Experimental Teaching and Demonstration Center of Basic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, PRC
| | - Yuanyuan Wang
- Pediatrics, Nanjing Tongren Hospital, Nanjing, Jiangsu, PRC
| | - Wen Yuan
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, PRC
| | - Fuxing Dong
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, PRC
| | - Fei Zhen
- Department of Pathology, Hongze District People's Hospital, Huai 'an, Jiangsu, PRC
| | - Jing Liu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, PRC
| | - Lihua Yang
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, PRC
| | - Xuebin Qu
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, PRC
| | - Ruiqin Yao
- Department of Cell Biology and Neurobiology, Xuzhou Key Laboratory of Neurobiology, Xuzhou Medical University, Xuzhou, Jiangsu, PRC
| |
Collapse
|
42
|
Pahle J, Muhia M, Wagener RJ, Tippmann A, Bock HH, Graw J, Herz J, Staiger JF, Drakew A, Kneussel M, Rune GM, Frotscher M, Brunne B. Selective Inactivation of Reelin in Inhibitory Interneurons Leads to Subtle Changes in the Dentate Gyrus But Leaves Cortical Layering and Behavior Unaffected. Cereb Cortex 2021; 30:1688-1707. [PMID: 31667489 PMCID: PMC7132935 DOI: 10.1093/cercor/bhz196] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Reelin is an extracellular matrix protein, known for its dual role in neuronal migration during brain development and in synaptic plasticity at adult stages. During the perinatal phase, Reelin expression switches from Cajal-Retzius (CR) cells, its main source before birth, to inhibitory interneurons (IN), the main source of Reelin in the adult forebrain. IN-derived Reelin has been associated with schizophrenia and temporal lobe epilepsy; however, the functional role of Reelin from INs is presently unclear. In this study, we used conditional knockout mice, which lack Reelin expression specifically in inhibitory INs, leading to a substantial reduction in total Reelin expression in the neocortex and dentate gyrus. Our results show that IN-specific Reelin knockout mice exhibit normal neuronal layering and normal behavior, including spatial reference memory. Although INs are the major source of Reelin within the adult stem cell niche, Reelin from INs does not contribute substantially to normal adult neurogenesis. While a closer look at the dentate gyrus revealed some unexpected alterations at the cellular level, including an increase in the number of Reelin expressing CR cells, overall our data suggest that Reelin derived from INs is less critical for cortex development and function than Reelin expressed by CR cells.
Collapse
Affiliation(s)
- Jasmine Pahle
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Mary Muhia
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Robin J Wagener
- Neurology Clinic, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Anja Tippmann
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Department of Systems Neuroscience, Johann-Friedrich-Blumenbach Institute for Zoology and Anthropology, University of Göttingen, 37075 Göttingen, Germany
| | - Hans H Bock
- Clinic of Gastroenterology and Hepatology, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Janice Graw
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center Göttingen, Georg-August-University Göttingen, 37075 Göttingen, Germany
| | - Alexander Drakew
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Institute of Clinical Neuroanatomy, Faculty of Medicine, 60590 Frankfurt, Germany
| | - Matthias Kneussel
- Institute of Molecular Neurogenetics, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Gabriele M Rune
- Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Michael Frotscher
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Bianka Brunne
- Institute for Structural Neurobiology, Center for Molecular Neurobiology, ZMNH, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.,Institute of Neuroanatomy, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| |
Collapse
|
43
|
Shin HY, Han KS, Park HW, Hong YH, Kim Y, Moon HE, Park KW, Park HR, Lee CJ, Lee K, Kim SJ, Heo MS, Park SH, Kim DG, Paek SH. Tumor Spheroids of an Aggressive Form of Central Neurocytoma Have Transit-Amplifying Progenitor Characteristics with Enhanced EGFR and Tumor Stem Cell Signaling. Exp Neurobiol 2021; 30:120-143. [PMID: 33972466 PMCID: PMC8118755 DOI: 10.5607/en21004] [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: 03/15/2021] [Revised: 03/26/2021] [Accepted: 03/26/2021] [Indexed: 11/19/2022] Open
Abstract
Central neurocytoma (CN) has been known as a benign neuronal tumor. In rare cases, CN undergoes malignant transformation to glioblastomas (GBM). Here we examined its cellular origin by characterizing differentiation potential and gene expression of CN-spheroids. First, we demonstrate that both CN tissue and cultured primary cells recapitulate the hierarchal cellular composition of subventricular zone (SVZ), which is comprised of neural stem cells (NSCs), transit amplifying progenitors (TAPs), and neuroblasts. We then derived spheroids from CN which displayed EGFR+/MASH+ TAP and BLBP+ radial glial cell (RGC) characteristic, and mitotic neurogenesis and gliogenesis by single spheroids were observed with cycling multipotential cells. CN-spheroids expressed increased levels of pluripotency and tumor stem cell genes such as KLF4 and TPD5L1, when compared to their differentiated cells and human NSCs. Importantly, Gene Set Enrichment Analysis showed that gene sets of GBM-Spheroids, EGFR Signaling, and Packaging of Telomere Ends are enriched in CN-spheroids in comparison with their differentiated cells. We speculate that CN tumor stem cells have TAP and RGC characteristics, and upregulation of EGFR signaling as well as downregulation of eph-ephrin signaling have critical roles in tumorigenesis of CN. And their ephemeral nature of TAPs destined to neuroblasts, might reflect benign nature of CN.
Collapse
Affiliation(s)
- Hye Young Shin
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Kyung-Seok Han
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Hyung Woo Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Yun Hwa Hong
- Department of Neurophysiology, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Yona Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Hyo Eun Moon
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Kwang Woo Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Hye Ran Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Kiyoung Lee
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Sang Jeong Kim
- Department of Neurophysiology, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Man Seung Heo
- Smart Healthcare Medical Device Research Center, Samsung Medical Center, Seoul 06351, Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Dong Gyu Kim
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea
| | - Sun Ha Paek
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul 03082, Korea.,Ischemic/Hypoxic Disease Institute, Cancer Research Institute, Seoul National University College of Medicine, Seoul 03082, Korea.,Clinical Research Institute, Seoul National University Hospital, Seoul 03082, Korea
| |
Collapse
|
44
|
Tsuneura Y, Sawahata M, Itoh N, Miyajima R, Mori D, Kohno T, Hattori M, Sobue A, Nagai T, Mizoguchi H, Nabeshima T, Ozaki N, Yamada K. Analysis of Reelin signaling and neurodevelopmental trajectory in primary cultured cortical neurons with RELN deletion identified in schizophrenia. Neurochem Int 2021; 144:104954. [PMID: 33388358 DOI: 10.1016/j.neuint.2020.104954] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/20/2020] [Accepted: 12/27/2020] [Indexed: 10/22/2022]
Abstract
Reelin, an extracellular matrix protein, is secreted by Cajal-Retzius cells and plays crucial roles in the development of brain structures and neuronal functions. Reductions in Reelin cause the brain dysfunctions associated with mental disorders, such as schizophrenia. A recent genome-wide copy number variation analysis of Japanese schizophrenia patients identified a novel deletion in RELN encoding Reelin. To clarify the pathophysiological role of the RELN deletion, we developed transgenic mice carrying the RELN deletion (Reln-del) and found abnormalities in their brain structures and social behavior. In the present study, we performed an in vitro analysis of Reelin expression, intracellular Reelin signaling, and the morphology of primary cultured cortical neurons from wild-type (WT) and Reln-del mice. Reelin protein levels were lower in Reln-del neurons than in WT neurons. Dab1 expression levels were significantly higher in Reln-del neurons than in WT neurons, suggesting that Reelin signaling was decreased in Reln-del neurons. Reelin was mainly expressed in γ-aminobutyric acid (GABA)-ergic inhibitory neurons, but not in parvalbumin (PV)-positive neurons. A small proportion of Ca2+/calmodulin-dependent protein kinase II α subunit (CaMKIIα)-positive excitatory neurons also expressed Reelin. In comparisons with WT neurons, significant decreases were observed in neurite lengths and branch points as well as in the number of postsynaptic density protein 95 (PSD95) immunoreactive puncta in Reln-del neurons. A disintegrin and metalloproteinase with thrombospondin motifs-3 (ADAMTS-3) is a protease that inactivates Reelin by cleavage at the N-t site. The knockdown of ADAMTS-3 by short hairpin RNAs suppressed Reelin cleavage in conditioned medium and reduced Dab1 expression, indicating that Reelin signaling was enhanced in the primary cultured cortical neurons of WT and heterozygous Reln-del. Accordingly, the inhibition of ADAMTS-3 may be a potential candidate in the clinical treatment of schizophrenia by enhancing Reelin signaling in the brain.
Collapse
Affiliation(s)
- Yumi Tsuneura
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Masahito Sawahata
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Norimichi Itoh
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Ryoya Miyajima
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan; Brain and Mind Research Center, Nagoya University, Nagoya, Aichi, Japan
| | - Takao Kohno
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Mitsuharu Hattori
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan
| | - Akira Sobue
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Taku Nagai
- Division of Behavioral Neuropharmacology, Project Office for Neuropsychological Research Center, Fujita Health University, Toyoake, Aichi, Japan
| | - Hiroyuki Mizoguchi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Graduate School of Health Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University, Graduate School of Medicine, Nagoya, Aichi, Japan.
| |
Collapse
|
45
|
Allen J, Caruncho HJ, Kalynchuk LE. Severe life stress, mitochondrial dysfunction, and depressive behavior: A pathophysiological and therapeutic perspective. Mitochondrion 2020; 56:111-117. [PMID: 33220501 DOI: 10.1016/j.mito.2020.11.010] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/28/2020] [Accepted: 11/11/2020] [Indexed: 01/11/2023]
Abstract
Mitochondria are responsible for providing our cells with energy, as well as regulating oxidative stress and apoptosis, and considerable evidence demonstrates that mitochondria-related alterations are prevalent during chronic stress and depression. Here, we discuss how chronic stress may induce depressive behavior by potentiating mitochondrial allostatic load, which ultimately decreases energy production, elevates the generation of harmful reactive oxygen species, damages mitochondrial DNA and increases membrane permeability and pro-apoptotic factor release. We also discuss how mitochondrial insults can exacerbate the immune response, contributing to depressive symptomology. Furthermore, we illustrate how depression symptoms are associated with specific mitochondrial defects, and how targeting of these defects with pharmacological agents may be a promising avenue for the development of novel, more efficacious antidepressants. In summary, this review supports the notion that severe psychosocial stress induces mitochondrial dysfunction, thereby increasing the vulnerability to developing depressive symptoms.
Collapse
Affiliation(s)
- Josh Allen
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Lisa E Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
46
|
Macrì S, Di-Poï N. Heterochronic Developmental Shifts Underlying Squamate Cerebellar Diversity Unveil the Key Features of Amniote Cerebellogenesis. Front Cell Dev Biol 2020; 8:593377. [PMID: 33195265 PMCID: PMC7642464 DOI: 10.3389/fcell.2020.593377] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 09/25/2020] [Indexed: 11/13/2022] Open
Abstract
Despite a remarkable conservation of architecture and function, the cerebellum of vertebrates shows extensive variation in morphology, size, and foliation pattern. These features make this brain subdivision a powerful model to investigate the evolutionary developmental mechanisms underlying neuroanatomical complexity both within and between anamniote and amniote species. Here, we fill a major evolutionary gap by characterizing the developing cerebellum in two non-avian reptile species-bearded dragon lizard and African house snake-representative of extreme cerebellar morphologies and neuronal arrangement patterns found in squamates. Our data suggest that developmental strategies regarded as exclusive hallmark of birds and mammals, including transit amplification in an external granule layer (EGL) and Sonic hedgehog expression by underlying Purkinje cells (PCs), contribute to squamate cerebellogenesis independently from foliation pattern. Furthermore, direct comparison of our models suggests the key importance of spatiotemporal patterning and dynamic interaction between granule cells and PCs in defining cortical organization. Especially, the observed heterochronic shifts in early cerebellogenesis events, including upper rhombic lip progenitor activity and EGL maintenance, are strongly expected to affect the dynamics of molecular interaction between neuronal cell types in snakes. Altogether, these findings help clarifying some of the morphogenetic and molecular underpinnings of amniote cerebellar corticogenesis, but also suggest new potential molecular mechanisms underlying cerebellar complexity in squamates. Furthermore, squamate models analyzed here are revealed as key animal models to further understand mechanisms of brain organization.
Collapse
Affiliation(s)
- Simone Macrì
- Program in Developmental Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Nicolas Di-Poï
- Program in Developmental Biology, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
47
|
Reelin Mediates Hippocampal Cajal-Retzius Cell Positioning and Infrapyramidal Blade Morphogenesis. J Dev Biol 2020; 8:jdb8030020. [PMID: 32962021 PMCID: PMC7558149 DOI: 10.3390/jdb8030020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/10/2020] [Accepted: 09/14/2020] [Indexed: 12/31/2022] Open
Abstract
We have previously described hypomorphic reelin (Reln) mutant mice, RelnCTRdel, in which the morphology of the dentate gyrus is distinct from that seen in reeler mice. In the RelnCTRdel mutant, the infrapyramidal blade of the dentate gyrus fails to extend, while the suprapyramidal blade forms with a relatively compact granule neuron layer. Underlying this defect, we now report several developmental anomalies in the RelnCTRdel dentate gyrus. Most strikingly, the distribution of Cajal-Retzius cells was aberrant; Cajal-Retzius neurons were increased in the suprapyramidal blade, but were greatly reduced along the subpial surface of the prospective infrapyramidal blade. We also observed multiple abnormalities of the fimbriodentate junction. Firstly, progenitor cells were distributed abnormally; the “neurogenic cluster” at the fimbriodentate junction was absent, lacking the normal accumulation of Tbr2-positive intermediate progenitors. However, the number of dividing cells in the dentate gyrus was not generally decreased. Secondly, a defect of secondary glial scaffold formation, limited to the infrapyramidal blade, was observed. The densely radiating glial fibers characteristic of the normal fimbriodentate junction were absent in mutants. These fibers might be required for migration of progenitors, which may account for the failure of neurogenic cluster formation. These findings suggest the importance of the secondary scaffold and neurogenic cluster of the fimbriodentate junction in morphogenesis of the mammalian dentate gyrus. Our study provides direct genetic evidence showing that normal RELN function is required for Cajal-Retzius cell positioning in the dentate gyrus, and for formation of the fimbriodentate junction to promote infrapyramidal blade extension.
Collapse
|
48
|
Ibi D, Nakasai G, Koide N, Sawahata M, Kohno T, Takaba R, Nagai T, Hattori M, Nabeshima T, Yamada K, Hiramatsu M. Reelin Supplementation Into the Hippocampus Rescues Abnormal Behavior in a Mouse Model of Neurodevelopmental Disorders. Front Cell Neurosci 2020; 14:285. [PMID: 32982694 PMCID: PMC7492784 DOI: 10.3389/fncel.2020.00285] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/11/2020] [Indexed: 12/19/2022] Open
Abstract
In the majority of schizophrenia patients, chronic atypical antipsychotic administration produces a significant reduction in or even complete remission of psychotic symptoms such as hallucinations and delusions. However, these drugs are not effective in improving cognitive and emotional deficits in patients with schizophrenia. Atypical antipsychotic drugs have a high affinity for the dopamine D2 receptor, and a modest affinity for the serotonin 5-HT2A receptor. The cognitive and emotional deficits in schizophrenia are thought to involve neural networks beyond the classical dopaminergic mesolimbic pathway, however, including serotonergic systems. For example, mutations in the RELN gene, which encodes Reelin, an extracellular matrix protein involved in neural development and synaptic plasticity, are associated with neurodevelopmental disorders such as schizophrenia and autism spectrum disorder. Furthermore, hippocampal Reelin levels are down-regulated in the brains of both schizophrenic patients and in rodent models of schizophrenia. In the present study, we investigated the effect of Reelin microinjection into the mouse hippocampus on behavioral phenotypes to evaluate the role of Reelin in neurodevelopmental disorders and to test a therapeutic approach that extends beyond classical monoamine targets. To model the cognitive and emotional deficits, as well as histological decreases in Reelin-positive cell numbers and hippocampal synaptoporin distribution, a synaptic vesicle protein, offspring that were prenatally exposed to maternal immune activation were used. Microinjections of recombinant Reelin protein into the hippocampus rescued impairments in object memory and anxiety-like behavior and recruited synaptoporin in the hippocampus in offspring exposed to antenatal inflammation. These results suggest that Reelin supplementation has the potential to treat cognitive and emotional impairments, as well as synaptic disturbances, in patients with neurodevelopmental disorders such as schizophrenia.
Collapse
Affiliation(s)
- Daisuke Ibi
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Genki Nakasai
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Nayu Koide
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Masahito Sawahata
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takao Kohno
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Rika Takaba
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Project Office for Neuropsychological Research Center, Fujita Health University, Toyoake, Japan
| | - Mitsuharu Hattori
- Department of Biomedical Science, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University, Graduate School of Health Sciences, Toyoake, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masayuki Hiramatsu
- Department of Chemical Pharmacology, Faculty of Pharmacy, Meijo University, Nagoya, Japan
| |
Collapse
|
49
|
Brymer KJ, Johnston J, Botterill JJ, Romay-Tallon R, Mitchell MA, Allen J, Pinna G, Caruncho HJ, Kalynchuk LE. Fast-acting antidepressant-like effects of Reelin evaluated in the repeated-corticosterone chronic stress paradigm. Neuropsychopharmacology 2020; 45:1707-1716. [PMID: 31926481 PMCID: PMC7419539 DOI: 10.1038/s41386-020-0609-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/10/2019] [Accepted: 01/02/2020] [Indexed: 02/07/2023]
Abstract
The present report examines the effects of repeated or single intrahippocampal Reelin infusions on measures of depressive-like behavior, cognition, and hippocampal neurogenesis in the repeated-corticosterone (CORT) paradigm. Rats received subcutaneous injections of CORT for 3 weeks and Reelin was infused through an inserted canula in the left hippocampus on days 7, 14, and 21, or only on day 21 of CORT injections. CORT increased immobility in the forced-swim test and impaired object-location memory. Notably, these effects were reversed by both repeated and single-Reelin infusions. CORT decreased both the number and complexity of doublecortin-labeled maturing newborn neurons in the dentate gyrus subgranular zone, and a single-Reelin infusion increased the number but not complexity of newborn neurons, while repeated Reelin infusions restored both. Injection of the AMPA antagonist CNQX blocked the rescue of the behavioral phenotype by Reelin but did completely block the effects of Reelin on hippocampal neurogenesis. Reelin is able to rescue the deficits in AMPA, NMDA, GABAA receptors, mTOR and p-mTOR induced by CORT. These novel results demonstrate that a single intrahippocampal Reelin infusion into the dorsal hippocampus has fast-acting antidepressant-like effects, and that some of these effects may be at least partially independent of Reelin actions on hippocampal neurogenesis.
Collapse
Affiliation(s)
- Kyle J Brymer
- Department of Psychology, University of Saskatchewan, Saskatoon, SK, S7N 5A5, Canada
| | - Jenessa Johnston
- Department of Psychology, University of Saskatchewan, Saskatoon, SK, S7N 5A5, Canada
| | - Justin J Botterill
- Center for Dementia Research, The Nathan Kline Institute for Psychiatric Research, Orangeburg, NY, 10962, USA
| | | | - Milann A Mitchell
- Department of Psychology, University of Saskatchewan, Saskatoon, SK, S7N 5A5, Canada
| | - Josh Allen
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Graziano Pinna
- The Psychiatric Institute. Department of Psychiatry, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
| | - Lisa E Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
50
|
Yu J, Cho E, Choi YG, Jeong YK, Na Y, Kim JS, Cho SR, Woo JS, Bae S. Purification of an Intact Human Protein Overexpressed from Its Endogenous Locus via Direct Genome Engineering. ACS Synth Biol 2020; 9:1591-1598. [PMID: 32584551 DOI: 10.1021/acssynbio.0c00090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The overproduction and purification of human proteins is a requisite of both basic and medical research. Although many recombinant human proteins have been purified, current protein production methods have several limitations; recombinant proteins are frequently truncated, fail to fold properly, and/or lack appropriate post-translational modifications. In addition, such methods require subcloning of the target gene into relevant plasmids, which can be difficult for long proteins with repeated domains. Here we devised a novel method for target protein production by introduction of a strong promoter for overexpression and an epitope tag for purification in front of the endogenous human gene, in a sense performing molecular cloning directly in the human genome, which does not require cloning of the target gene. As a proof of concept, we successfully purified intact human Reelin protein, which is lengthy (3460 amino acids) and contains repeating domains, and confirmed that it was biologically functional.
Collapse
Affiliation(s)
- Jihyeon Yu
- Department of Chemistry, Hanyang University, Seoul 04763, South Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, South Korea
| | - Eunju Cho
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul 03722, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Yeon-Gil Choi
- Department of Life Sciences, Korea University, Seoul 02841, South Korea
| | - You Kyeong Jeong
- Department of Chemistry, Hanyang University, Seoul 04763, South Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, South Korea
| | - Yongwoo Na
- Center for RNA Research, Institute for Basic Science (IBS), Seoul 08826, South Korea
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Jong-Seo Kim
- Center for RNA Research, Institute for Basic Science (IBS), Seoul 08826, South Korea
- School of Biological Sciences, Seoul National University, Seoul 08826, South Korea
| | - Sung-Rae Cho
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul 03722, South Korea
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, South Korea
- Graduate Program of Nano Science and Technology, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Jae-Sung Woo
- Department of Life Sciences, Korea University, Seoul 02841, South Korea
| | - Sangsu Bae
- Department of Chemistry, Hanyang University, Seoul 04763, South Korea
- Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, South Korea
| |
Collapse
|