1
|
Shahbaz S, Rezaeifar M, Syed H, Redmond D, Terveart JWC, Osman M, Elahi S. Upregulation of olfactory receptors and neuronal-associated genes highlights complex immune and neuronal dysregulation in Long COVID patients. Brain Behav Immun 2025; 124:97-114. [PMID: 39615603 DOI: 10.1016/j.bbi.2024.11.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/04/2024] [Accepted: 11/27/2024] [Indexed: 01/20/2025] Open
Abstract
A substantial portion of patients infected with SARS-CoV-2 experience prolonged complications, known as Long COVID (LC). A subset of these patients exhibits the most debilitating symptoms, similar to those defined in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). We performed bulk RNA sequencing (RNAseq) on the whole blood of LC with ME/CFS, at least 12 months post-onset of the acute disease, and compared them with controls. We found that LC patients had a distinct transcriptional profile compared to controls. Key findings include the upregulation of genes involved in immune dysregulation and neuronal development, such as Fezf2, BRINP2, HOXC12, MEIS2, ZFHX3, and RELN. These genes are linked to neuroinflammatory responses, cognitive impairments, and hematopoietic disturbances, suggesting ongoing neurological and immune disturbances in LC patients. RELN, encoding the Reelin protein, was notably elevated in LC patients, potentially serving as a biomarker for LC pathogenesis due to its role in inflammation and neuronal function. Immune cell analysis showed altered profiles in LC patients, with increased activated memory CD4 + T cells and neutrophils, and decreased regulatory T cells and NK cells, reflecting immune dysregulation. Changes in cytokine and chemokine expression further underscore the chronic inflammatory state in LC patients. Notably, a unique upregulation of olfactory receptors (ORs) suggest alternative roles for ORs in non-olfactory tissues. Pathway analysis revealed upregulation in ribosomal RNA processing, amino acid metabolism, protein synthesis, cell proliferation, DNA repair, and mitochondrial pathways, indicating heightened metabolic and immune demands. Conversely, downregulated pathways, such as VEGF signaling and TP53 activity, point to impaired tissue repair and cellular stress responses. Overall, our study underscores the complex interplay between immune and neuronal dysfunction in LC patients, providing insights into potential diagnostic biomarkers and therapeutic targets. Future research is needed to fully understand the roles and interactions of these genes in LC pathophysiology.
Collapse
Affiliation(s)
- Shima Shahbaz
- Mike Petryk School of Dentistry, Division of Foundational Sciences, University of Alberta, Edmonton T6G 2E1, AB, Canada
| | - Maryam Rezaeifar
- Mike Petryk School of Dentistry, Division of Foundational Sciences, University of Alberta, Edmonton T6G 2E1, AB, Canada
| | - Hussein Syed
- Department of Medicine, Division of Gastroenterology, University of Alberta, Edmonton T6G 2E1, AB, Canada
| | - Desiree Redmond
- Department of Medicine, Division of Rheumatology, University of Alberta, Edmonton T6G 2E1, AB, Canada
| | - Jan Willem Cohen Terveart
- Department of Medicine, Division of Rheumatology, University of Alberta, Edmonton T6G 2E1, AB, Canada
| | - Mohammed Osman
- Department of Medicine, Division of Rheumatology, University of Alberta, Edmonton T6G 2E1, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton T6G 2E1, AB, Canada; Women and Children Health Research Institute, University of Alberta, Edmonton T6G 2E1, AB, Canada.
| | - Shokrollah Elahi
- Mike Petryk School of Dentistry, Division of Foundational Sciences, University of Alberta, Edmonton T6G 2E1, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton T6G 2E1, AB, Canada; Women and Children Health Research Institute, University of Alberta, Edmonton T6G 2E1, AB, Canada; Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton T6G 2E1, AB, Canada; Glycomics Institute of Alberta, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G 2E1, AB, Canada.
| |
Collapse
|
2
|
Scheil KKA, Sánchez-Lafuente CL, Reive BS, Halvorson CS, Floyd J, Reid HMO, Johnston JN, Kalynchuk LE, Caruncho HJ. Time-dependent antidepressant-like effects of reelin and ketamine in the repeated-corticosterone model of chronic stress. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110998. [PMID: 38552775 DOI: 10.1016/j.pnpbp.2024.110998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/01/2024]
Abstract
There is an urgent need for novel antidepressants, given that approximately 30% of those diagnosed with depression do not respond adequately to first-line treatment. Additionally, monoaminergic-based antidepressants have a substantial therapeutic time-lag, often taking months to reach full therapeutic effect. Ketamine, an N-methyl-d-aspartate receptor (NMDAR) antagonist is the only current effective rapid-acting antidepressant, demonstrating efficacy within hours and lasting up to two weeks with an acute dose. Reelin, an extracellular matrix glycoprotein, has demonstrated rapid-acting antidepressant-like effects at 24 h, however the exact timescale of these effects has not been investigated. To determine the short and long-term effects of reelin, female Long Evans rats (n = 120) underwent a chronic corticosterone (CORT; or vehicle) paradigm (40 mg/kg, 21 days). On day 21, rats were treated with reelin (3μg; i.v.), ketamine (10 mg/kg; i.p.), both reelin and ketamine (same doses), or vehicle (saline). Behavioural and biological effects were then evaluated at 1 h, 6 h, 12 h, and 1 week after treatment. The 1-week cohort continued CORT injections to ensure the effect of chronic stress was not lost. Individually, both reelin and ketamine significantly rescued CORT-induced behaviour and hippocampal reelin expression at all timepoints. Ketamine rescued a decrease in dendritic maturity as induced by CORT. Synergistic effects of reelin and ketamine appeared at 1-week, suggesting a potential additive effect of the antidepressant-like actions. Taken together, this study provides further support for reelin-based therapeutics to develop rapid-acting antidepressant.
Collapse
Affiliation(s)
- Kaylene K A Scheil
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | | | - Brady S Reive
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Ciara S Halvorson
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Jennifer Floyd
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Hannah M O Reid
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Jenessa N Johnston
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Lisa E Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C2, Canada.
| |
Collapse
|
3
|
Ramsden CE, Zamora D, Horowitz MS, Jahanipour J, Calzada E, Li X, Keyes GS, Murray HC, Curtis MA, Faull RM, Sedlock A, Maric D. ApoER2-Dab1 disruption as the origin of pTau-associated neurodegeneration in sporadic Alzheimer's disease. Acta Neuropathol Commun 2023; 11:197. [PMID: 38093390 PMCID: PMC10720169 DOI: 10.1186/s40478-023-01693-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/16/2023] [Indexed: 12/17/2023] Open
Abstract
In sporadic Alzheimer's disease (sAD) specific regions, layers and neurons accumulate hyperphosphorylated Tau (pTau) and degenerate early while others remain unaffected even in advanced disease. ApoER2-Dab1 signaling suppresses Tau phosphorylation as part of a four-arm pathway that regulates lipoprotein internalization and the integrity of actin, microtubules, and synapses; however, the role of this pathway in sAD pathogenesis is not fully understood. We previously showed that multiple ApoER2-Dab1 pathway components including ApoE, Reelin, ApoER2, Dab1, pP85αTyr607, pLIMK1Thr508, pTauSer202/Thr205 and pPSD95Thr19 accumulate together within entorhinal-hippocampal terminal zones in sAD, and proposed a unifying hypothesis wherein disruption of this pathway underlies multiple aspects of sAD pathogenesis. However, it is not yet known whether ApoER2-Dab1 disruption can help explain the origin(s) and early progression of pTau pathology in sAD. In the present study, we applied in situ hybridization and immunohistochemistry (IHC) to characterize ApoER2 expression and accumulation of ApoER2-Dab1 pathway components in five regions known to develop early pTau pathology in 64 rapidly autopsied cases spanning the clinicopathological spectrum of sAD. We found that (1) these selectively vulnerable neuron populations strongly express ApoER2; and (2) multiple ApoER2-Dab1 components representing all four arms of this pathway accumulate in abnormal neurons and neuritic plaques in mild cognitive impairment (MCI) and sAD cases and correlate with histological progression and cognitive deficits. Multiplex-IHC revealed that Dab1, pP85αTyr607, pLIMK1Thr508, pTauSer202/Thr205 and pPSD95Thr19 accumulate together within many of the same ApoER2-expressing neurons and in the immediate vicinity of ApoE/ApoJ-enriched extracellular plaques. Collective findings reveal that pTau is only one of many ApoER2-Dab1 pathway components that accumulate in multiple neuroanatomical sites in the earliest stages of sAD and provide support for the concept that ApoER2-Dab1 disruption drives pTau-associated neurodegeneration in human sAD.
Collapse
Affiliation(s)
- Christopher E Ramsden
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH (NIA/NIH), 251 Bayview Blvd., Baltimore, MD, 21224, USA.
- Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA.
| | - Daisy Zamora
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH (NIA/NIH), 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Mark S Horowitz
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH (NIA/NIH), 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Jahandar Jahanipour
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| | - Elizabeth Calzada
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH (NIA/NIH), 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Xiufeng Li
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH (NIA/NIH), 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Gregory S Keyes
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH (NIA/NIH), 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Helen C Murray
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Private Bag, Auckland, 92019, New Zealand
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| | - Maurice A Curtis
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Private Bag, Auckland, 92019, New Zealand
| | - Richard M Faull
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Private Bag, Auckland, 92019, New Zealand
| | - Andrea Sedlock
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| |
Collapse
|
4
|
Tsang CK, Mi Q, Su G, Hwa Lee G, Xie X, D'Arcangelo G, Huang L, Steven Zheng XF. Maf1 is an intrinsic suppressor against spontaneous neural repair and functional recovery after ischemic stroke. J Adv Res 2023; 51:73-90. [PMID: 36402285 PMCID: PMC10491990 DOI: 10.1016/j.jare.2022.11.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/28/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION Spontaneous recovery after CNS injury is often very limited and incomplete, leaving most stroke patients with permanent disability. Maf1 is known as a key growth suppressor in proliferating cells. However, its role in neuronal cells after stroke remains unclear. OBJECTIVE We aimed to investigate the mechanistic role of Maf1 in spontaneous neural repair and evaluated the therapeutic effect of targeting Maf1 on stroke recovery. METHODS We used mouse primary neurons to determine the signaling mechanism of Maf1, and the cleavage-under-targets-and-tagmentation-sequencing to map the whole-genome promoter binding sites of Maf1 in isolated mature cortical neurons. Photothrombotic stroke model was used to determine the therapeutic effect on neural repair and functional recovery by AAV-mediated Maf1 knockdown. RESULTS We found that Maf1 mediates mTOR signaling to regulate RNA polymerase III (Pol III)-dependent rRNA and tRNA transcription in mouse cortical neurons. mTOR regulates neuronal Maf1 phosphorylation and subcellular localization. Maf1 knockdown significantly increases Pol III transcription, neurite outgrowth and dendritic spine formation in neurons. Conversely, Maf1 overexpression suppresses such activities. In response to photothrombotic stroke in mice, Maf1 expression is increased and accumulates in the nucleus of neurons in the peripheral region of infarcted cortex, which is the key region for neural remodeling and repair during spontaneous recovery. Intriguingly, Maf1 knockdown in the peri-infarct cortex significantly enhances neural plasticity and functional recovery. Mechanistically, Maf1 not only interacts with the promoters and represses Pol III-transcribed genes, but also those of CREB-associated genes that are critical for promoting plasticity during neurodevelopment and neural repair. CONCLUSION These findings indicate Maf1 as an intrinsic neural repair suppressor against regenerative capability of mature CNS neurons, and suggest that Maf1 is a potential therapeutic target for enhancing functional recovery after ischemic stroke and other CNS injuries.
Collapse
Affiliation(s)
- Chi Kwan Tsang
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA.
| | - Qiongjie Mi
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Department of Neurology, The First Clinical Medical School of Jinan University, Guangzhou, China
| | - Guangpu Su
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Department of Neurology, The First Clinical Medical School of Jinan University, Guangzhou, China
| | - Gum Hwa Lee
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Xuemin Xie
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Department of Neurology, The First Clinical Medical School of Jinan University, Guangzhou, China
| | - Gabriella D'Arcangelo
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | - Li'an Huang
- Clinical Neuroscience Institute, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China; Department of Neurology, The First Clinical Medical School of Jinan University, Guangzhou, China; Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University Guangzhou, Guangdong, China.
| | - X F Steven Zheng
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA.
| |
Collapse
|
5
|
Johnston JN, Allen J, Shkolnikov I, Sanchez-Lafuente CL, Reive BS, Scheil K, Liang S, Christie BR, Kalynchuk LE, Caruncho HJ. Reelin Rescues Behavioral, Electrophysiological, and Molecular Metrics of a Chronic Stress Phenotype in a Similar Manner to Ketamine. eNeuro 2023; 10:ENEURO.0106-23.2023. [PMID: 37550058 PMCID: PMC10431216 DOI: 10.1523/eneuro.0106-23.2023] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/24/2023] [Accepted: 06/20/2023] [Indexed: 08/09/2023] Open
Abstract
Over the past decade, ketamine, an NMDA receptor antagonist, has demonstrated fast-acting antidepressant effects previously unseen with monoaminergic-based therapeutics. Concerns regarding psychotomimetic effects limit the use of ketamine for certain patient populations. Reelin, an extracellular matrix glycoprotein, has shown promise as a putative fast-acting antidepressant in a model of chronic stress. However, research has not yet demonstrated the changes that occur rapidly after peripheral reelin administration. To address this key gap in knowledge, male Long-Evans rats underwent a chronic corticosterone (CORT; or vehicle) paradigm (40 mg/kg, 21 d). On day 21, rats were then administered an acute dose of ketamine (10 mg/kg, i.p.), reelin (3 µg, i.v.), or vehicle. Twenty-four hours after administration, rats underwent behavioral or in vivo electrophysiological testing before killing. Immunohistochemistry was used to confirm changes in hippocampal reelin immunoreactivity. Lastly, the hippocampus was microdissected from fresh tissue to ascertain whole cell and synaptic-specific changes in protein expression through Western blotting. Chronic corticosterone induced a chronic stress phenotype in the forced swim test and sucrose preference test (SPT). Both reelin and ketamine rescued immobility and swimming, however reelin alone rescued latency to immobility. In vivo electrophysiology revealed decreases in hippocampal long-term potentiation (LTP) after chronic stress which was increased significantly by both ketamine and reelin. Reelin immunoreactivity in the dentate gyrus paralleled the behavioral and electrophysiological findings, but no significant changes were observed in synaptic-level protein expression. This exploratory research supports the putative rapid-acting antidepressant effects of an acute dose of reelin across behavioral, electrophysiological, and molecular measures.
Collapse
Affiliation(s)
- Jenessa N Johnston
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Josh Allen
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Irene Shkolnikov
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Carla L Sanchez-Lafuente
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Brady S Reive
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Kaylene Scheil
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Stanley Liang
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Brian R Christie
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Lisa E Kalynchuk
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| | - Hector J Caruncho
- Division of Medical Sciences, University of Victoria, Victoria, British Columbia V8P 5C2, Canada
| |
Collapse
|
6
|
Ramsden CE, Zamora D, Horowitz M, Jahanipour J, Keyes G, Li X, Murray HC, Curtis MA, Faull RM, Sedlock A, Maric D. ApoER2-Dab1 disruption as the origin of pTau-related neurodegeneration in sporadic Alzheimer's disease. RESEARCH SQUARE 2023:rs.3.rs-2968020. [PMID: 37461602 PMCID: PMC10350181 DOI: 10.21203/rs.3.rs-2968020/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
BACKGROUND Sporadic Alzheimer's disease (sAD) is not a global brain disease. Specific regions, layers and neurons degenerate early while others remain untouched even in advanced disease. The prevailing model used to explain this selective neurodegeneration-prion-like Tau spread-has key limitations and is not easily integrated with other defining sAD features. Instead, we propose that in humans Tau hyperphosphorylation occurs locally via disruption in ApoER2-Dab1 signaling and thus the presence of ApoER2 in neuronal membranes confers vulnerability to degeneration. Further, we propose that disruption of the Reelin/ApoE/ApoJ-ApoER2-Dab1-P85α-LIMK1-Tau-PSD95 (RAAAD-P-LTP) pathway induces deficits in memory and cognition by impeding neuronal lipoprotein internalization and destabilizing actin, microtubules, and synapses. This new model is based in part on our recent finding that ApoER2-Dab1 disruption is evident in entorhinal-hippocampal terminal zones in sAD. Here, we hypothesized that neurons that degenerate in the earliest stages of sAD (1) strongly express ApoER2 and (2) show evidence of ApoER2-Dab1 disruption through co-accumulation of multiple RAAAD-P-LTP components. METHODS We applied in situ hybridization and immunohistochemistry to characterize ApoER2 expression and accumulation of RAAAD-P-LTP components in five regions that are prone to early pTau pathology in 64 rapidly autopsied cases spanning the clinicopathological spectrum of sAD. RESULTS We found that: (1) selectively vulnerable neuron populations strongly express ApoER2; (2) numerous RAAAD-P-LTP pathway components accumulate in neuritic plaques and abnormal neurons; and (3) RAAAD-P-LTP components were higher in MCI and sAD cases and correlated with histological progression and cognitive deficits. Multiplex-IHC revealed that Dab1, pP85αTyr607, pLIMK1Thr508, pTau and pPSD95Thr19 accumulated together within dystrophic dendrites and soma of ApoER2-expressing neurons in the vicinity of ApoE/ApoJ-enriched extracellular plaques. These observations provide evidence for molecular derangements that can be traced back to ApoER2-Dab1 disruption, in each of the sampled regions, layers, and neuron populations that are prone to early pTau pathology. CONCLUSION Findings support the RAAAD-P-LTP hypothesis, a unifying model that implicates dendritic ApoER2-Dab1 disruption as the major driver of both pTau accumulation and neurodegeneration in sAD. This model provides a new conceptual framework to explain why specific neurons degenerate and identifies RAAAD-P-LTP pathway components as potential mechanism-based biomarkers and therapeutic targets for sAD.
Collapse
Affiliation(s)
| | - Daisy Zamora
- National Institute on Aging Laboratory of Clinical Investigation
| | - Mark Horowitz
- National Institute on Aging Intramural Research Program
| | | | - Gregory Keyes
- National Institute on Aging Laboratory of Clinical Investigation
| | - Xiufeng Li
- National Institute on Aging Laboratory of Clinical Investigation
| | - Helen C Murray
- The University of Auckland Faculty of Medical and Health Sciences
| | - Maurice A Curtis
- The University of Auckland Faculty of Medical and Health Sciences
| | - Richard M Faull
- The University of Auckland Faculty of Medical and Health Sciences
| | - Andrea Sedlock
- NINDS: National Institute of Neurological Disorders and Stroke
| | - Dragan Maric
- NINDS: National Institute of Neurological Disorders and Stroke
| |
Collapse
|
7
|
Ramsden CE, Zamora D, Horowitz MS, Jahanipour J, Keyes GS, Li X, Murray HC, Curtis MA, Faull RM, Sedlock A, Maric D. ApoER2-Dab1 disruption as the origin of pTau-related neurodegeneration in sporadic Alzheimer's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.19.23290250. [PMID: 37333406 PMCID: PMC10274982 DOI: 10.1101/2023.05.19.23290250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
BACKGROUND Sporadic Alzheimer's disease (sAD) is not a global brain disease. Specific regions, layers and neurons degenerate early while others remain untouched even in advanced disease. The prevailing model used to explain this selective neurodegeneration-prion-like Tau spread-has key limitations and is not easily integrated with other defining sAD features. Instead, we propose that in humans Tau hyperphosphorylation occurs locally via disruption in ApoER2-Dab1 signaling and thus the presence of ApoER2 in neuronal membranes confers vulnerability to degeneration. Further, we propose that disruption of the Reelin/ApoE/ApoJ-ApoER2-Dab1-P85α-LIMK1-Tau-PSD95 (RAAAD-P-LTP) pathway induces deficits in memory and cognition by impeding neuronal lipoprotein internalization and destabilizing actin, microtubules, and synapses. This new model is based in part on our recent finding that ApoER2-Dab1 disruption is evident in entorhinal-hippocampal terminal zones in sAD. Here, we hypothesized that neurons that degenerate in the earliest stages of sAD (1) strongly express ApoER2 and (2) show evidence of ApoER2-Dab1 disruption through co-accumulation of multiple RAAAD-P-LTP components. METHODS We applied in situ hybridization and immunohistochemistry to characterize ApoER2 expression and accumulation of RAAAD-P-LTP components in five regions that are prone to early pTau pathology in 64 rapidly autopsied cases spanning the clinicopathological spectrum of sAD. RESULTS We found that: (1) selectively vulnerable neuron populations strongly express ApoER2; (2) numerous RAAAD-P-LTP pathway components accumulate in neuritic plaques and abnormal neurons; and (3) RAAAD-P-LTP components were higher in MCI and sAD cases and correlated with histological progression and cognitive deficits. Multiplex-IHC revealed that Dab1, pP85αTyr607, pLIMK1Thr508, pTau and pPSD95Thr19 accumulated together within dystrophic dendrites and soma of ApoER2-expressing neurons in the vicinity of ApoE/ApoJ-enriched extracellular plaques. These observations provide evidence for molecular derangements that can be traced back to ApoER2-Dab1 disruption, in each of the sampled regions, layers, and neuron populations that are prone to early pTau pathology. CONCLUSION Findings support the RAAAD-P-LTP hypothesis, a unifying model that implicates dendritic ApoER2-Dab1 disruption as the major driver of both pTau accumulation and neurodegeneration in sAD. This model provides a new conceptual framework to explain why specific neurons degenerate and identifies RAAAD-P-LTP pathway components as potential mechanism-based biomarkers and therapeutic targets for sAD.
Collapse
Affiliation(s)
- Christopher E. Ramsden
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD, 21224, USA
- Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, 20892, USA
| | - Daisy Zamora
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD, 21224, USA
- Department of Physical Medicine and Rehabilitation, School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Mark S. Horowitz
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Jahandar Jahanipour
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| | - Gregory S. Keyes
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Xiufeng Li
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH 251 Bayview Blvd., Baltimore, MD, 21224, USA
| | - Helen C. Murray
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Private Bag, Auckland, 92019, New Zealand
- Laboratory of Functional and Molecular Imaging, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| | - Maurice A. Curtis
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Private Bag, Auckland, 92019, New Zealand
| | - Richard M. Faull
- Department of Anatomy and Medical Imaging and Centre for Brain Research, Faculty of Medical and Health Science, University of Auckland, Private Bag, Auckland, 92019, New Zealand
| | - Andrea Sedlock
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, 20892, USA
| |
Collapse
|
8
|
Woods RM, Lorusso JM, Harris I, Kowash HM, Murgatroyd C, Neill JC, Glazier JD, Harte M, Hager R. Maternal Immune Activation Induces Adolescent Cognitive Deficits Preceded by Developmental Perturbations in Cortical Reelin Signalling. Biomolecules 2023; 13:biom13030489. [PMID: 36979424 PMCID: PMC10046789 DOI: 10.3390/biom13030489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/02/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Exposure to maternal immune activation (MIA) in utero significantly elevates the risk of developing schizophrenia and other neurodevelopmental disorders. To understand the biological mechanisms underlying the link between MIA and increased risk, preclinical animal models have focussed on specific signalling pathways in the brain that mediate symptoms associated with neurodevelopmental disorders such as cognitive dysfunction. Reelin signalling in multiple brain regions is involved in neuronal migration, synaptic plasticity and long-term potentiation, and has been implicated in cognitive deficits. However, how regulation of Reelin expression is affected by MIA across cortical development and associated cognitive functions remains largely unclear. Using a MIA rat model, here we demonstrate cognitive deficits in adolescent object-location memory in MIA offspring and reductions in Reln expression prenatally and in the adult prefrontal cortex. Further, developmental disturbances in gene/protein expression and DNA methylation of downstream signalling components occurred subsequent to MIA-induced Reelin dysregulation and prior to cognitive deficits. We propose that MIA-induced dysregulation of Reelin signalling contributes to the emergence of prefrontal cortex-mediated cognitive deficits through altered NMDA receptor function, resulting in inefficient long-term potentiation. Our data suggest a developmental window during which attenuation of Reelin signalling may provide a possible therapeutic target.
Collapse
Affiliation(s)
- Rebecca M. Woods
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M139PL, UK
- Correspondence: (R.M.W.); (J.M.L.)
| | - Jarred M. Lorusso
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M139PL, UK
- Correspondence: (R.M.W.); (J.M.L.)
| | - Isabella Harris
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M139PL, UK
| | - Hager M. Kowash
- Maternal and Fetal Health Research Centre, School of Medical Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M139PL, UK
| | | | - Joanna C. Neill
- Division of Pharmacy & Optometry, School of Health Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M139PL, UK
| | - Jocelyn D. Glazier
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M139PL, UK
| | - Michael Harte
- Division of Pharmacy & Optometry, School of Health Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M139PL, UK
| | - Reinmar Hager
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Manchester Academic Health Science Centre, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M139PL, UK
| |
Collapse
|
9
|
Lee HJ, Park JH, Trotter JH, Maher JN, Keenoy KE, Jang YM, Lee Y, Kim JI, Weeber EJ, Hoe HS. Reelin and APP Cooperatively Modulate Dendritic Spine Formation In Vitro and In Vivo. Exp Neurobiol 2023; 32:42-55. [PMID: 36919335 PMCID: PMC10017845 DOI: 10.5607/en22044] [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: 11/15/2022] [Revised: 01/09/2023] [Accepted: 02/08/2023] [Indexed: 03/16/2023] Open
Abstract
Amyloid precursor protein (APP) plays an important role in the pathogenesis of Alzheimer's disease (AD), but the normal function of APP at synapses is poorly understood. We and others have found that APP interacts with Reelin and that each protein is individually important for dendritic spine formation, which is associated with learning and memory, in vitro. However, whether Reelin acts through APP to modulate dendritic spine formation or synaptic function remains unknown. In the present study, we found that Reelin treatment significantly increased dendritic spine density and PSD-95 puncta number in primary hippocampal neurons. An examination of the molecular mechanisms by which Reelin regulates dendritic spinogenesis revealed that Reelin enhanced hippocampal dendritic spine formation in a Ras/ERK/CREB signaling-dependent manner. Interestingly, Reelin did not increase dendritic spine number in primary hippocampal neurons when APP expression was reduced or in vivo in APP knockout (KO) mice. Taken together, our data are the first to demonstrate that Reelin acts cooperatively with APP to modulate dendritic spine formation and suggest that normal APP function is critical for Reelin-mediated dendritic spinogenesis at synapses.
Collapse
Affiliation(s)
- Hyun-Ju Lee
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| | - Jin-Hee Park
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu 41062, Korea.,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea
| | - Justin H Trotter
- Department of Molecular Pharmacology and Physiology, USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA
| | - James N Maher
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Kathleen E Keenoy
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| | - You Mi Jang
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu 41062, Korea
| | - Youngeun Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Jae-Ick Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Edwin J Weeber
- Department of Molecular Pharmacology and Physiology, USF Health Byrd Alzheimer's Institute, University of South Florida, Tampa, FL 33613, USA
| | - Hyang-Sook Hoe
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu 41062, Korea.,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea.,Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
10
|
Reive B, Johnston JN, Sánchez-Lafuente CL, Zhang L, Chang A, Zhang J, Allen J, Romay-Tallon R, Kalynchuk LE, Caruncho HJ. Intravenous Reelin Treatment Rescues Atrophy of Spleen White Pulp and Correlates to Rescue of Forced Swim Test Immobility and Neurochemical Alterations Induced by Chronic Stress. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2023; 7:24705470231164920. [PMID: 36970446 PMCID: PMC10034288 DOI: 10.1177/24705470231164920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 03/06/2023] [Indexed: 03/24/2023]
Abstract
Reelin, an extracellular matrix protein with putative antidepressant-like properties, becomes dysregulated by chronic stress. Improvement in cognitive dysfunction and depression-like behavior induced by chronic stress has been reported with both intrahippocampal and intravenous Reelin treatment but the mechanisms responsible are not clear. To determine if treatment with Reelin modifies chronic stress-induced dysfunction in immune organs and whether this relates to behavioral and/or neurochemical outcomes, spleens were collected from both male (n = 62) and female (n = 53) rats treated with daily corticosterone injections for three weeks that received Reelin or vehicle. Reelin was intravenously administered once on the final day of chronic stress, or repeatedly, with weekly treatments throughout chronic stress. Behavior was assessed during the forced swim test and the object-in-place test. Chronic corticosterone caused significant atrophy of the spleen white pulp, but treatment with a single shot of Reelin restored white pulp in both males and females. Repeated Reelin injections also resolved atrophy in females. Correlations were observed between recovery of white pulp atrophy and recovery of behavioral deficits and expression of both Reelin and glutamate receptor 1 in the hippocampus, supporting a role of the peripheral immune system in the recovery of chronic stress-induced behaviors following treatment with Reelin. Our data adds to research indicating Reelin could be a valuable therapeutic target for chronic stress-related disorders including major depression.
Collapse
Affiliation(s)
- B.S. Reive
- Division of Medical Sciences, University of
Victoria, Victoria, Canada
| | | | | | - Lucy Zhang
- Mount Douglas
Secondary School, Victoria, Canada
| | - Aland Chang
- Mount Douglas
Secondary School, Victoria, Canada
| | | | - Josh Allen
- Division of Medical Sciences, University of
Victoria, Victoria, Canada
| | | | - Lisa E. Kalynchuk
- Division of Medical Sciences, University of
Victoria, Victoria, Canada
| | | |
Collapse
|
11
|
Bracher-Smith M, Leonenko G, Baker E, Crawford K, Graham AC, Salih DA, Howell BW, Hardy J, Escott-Price V. Whole genome analysis in APOE4 homozygotes identifies the DAB1-RELN pathway in Alzheimer's disease pathogenesis. Neurobiol Aging 2022; 119:67-76. [PMID: 35977442 PMCID: PMC9548409 DOI: 10.1016/j.neurobiolaging.2022.07.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/04/2022] [Accepted: 07/23/2022] [Indexed: 11/20/2022]
Abstract
The APOE-ε4 allele is known to predispose to amyloid deposition and consequently is strongly associated with Alzheimer's disease (AD) risk. There is debate as to whether the APOE gene accounts for all genetic variation of the APOE locus. Another question which remains is whether APOE-ε4 carriers have other genetic factors influencing the progression of amyloid positive individuals to AD. We conducted a genome-wide association study in a sample of 5,390 APOE-ε4 homozygous (ε4ε4) individuals (288 cases and 5102 controls) aged 65 or over in the UK Biobank. We found no significant associations of SNPs in the APOE locus with AD in the sample of ε4ε4 individuals. However, we identified a novel genome-wide significant locus associated to AD, mapping to DAB1 (rs112437613, OR = 2.28, CI = 1.73-3.01, p = 5.4 × 10-9). This identification of DAB1 led us to investigate other components of the DAB1-RELN pathway for association. Analysis of the DAB1-RELN pathway indicated that the pathway itself was associated with AD, therefore suggesting an epistatic interaction between the APOE locus and the DAB1-RELN pathway.
Collapse
Affiliation(s)
- Matthew Bracher-Smith
- Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, UK; Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Ganna Leonenko
- Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Emily Baker
- Dementia Research Institute, Cardiff University, Cardiff, UK
| | - Karen Crawford
- Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, UK
| | | | - Dervis A Salih
- Dementia Research Institute, University College London, UK
| | - Brian W Howell
- Neuroscience and Physiology, State University of New York, Albany, NY, USA
| | - John Hardy
- Dementia Research Institute, University College London, UK.
| | - Valentina Escott-Price
- Division of Psychological Medicine & Clinical Neurosciences, Cardiff University, Cardiff, UK.
| |
Collapse
|
12
|
Ramsden CE, Keyes GS, Calzada E, Horowitz MS, Zamora D, Jahanipour J, Sedlock A, Indig FE, Moaddel R, Kapogiannis D, Maric D. Lipid Peroxidation Induced ApoE Receptor-Ligand Disruption as a Unifying Hypothesis Underlying Sporadic Alzheimer's Disease in Humans. J Alzheimers Dis 2022; 87:1251-1290. [PMID: 35466940 DOI: 10.3233/jad-220071] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Sporadic Alzheimer's disease (sAD) lacks a unifying hypothesis that can account for the lipid peroxidation observed early in the disease, enrichment of ApoE in the core of neuritic plaques, hallmark plaques and tangles, and selective vulnerability of entorhinal-hippocampal structures. OBJECTIVE We hypothesized that 1) high expression of ApoER2 (receptor for ApoE and Reelin) helps explain this anatomical vulnerability; 2) lipid peroxidation of ApoE and ApoER2 contributes to sAD pathogenesis, by disrupting neuronal ApoE delivery and Reelin-ApoER2-Dab1 signaling cascades. METHODS In vitro biochemical experiments; Single-marker and multiplex fluorescence-immunohistochemistry (IHC) in postmortem specimens from 26 individuals who died cognitively normal, with mild cognitive impairment or with sAD. RESULTS ApoE and ApoER2 peptides and proteins were susceptible to attack by reactive lipid aldehydes, generating lipid-protein adducts and crosslinked ApoE-ApoER2 complexes. Using in situ hybridization alongside IHC, we observed that: 1) ApoER2 is strongly expressed in terminal zones of the entorhinal-hippocampal 'perforant path' projections that underlie memory; 2) ApoE, lipid aldehyde-modified ApoE, Reelin, ApoER2, and the downstream Reelin-ApoER2 cascade components Dab1 and Thr19-phosphorylated PSD95 accumulated in the vicinity of neuritic plaques in perforant path terminal zones in sAD cases; 3) several ApoE/Reelin-ApoER2-Dab1 pathway markers were higher in sAD cases and positively correlated with histological progression and cognitive deficits. CONCLUSION Results demonstrate derangements in multiple ApoE/Reelin-ApoER2-Dab1 axis components in perforant path terminal zones in sAD and provide proof-of-concept that ApoE and ApoER2 are vulnerable to aldehyde-induced adduction and crosslinking. Findings provide the foundation for a unifying hypothesis implicating lipid peroxidation of ApoE and ApoE receptors in sAD.
Collapse
Affiliation(s)
- Christopher E Ramsden
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, USA.,Intramural Program of the National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA
| | - Gregory S Keyes
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Elizabeth Calzada
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Mark S Horowitz
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Daisy Zamora
- Lipid Peroxidation Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Jahandar Jahanipour
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Andrea Sedlock
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Fred E Indig
- Confocal Imaging Facility, National Institute on Aging Intramural Research Program, NIH, Baltimore, MD, USA
| | - Ruin Moaddel
- Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Dimitrios Kapogiannis
- Human Neuroscience Unit, Laboratory of Clinical Investigation, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Dragan Maric
- Flow and Imaging Cytometry Core Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| |
Collapse
|
13
|
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: 7] [Impact Index Per Article: 2.3] [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
|
14
|
Durakoglugil MS, Wasser CR, Wong CH, Pohlkamp T, Xian X, Lane-Donovan C, Fritschle K, Naestle L, Herz J. Reelin Regulates Neuronal Excitability through Striatal-Enriched Protein Tyrosine Phosphatase (STEP 61) and Calcium Permeable AMPARs in an NMDAR-Dependent Manner. J Neurosci 2021; 41:7340-7349. [PMID: 34290083 PMCID: PMC8412985 DOI: 10.1523/jneurosci.0388-21.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 06/13/2021] [Accepted: 07/08/2021] [Indexed: 11/21/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease marked by the accumulation of amyloid-β (Aβ) plaques and neurofibrillary tangles. Aβ oligomers cause synaptic dysfunction early in AD by enhancing long-term depression (LTD; a paradigm for forgetfulness) via metabotropic glutamate receptor (mGluR)-dependent regulation of striatal-enriched tyrosine phosphatase (STEP61). Reelin is a neuromodulator that signals through ApoE (apolipoprotein E) receptors to protect the synapse against Aβ toxicity (Durakoglugil et al., 2009) Reelin signaling is impaired by ApoE4, the most important genetic risk factor for AD, and Aβ-oligomers activate metabotropic glutamate receptors (Renner et al., 2010). We therefore asked whether Reelin might also affect mGluR-LTD. To this end, we induced chemical mGluR-LTD using DHPG (Dihydroxyphenylglycine), a selective mGluR5 agonist. We found that exogenous Reelin reduces the DHPG-induced increase in STEP61, prevents the dephosphorylation of GluA2, and concomitantly blocks mGluR-mediated LTD. By contrast, Reelin deficiency increased expression of Ca2+-permeable GluA2-lacking AMPA receptors along with higher STEP61 levels, resulting in occlusion of DHPG-induced LTD in hippocampal CA1 neurons. We propose a model in which Reelin modulates local protein synthesis as well as AMPA receptor subunit composition through modulation of mGluR-mediated signaling with implications for memory consolidation or neurodegeneration.SIGNIFICANCE STATEMENT Reelin is an important neuromodulator, which in the adult brain controls synaptic plasticity and protects against neurodegeneration. Amyloid-β has been shown to use mGluRs to induce synaptic depression through endocytosis of NMDA and AMPA receptors, a mechanism referred to as LTD, a paradigm of forgetfulness. Our results show that Reelin regulates the phosphatase STEP, which plays an important role in neurodegeneration, as well as the expression of calcium-permeable AMPA receptors, which play a role in memory formation. These data suggest that Reelin uses mGluR LTD pathways to regulate memory formation as well as neurodegeneration.
Collapse
Affiliation(s)
- Murat S Durakoglugil
- Department of Molecular Genetics
- Center for Translational Neurodegeneration Research
| | - Catherine R Wasser
- Department of Molecular Genetics
- Center for Translational Neurodegeneration Research
| | - Connie H Wong
- Department of Molecular Genetics
- Center for Translational Neurodegeneration Research
| | - Theresa Pohlkamp
- Department of Molecular Genetics
- Center for Translational Neurodegeneration Research
| | - Xunde Xian
- Institute of Cardiovascular Sciences and Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Peking University, Beijing 100871, China
| | - Courtney Lane-Donovan
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, California 94158
| | | | - Lea Naestle
- Ludwig-Maximilians University of Munich, 80539, Munich, Germany
| | - Joachim Herz
- Department of Molecular Genetics
- Center for Translational Neurodegeneration Research
- Departments of Neuroscience and
- Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
15
|
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
|
16
|
Lee SE, Lee GH. Reelin Affects Signaling Pathways of a Group of Inhibitory Neurons and the Development of Inhibitory Synapses in Primary Neurons. Int J Mol Sci 2021; 22:ijms22147510. [PMID: 34299127 PMCID: PMC8305533 DOI: 10.3390/ijms22147510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/26/2021] [Accepted: 07/10/2021] [Indexed: 11/16/2022] Open
Abstract
Reelin is a secretory protein involved in a variety of processes in forebrain development and function, including neuronal migration, dendrite growth, spine formation, and synaptic plasticity. Most of the function of Reelin is focused on excitatory neurons; however, little is known about its effects on inhibitory neurons and inhibitory synapses. In this study, we investigated the phosphatidylinositol 3-kinase/Akt pathway of Reelin in primary cortical and hippocampal neurons. Individual neurons were visualized using immunofluorescence to distinguish inhibitory neurons from excitatory neurons. Reelin-rich protein supplementation significantly induced the phosphorylation of Akt and ribosomal S6 protein in excitatory neurons, but not in most inhibitory neurons. In somatostatin-expressing inhibitory neurons, one of major subtypes of inhibitory neurons, Reelin-rich protein supplementation induced the phosphorylation of S6. Subsequently, we investigated whether or not Reelin-rich protein supplementation affected dendrite development in cultured inhibitory neurons. Reelin-rich protein supplementation did not change the total length of dendrites in inhibitory neurons in vitro. Finally, we examined the development of inhibitory synapses in primary hippocampal neurons and found that Reelin-rich protein supplementation significantly reduced the density of gephyrin-VGAT-positive clusters in the dendritic regions without changing the expression levels of several inhibitory synapse-related proteins. These findings indicate a new role for Reelin in specific groups of inhibitory neurons and the development of inhibitory synapses, which may contribute to the underlying cellular mechanisms of RELN-associated neurological disorders.
Collapse
|
17
|
Melrose J, Hayes AJ, Bix G. The CNS/PNS Extracellular Matrix Provides Instructive Guidance Cues to Neural Cells and Neuroregulatory Proteins in Neural Development and Repair. Int J Mol Sci 2021; 22:5583. [PMID: 34070424 PMCID: PMC8197505 DOI: 10.3390/ijms22115583] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The extracellular matrix of the PNS/CNS is unusual in that it is dominated by glycosaminoglycans, especially hyaluronan, whose space filling and hydrating properties make essential contributions to the functional properties of this tissue. Hyaluronan has a relatively simple structure but its space-filling properties ensure micro-compartments are maintained in the brain ultrastructure, ensuring ionic niches and gradients are maintained for optimal cellular function. Hyaluronan has cell-instructive, anti-inflammatory properties and forms macro-molecular aggregates with the lectican CS-proteoglycans, forming dense protective perineuronal net structures that provide neural and synaptic plasticity and support cognitive learning. AIMS To highlight the central nervous system/peripheral nervous system (CNS/PNS) and its diverse extracellular and cell-associated proteoglycans that have cell-instructive properties regulating neural repair processes and functional recovery through interactions with cell adhesive molecules, receptors and neuroregulatory proteins. Despite a general lack of stabilising fibrillar collagenous and elastic structures in the CNS/PNS, a sophisticated dynamic extracellular matrix is nevertheless important in tissue form and function. CONCLUSIONS This review provides examples of the sophistication of the CNS/PNS extracellular matrix, showing how it maintains homeostasis and regulates neural repair and regeneration.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Sydney Medical School, Northern, The University of Sydney, Sydney, NSW 2052, Australia
- Faculty of Medicine and Health, The University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| | - Anthony J. Hayes
- Bioimaging Research Hub, Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK;
| | - Gregory Bix
- Clinical Neuroscience Research Center, Departments of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| |
Collapse
|
18
|
Hafner G, Guy J, Witte M, Truschow P, Rüppel A, Sirmpilatze N, Dadarwal R, Boretius S, Staiger JF. Increased Callosal Connectivity in Reeler Mice Revealed by Brain-Wide Input Mapping of VIP Neurons in Barrel Cortex. Cereb Cortex 2021; 31:1427-1443. [PMID: 33135045 PMCID: PMC7869096 DOI: 10.1093/cercor/bhaa280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 08/25/2020] [Accepted: 08/25/2020] [Indexed: 01/22/2023] Open
Abstract
The neocortex is composed of layers. Whether layers constitute an essential framework for the formation of functional circuits is not well understood. We investigated the brain-wide input connectivity of vasoactive intestinal polypeptide (VIP) expressing neurons in the reeler mouse. This mutant is characterized by a migration deficit of cortical neurons so that no layers are formed. Still, neurons retain their properties and reeler mice show little cognitive impairment. We focused on VIP neurons because they are known to receive strong long-range inputs and have a typical laminar bias toward upper layers. In reeler, these neurons are more dispersed across the cortex. We mapped the brain-wide inputs of VIP neurons in barrel cortex of wild-type and reeler mice with rabies virus tracing. Innervation by subcortical inputs was not altered in reeler, in contrast to the cortical circuitry. Numbers of long-range ipsilateral cortical inputs were reduced in reeler, while contralateral inputs were strongly increased. Reeler mice had more callosal projection neurons. Hence, the corpus callosum was larger in reeler as shown by structural imaging. We argue that, in the absence of cortical layers, circuits with subcortical structures are maintained but cortical neurons establish a different network that largely preserves cognitive functions.
Collapse
Affiliation(s)
- Georg Hafner
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Göttingen, 37075 Göttingen, Germany
| | - Julien Guy
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Göttingen, 37075 Göttingen, Germany
| | - Mirko Witte
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Göttingen, 37075 Göttingen, Germany
| | - Pavel Truschow
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Göttingen, 37075 Göttingen, Germany
| | - Alina Rüppel
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Göttingen, 37075 Göttingen, Germany
| | - Nikoloz Sirmpilatze
- Functional Imaging Laboratory, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Rakshit Dadarwal
- Functional Imaging Laboratory, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Susann Boretius
- Functional Imaging Laboratory, German Primate Center, Leibniz Institute for Primate Research, 37077 Göttingen, Germany
| | - Jochen F Staiger
- Institute for Neuroanatomy, University Medical Center, Georg-August-University Göttingen, 37075 Göttingen, Germany
| |
Collapse
|
19
|
Yvone GM, Chavez-Martinez CL, Nguyen AR, Wang DJ, Phelps PE. Reelin dorsal horn neurons co-express Lmx1b and are mispositioned in disabled-1 mutant mice. Eur J Neurosci 2020; 52:3322-3338. [PMID: 32492253 PMCID: PMC9451954 DOI: 10.1111/ejn.14847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 11/30/2022]
Abstract
Mice missing either Reelin or Disabled-1 (Dab1) exhibit dorsal horn neuronal positioning errors and display heat hypersensitivity and mechanical insensitivity. Reelin binds its receptors, apolipoprotein E receptor 2 and very low-density lipoprotein receptor, leading to the recruitment and phosphorylation of Dab1 and activation of downstream pathways that regulate neuronal migration. Previously, we reported that 70% of Dab1 laminae I-II neurons co-expressed LIM-homeobox transcription factor 1-beta (Lmx1b). Here, we asked whether Reelin-expressing dorsal horn neurons co-express Lmx1b, are mispositioned in dab1 mutants, and contribute to nociceptive abnormalities. About 90% of Reelin-labeled neurons are Lmx1b-positive in laminae I-II, confirming that most Reelin and Dab1 neurons are glutamatergic. We determined that Reelin-Lmx1b and Dab1-Lmx1b dorsal horn neurons are separate populations, and together, comprise 37% of Lmx1b-positive cells within and above the Isolectin B4 (IB4) layer in wild-type mice. Compared to wild-type mice, dab1 mutants have a reduced area of laminae I-II outer (above the IB4 layer), more Reelin-Lmx1b neurons within the IB4 layer, and fewer Reelin-Lmx1b neurons within the lateral reticulated area of lamina V and lateral spinal nucleus. Interestingly, both Reelin- and Dab1-labeled dorsal horn neurons sustain similar positioning errors in mutant mice. After noxious thermal and mechanical stimulation, Reelin, Lmx1b, and Reelin-Lmx1b neurons expressed Fos in laminae I-II and the lateral reticulated area in wild-type mice and, therefore, participate in nociceptive circuits. Together, our data suggest that disruption of the Reelin-signaling pathway results in neuroanatomical abnormalities that contribute to the nociceptive changes that characterize these mutant mice.
Collapse
Affiliation(s)
- Griselda M Yvone
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA, USA
| | | | - Amanda R Nguyen
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA, USA
| | - Deborah J Wang
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA, USA
| | - Patricia E Phelps
- Department of Integrative Biology and Physiology, UCLA, Los Angeles, CA, USA
| |
Collapse
|
20
|
Dal Pozzo V, Crowell B, Briski N, Crockett DP, D’Arcangelo G. Reduced Reelin Expression in the Hippocampus after Traumatic Brain Injury. Biomolecules 2020; 10:biom10070975. [PMID: 32610618 PMCID: PMC7407987 DOI: 10.3390/biom10070975] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 01/06/2023] Open
Abstract
Traumatic brain injury (TBI) is a relatively common occurrence following accidents or violence, and often results in long-term cognitive or motor disability. Despite the high health cost associated with this type of injury, presently there are no effective treatments for many neurological symptoms resulting from TBI. This is due in part to our limited understanding of the mechanisms underlying brain dysfunction after injury. In this study, we used the mouse controlled cortical impact (CCI) model to investigate the effects of TBI, and focused on Reelin, an extracellular protein that critically regulates brain development and modulates synaptic activity in the adult brain. We found that Reelin expression decreases in forebrain regions after TBI, and that the number of Reelin-expressing cells decrease specifically in the hippocampus, an area of the brain that plays an important role in learning and memory. We also conducted in vitro experiments using mouse neuronal cultures and discovered that Reelin protects hippocampal neuronal cells from glutamate-induced neurotoxicity, a well-known secondary effect of TBI. Together our findings suggest that the loss of Reelin expression may contribute to neuronal death in the hippocampus after TBI, and raise the possibility that increasing Reelin levels or signaling activity may promote functional recovery.
Collapse
Affiliation(s)
- Valentina Dal Pozzo
- Graduate Program in Neuroscience, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA;
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA; (B.C.); (N.B.)
| | - Beth Crowell
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA; (B.C.); (N.B.)
| | - Nicholas Briski
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA; (B.C.); (N.B.)
| | - David P. Crockett
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA;
| | - Gabriella D’Arcangelo
- Department of Cell Biology and Neuroscience, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA; (B.C.); (N.B.)
- Correspondence:
| |
Collapse
|
21
|
Jossin Y. Reelin Functions, Mechanisms of Action and Signaling Pathways During Brain Development and Maturation. Biomolecules 2020; 10:biom10060964. [PMID: 32604886 PMCID: PMC7355739 DOI: 10.3390/biom10060964] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 06/24/2020] [Accepted: 06/24/2020] [Indexed: 12/14/2022] Open
Abstract
During embryonic development and adulthood, Reelin exerts several important functions in the brain including the regulation of neuronal migration, dendritic growth and branching, dendritic spine formation, synaptogenesis and synaptic plasticity. As a consequence, the Reelin signaling pathway has been associated with several human brain disorders such as lissencephaly, autism, schizophrenia, bipolar disorder, depression, mental retardation, Alzheimer’s disease and epilepsy. Several elements of the signaling pathway are known. Core components, such as the Reelin receptors very low-density lipoprotein receptor (VLDLR) and Apolipoprotein E receptor 2 (ApoER2), Src family kinases Src and Fyn, and the intracellular adaptor Disabled-1 (Dab1), are common to most but not all Reelin functions. Other downstream effectors are, on the other hand, more specific to defined tasks. Reelin is a large extracellular protein, and some aspects of the signal are regulated by its processing into smaller fragments. Rather than being inhibitory, the processing at two major sites seems to be fulfilling important physiological functions. In this review, I describe the various cellular events regulated by Reelin and attempt to explain the current knowledge on the mechanisms of action. After discussing the shared and distinct elements of the Reelin signaling pathway involved in neuronal migration, dendritic growth, spine development and synaptic plasticity, I briefly outline the data revealing the importance of Reelin in human brain disorders.
Collapse
Affiliation(s)
- Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium
| |
Collapse
|
22
|
Fyn Tyrosine Kinase as Harmonizing Factor in Neuronal Functions and Dysfunctions. Int J Mol Sci 2020; 21:ijms21124444. [PMID: 32580508 PMCID: PMC7352836 DOI: 10.3390/ijms21124444] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/19/2020] [Accepted: 06/20/2020] [Indexed: 12/25/2022] Open
Abstract
Fyn is a non-receptor or cytoplasmatic tyrosine kinase (TK) belonging to the Src family kinases (SFKs) involved in multiple transduction pathways in the central nervous system (CNS) including synaptic transmission, myelination, axon guidance, and oligodendrocyte formation. Almost one hundred years after the original description of Fyn, this protein continues to attract extreme interest because of its multiplicity of actions in the molecular signaling pathways underlying neurodevelopmental as well as neuropathologic events. This review highlights and summarizes the most relevant recent findings pertinent to the role that Fyn exerts in the brain, emphasizing aspects related to neurodevelopment and synaptic plasticity. Fyn is a common factor in healthy and diseased brains that targets different proteins and shapes different transduction signals according to the neurological conditions. We will primarily focus on Fyn-mediated signaling pathways involved in neuronal differentiation and plasticity that have been subjected to considerable attention lately, opening the fascinating scenario to target Fyn TK for the development of potential therapeutic interventions for the treatment of CNS injuries and certain neurodegenerative disorders like Alzheimer’s disease.
Collapse
|
23
|
Wang L, Zhao D, Wang M, Wang Y, Vreugdenhil M, Lin J, Lu C. Modulation of Hippocampal Gamma Oscillations by Dopamine in Heterozygous Reeler Mice in vitro. Front Cell Neurosci 2020; 13:586. [PMID: 32116553 PMCID: PMC7026475 DOI: 10.3389/fncel.2019.00586] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/23/2019] [Indexed: 11/14/2022] Open
Abstract
The reelin haploinsufficient heterozygous reeler mice (HRM), an animal model of schizophrenia, have altered mesolimbic dopaminergic pathways and share similar neurochemical and behavioral properties with patients with schizophrenia. Dysfunctional neural circuitry with impaired gamma (γ) oscillation (30–80 Hz) has been implicated in abnormal cognition in patients with schizophrenia. However, the function of neural circuitry in terms of γ oscillation and its modulation by dopamine (DA) has not been reported in HRM. In this study, first, we recorded γ oscillations in CA3 from wild-type mice (WTM) and HRM hippocampal slices, and we studied the effects of DA on γ oscillations. We found that there was no difference in γ power between WTM and HRM and that DA increased γ power of WTM but not HRM, suggesting that DA modulations of network oscillations in HRM are impaired. Second, we found that N-methyl-D-aspartate receptor (NMDAR) antagonist MK-801 itself increased γ power and occluded DA-mediated enhancement of γ power in WTM but partially restored DA modulation of γ oscillations in HRM. Third, inhibition of phosphatidylinositol 3-kinase (PI3K), a downstream molecule of NMDAR, increased γ power and blocked the effects of DA on γ oscillation in WTM and had no significant effect on γ power but largely restored DA modulation of γ oscillations in HRM. Our results reveal that impaired DA function in HRM is associated with dysregulated NMDAR–PI3K signaling, a mechanism that may be relevant in the pathology of schizophrenia.
Collapse
Affiliation(s)
- Lu Wang
- The International-Joint Lab for Non-Invasive Neural Modulation, Xinxiang Medical University, Xinxiang, China.,Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University, Xinxiang, China.,Department of Neurobiology and Physiology, Xinxiang Medical University, Xinxiang, China
| | - Dandan Zhao
- The International-Joint Lab for Non-Invasive Neural Modulation, Xinxiang Medical University, Xinxiang, China.,Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University, Xinxiang, China.,Department of Neurobiology and Physiology, Xinxiang Medical University, Xinxiang, China
| | - Mengmeng Wang
- The International-Joint Lab for Non-Invasive Neural Modulation, Xinxiang Medical University, Xinxiang, China.,Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University, Xinxiang, China.,Department of Neurobiology and Physiology, Xinxiang Medical University, Xinxiang, China
| | - Yuan Wang
- The International-Joint Lab for Non-Invasive Neural Modulation, Xinxiang Medical University, Xinxiang, China.,Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University, Xinxiang, China
| | - Martin Vreugdenhil
- Department of Life Science, School of Health Sciences, Birmingham City University, Birmingham, United Kingdom
| | - Juntang Lin
- School of Biomedical Engineering, Xinxiang Medical University, Xinxiang, China
| | - Chengbiao Lu
- The International-Joint Lab for Non-Invasive Neural Modulation, Xinxiang Medical University, Xinxiang, China.,Key Laboratory for the Brain Research of Henan Province, Xinxiang Medical University, Xinxiang, China.,Department of Neurobiology and Physiology, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
24
|
Castagna C, Merighi A, Lossi L. Decreased Expression of Synaptophysin 1 (SYP1 Major Synaptic Vesicle Protein p38) and Contactin 6 (CNTN6/NB3) in the Cerebellar Vermis of reln Haplodeficient Mice. Cell Mol Neurobiol 2019; 39:833-856. [PMID: 31098770 PMCID: PMC11462894 DOI: 10.1007/s10571-019-00683-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 05/02/2019] [Indexed: 01/17/2023]
Abstract
Reeler heterozygous mice (reln+/-) are seemingly normal but haplodeficient in reln, a gene implicated in autism. Structural/neurochemical alterations in the reln+/- brain are subtle and difficult to demonstrate. Therefore, the usefulness of these mice in translational research is still debated. As evidence implicated several synapse-related genes in autism and the cerebellar vermis is structurally altered in the condition, we have investigated the expression of synaptophysin 1 (SYP1) and contactin 6 (CNTN6) within the vermis of reln+/- mice. Semi-thin plastic sections of the vermis from adult mice of both sexes and different genotypes (reln+/- and reln+/+) were processed with an indirect immunofluorescence protocol. Immunofluorescence was quantified on binary images and statistically analyzed. Reln+/- males displayed a statistically significant reduction of 11.89% in the expression of SYP1 compared to sex-matched wild-type animals, whereas no differences were observed between reln+/+ and reln+/- females. In reln+/- male mice, reductions were particularly evident in the molecular layer: 10.23% less SYP1 than reln+/+ males and 5.84% < reln+/+ females. In reln+/- females, decrease was 9.84% versus reln+/+ males and 5.43% versus reln+/+ females. Both reln+/- males and females showed a stronger decrease in CNTN6 expression throughout all the three cortical layers of the vermis: 17-23% in the granular layer, 24-26% in the Purkinje cell layer, and 9-14% in the molecular layer. Altogether, decrease of vermian SYP1 and CNTN6 in reln+/- mice displayed patterns compatible with the structural modifications of the autistic cerebellum. Therefore, these mice may be a good model in translational studies.
Collapse
Affiliation(s)
- Claudia Castagna
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095, Turin, Grugliasco (TO), Italy.
| | - Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095, Turin, Grugliasco (TO), Italy
| | - Laura Lossi
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095, Turin, Grugliasco (TO), Italy
| |
Collapse
|
25
|
Anstötz M, Lee SK, Neblett TI, Rune GM, Maccaferri G. Experience-Dependent Regulation of Cajal-Retzius Cell Networks in the Developing and Adult Mouse Hippocampus. ACTA ACUST UNITED AC 2019. [PMID: 28637318 DOI: 10.1093/cercor/bhx153] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In contrast to their near-disappearance in the adult neocortex, Cajal-Retzius cells have been suggested to persist longer in the hippocampus. A distinctive feature of the mature hippocampus, not maintained by other cortical areas, is its ability to sustain adult neurogenesis. Here, we have investigated whether environmental manipulations affecting hippocampal postnatal neurogenesis have a parallel impact on Cajal-Retzius cells. We used multiple mouse reporter lines to unequivocally identify Cajal-Retzius cells and quantify their densities during postnatal development. We found that exposure to an enriched environment increased the persistence of Cajal-Retzius cells in the hippocampus, but not in adjacent cortical regions. We did not observe a similar effect for parvalbumin-expressing interneurons, which suggested the occurrence of a cell type-specific process. In addition, we did not detect obvious changes either in Cajal-Retzius cell electrophysiological or morphological features, when compared with what previously reported in animals not exposed to enriched conditions. However, optogenetically triggered synaptic output of Cajal-Retzius cells onto local interneurons was enhanced, consistent with our observation of higher Cajal-Retzius cell densities. In conclusion, our data reveal a novel form of hippocampal, cell type-specific, experience-dependent network plasticity. We propose that this phenomenon may be involved in the regulation of enrichment-dependent enhanced hippocampal postnatal neurogenesis.
Collapse
Affiliation(s)
- Max Anstötz
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611-3008, USA.,Institute for Neuroanatomy, University/University Hospital Hamburg, 20246 Hamburg, Germany
| | - Sun Kyong Lee
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611-3008, USA
| | - Tamra I Neblett
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611-3008, USA
| | - Gabriele M Rune
- Institute for Neuroanatomy, University/University Hospital Hamburg, 20246 Hamburg, Germany
| | - Gianmaria Maccaferri
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611-3008, USA
| |
Collapse
|
26
|
Juric-Sekhar G, Hevner RF. Malformations of Cerebral Cortex Development: Molecules and Mechanisms. ANNUAL REVIEW OF PATHOLOGY 2019; 14:293-318. [PMID: 30677308 PMCID: PMC6938687 DOI: 10.1146/annurev-pathmechdis-012418-012927] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Malformations of cortical development encompass heterogeneous groups of structural brain anomalies associated with complex neurodevelopmental disorders and diverse genetic and nongenetic etiologies. Recent progress in understanding the genetic basis of brain malformations has been driven by extraordinary advances in DNA sequencing technologies. For example, somatic mosaic mutations that activate mammalian target of rapamycin signaling in cortical progenitor cells during development are now recognized as the cause of hemimegalencephaly and some types of focal cortical dysplasia. In addition, research on brain development has begun to reveal the cellular and molecular bases of cortical gyrification and axon pathway formation, providing better understanding of disorders involving these processes. New neuroimaging techniques with improved resolution have enhanced our ability to characterize subtle malformations, such as those associated with intellectual disability and autism. In this review, we broadly discuss cortical malformations and focus on several for which genetic etiologies have elucidated pathogenesis.
Collapse
Affiliation(s)
- Gordana Juric-Sekhar
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington 98195, USA; ,
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | - Robert F Hevner
- Department of Pathology, University of Washington School of Medicine, Seattle, Washington 98195, USA; ,
- Department of Neurological Surgery, University of Washington School of Medicine, Seattle, Washington 98195, USA
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, Washington 98105, USA
- Current affiliation: Department of Pathology, University of California, San Diego, California 92093, USA
| |
Collapse
|
27
|
Prume M, Rollenhagen A, Lübke JHR. Structural and Synaptic Organization of the Adult Reeler Mouse Somatosensory Neocortex: A Comparative Fine-Scale Electron Microscopic Study of Reeler With Wild Type Mice. Front Neuroanat 2018; 12:80. [PMID: 30344480 PMCID: PMC6182073 DOI: 10.3389/fnana.2018.00080] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 09/13/2018] [Indexed: 11/17/2022] Open
Abstract
The reeler mouse has been widely used to study various aspects of cortico- and synaptogenesis, but also as a model for several neurological and neurodegenerative disorders. In contrast to development, comparably little is known about the neuronal composition and synaptic organization of the adult reeler mouse neocortex, in particular at the fine-scale electron microscopic level, which was investigated here and compared with wild type (WT) mice. In this study, the “barrel field” of the adult reeler and WT mouse somatosensory neocortex is used as a model system. In reeler the characteristic six-layered structure is no longer existent, but replaced by a conglomerate of neurons organized in homologous clusters with maintained morphological identity and heterologous clusters between neurons and/or oligodendrocytes. These clusters are loosely scattered throughout the neocortical mass between the pial surface and the white matter. In contrast to WT, layer 1 (L1), if existent, seems to be diluted into the volume of the neocortical mass with no clear boundary. L1 also contains clusters of migrated or persistent neurons, oligodendro- and astrocytes. As in WT, myelinated and unmyelinated axons were found throughout the neocortical mass, but in reeler they were organized in massive fiber bundles with a high fiber packing density. A prominent and massive thalamocortical projection traverses through the neocortical mass, always accompanied by numerous “active” oligodendrocytes whereas in WT no such projections were found and “silent” oligodendrocytes were restricted to the white matter. In the adult reeler mouse neocortex, synaptic boutons terminate on somata, dendritic shafts, spines of different types and axon initial segments with no signs of structural distortion and/or degeneration, indicating a “normal” postsynaptic innervation pattern of neurons. In addition, synaptic complexes between boutons and their postsynaptic targets are tightly ensheathed by fine astrocytic processes, as in WT. In conclusion, the neuronal clusters may represent a possible alternative organization principle in adult reeler mice “replacing” layer formation. If so, these homologous clusters may represent individual “functional units” where neurons are highly interconnected and may function as the equivalent of neurons integrated in a cortical layer. The structural composition and postsynaptic innervation pattern of neurons by synaptic boutons provide the structural basis for the establishment of a functional although altered cortical network in the adult reeler mouse.
Collapse
Affiliation(s)
- Miriam Prume
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Jülich, Germany
| | - Astrid Rollenhagen
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Jülich, Germany
| | - Joachim H R Lübke
- Institute of Neuroscience and Medicine INM-10, Research Centre Jülich GmbH, Jülich, Germany.,Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH University Hospital Aachen, Aachen, Germany.,JARA Translational Brain Medicine, Jülich, Germany
| |
Collapse
|
28
|
Schroeder A, van den Buuse M, Hill RA. Reelin Haploinsufficiency and Late-Adolescent Corticosterone Treatment Induce Long-Lasting and Female-Specific Molecular Changes in the Dorsal Hippocampus. Brain Sci 2018; 8:brainsci8070118. [PMID: 29941797 PMCID: PMC6070826 DOI: 10.3390/brainsci8070118] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 06/20/2018] [Accepted: 06/22/2018] [Indexed: 12/24/2022] Open
Abstract
Reelin depletion and stress seem to affect similar pathways including GABAergic and glutamatergic signaling and both are implicated in psychiatric disorders in late adolescence/early adulthood. The interaction between reelin depletion and stress, however, remains unclear. To investigate this, male and female heterozygous reelin mice (HRM) and wildtype (WT) controls were treated with the stress hormone, corticosterone (CORT), during late adolescence to simulate chronic stress. Glucocorticoid receptors (GR), N-methyl-d-aspartate receptor (NMDAr) subunits, glutamic acid decarboxylase (GAD67) and parvalbumin (PV) were measured in the hippocampus and the prefrontal cortex (PFC) in adulthood. While no changes were seen in male mice, female HRM showed a significant reduction in GR expression in the dorsal hippocampus. In addition, CORT reduced GR levels as well as GluN2B and GluN2C subunits of NMDAr in the dorsal hippocampus in female mice only. CORT furthermore reduced GluN1 levels in the PFC of female mice. The combined effect of HRM and CORT treatment appeared to be additive in terms of GR expression in the dorsal hippocampus. Female-specific CORT-induced changes were associated with overall higher circulating CORT levels in female compared to male mice. This study shows differential effects of reelin depletion and CORT treatment on GR and NMDAr protein expression in male and female mice, suggesting that females are more susceptible to reelin haploinsufficiency as well as late-adolescent stress. These findings shed more light on female-specific vulnerability to stress and have implications for stress-associated mental illnesses with a female bias including anxiety and major depression.
Collapse
Affiliation(s)
- Anna Schroeder
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Australia.
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton 3168, Australia.
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Bundoora 3086 Australia.
- Department of Pharmacology, University of Melbourne, Parkville 3052, Australia.
- The College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville 4810, Australia.
| | - Rachel A Hill
- The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3052, Australia.
- Department of Psychiatry, School of Clinical Sciences, Monash University, Clayton 3168, Australia.
| |
Collapse
|
29
|
Bosch C, Masachs N, Exposito-Alonso D, Martínez A, Teixeira CM, Fernaud I, Pujadas L, Ulloa F, Comella JX, DeFelipe J, Merchán-Pérez A, Soriano E. Reelin Regulates the Maturation of Dendritic Spines, Synaptogenesis and Glial Ensheathment of Newborn Granule Cells. Cereb Cortex 2018; 26:4282-4298. [PMID: 27624722 PMCID: PMC5066826 DOI: 10.1093/cercor/bhw216] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 06/20/2016] [Indexed: 02/02/2023] Open
Abstract
Significance Statement The extracellular protein Reelin has an important role in neurological diseases, including epilepsy, Alzheimer's disease and psychiatric diseases, targeting hippocampal circuits. Here we address the role of Reelin in the development of synaptic contacts in adult-generated granule cells (GCs), a neuronal population that is crucial for learning and memory and implicated in neurological and psychiatric diseases. We found that the Reelin pathway controls the shapes, sizes, and types of dendritic spines, the complexity of multisynaptic innervations and the degree of the perisynaptic astroglial ensheathment that controls synaptic homeostasis. These findings show a pivotal role of Reelin in GC synaptogenesis and provide a foundation for structural circuit alterations caused by Reelin deregulation that may occur in neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Carles Bosch
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.,Institut de Recerca de l'Hospital Universitari de la Vall d'Hebron (VHIR), Barcelona 08023, Spain
| | - Nuria Masachs
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain
| | - David Exposito-Alonso
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
| | - Albert Martínez
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain
| | - Cátia M Teixeira
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain
| | - Isabel Fernaud
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.,Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus de Montegancedo, Madrid 28223, Spain.,Instituto Cajal (Consejo Superior de Investigaciones Científicas), Madrid 28002, Spain
| | - Lluís Pujadas
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.,Institut de Recerca de l'Hospital Universitari de la Vall d'Hebron (VHIR), Barcelona 08023, Spain
| | - Fausto Ulloa
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain
| | - Joan X Comella
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.,Institut de Recerca de l'Hospital Universitari de la Vall d'Hebron (VHIR), Barcelona 08023, Spain.,Institut de Neurociències, Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Javier DeFelipe
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.,Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus de Montegancedo, Madrid 28223, Spain.,Instituto Cajal (Consejo Superior de Investigaciones Científicas), Madrid 28002, Spain
| | - Angel Merchán-Pérez
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.,Laboratorio Cajal de Circuitos Corticales, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Campus de Montegancedo, Madrid 28223, Spain.,Departamento de Arquitectura y Tecnología de Sistemas Informáticos, Escuela Técnica Superior de Ingenieros Informáticos, Universidad Politécnica de Madrid, Madrid 28660, Spain
| | - Eduardo Soriano
- Developmental Neurobiology and Regeneration Unit, Department of Cell Biology, Parc Científic de Barcelona and Institute of Neurosciences, University of Barcelona, Barcelona 08028, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.,Institut de Recerca de l'Hospital Universitari de la Vall d'Hebron (VHIR), Barcelona 08023, Spain.,Institució Catalana de Recerca i Estudis Avançats Academia, Barcelona 08010, Spain
| |
Collapse
|
30
|
The functions of Reelin in membrane trafficking and cytoskeletal dynamics: implications for neuronal migration, polarization and differentiation. Biochem J 2017; 474:3137-3165. [PMID: 28887403 DOI: 10.1042/bcj20160628] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/27/2017] [Accepted: 08/01/2017] [Indexed: 02/06/2023]
Abstract
Reelin is a large extracellular matrix protein with relevant roles in mammalian central nervous system including neurogenesis, neuronal polarization and migration during development; and synaptic plasticity with its implications in learning and memory, in the adult. Dysfunctions in reelin signaling are associated with brain lamination defects such as lissencephaly, but also with neuropsychiatric diseases like autism, schizophrenia and depression as well with neurodegeneration. Reelin signaling involves a core pathway that activates upon reelin binding to its receptors, particularly ApoER2 (apolipoprotein E receptor 2)/LRP8 (low-density lipoprotein receptor-related protein 8) and very low-density lipoprotein receptor, followed by Src/Fyn-mediated phosphorylation of the adaptor protein Dab1 (Disabled-1). Phosphorylated Dab1 (pDab1) is a hub in the signaling cascade, from which several other downstream pathways diverge reflecting the different roles of reelin. Many of these pathways affect the dynamics of the actin and microtubular cytoskeleton, as well as membrane trafficking through the regulation of the activity of small GTPases, including the Rho and Rap families and molecules involved in cell polarity. The complexity of reelin functions is reflected by the fact that, even now, the precise mode of action of this signaling cascade in vivo at the cellular and molecular levels remains unclear. This review addresses and discusses in detail the participation of reelin in the processes underlying neurogenesis, neuronal migration in the cerebral cortex and the hippocampus; and the polarization, differentiation and maturation processes that neurons experiment in order to be functional in the adult brain. In vivo and in vitro evidence is presented in order to facilitate a better understanding of this fascinating system.
Collapse
|
31
|
Antonioli-Santos R, Lanzillotta-Mattos B, Hedin-Pereira C, Serfaty CA. The fine tuning of retinocollicular topography depends on reelin signaling during early postnatal development of the rat visual system. Neuroscience 2017; 357:264-272. [PMID: 28602919 DOI: 10.1016/j.neuroscience.2017.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Revised: 05/27/2017] [Accepted: 06/01/2017] [Indexed: 10/19/2022]
Abstract
During postnatal development, neural circuits are extremely dynamic and develop precise connection patterns that emerge as a result of the elimination of synaptic terminals, a process instructed by molecular cues and patterns of electrical activity. In the rodent visual system, this process begins during the first postnatal week and proceeds during the second and third postnatal weeks as spontaneous retinal activity and finally use-dependent fine tuning takes place. Reelin is a large extracellular matrix glycoprotein able to affect several steps of brain development, from neuronal migration to the maturation of dendritic spines and use-dependent synaptic development. In the present study, we investigated the role of reelin on the topographical refinement of primary sensory connections studying the development of retinal ganglion cell axon terminals in the rat superior colliculus. We found that reelin levels in the visual layers of the superior colliculus are the highest between the second and third postnatal weeks. Blocking reelin signaling with a neutralizing antibody (CR-50) from PND 7 to PND 14 induced a non-specific sprouting of ipsilateral retinocollicular axons outside their typical distribution of discrete patches of axon terminals. Also we found that reelin blockade resulted in reduced levels of phospho-GAP43, increased GluN1 and GluN2B-NMDA subunits and decreased levels of GAD65 content in the visual layers of the superior colliculus. The results suggest that reelin signaling is associated with the maturation of excitatory and inhibitory synaptic machinery influencing the development and fine tuning of topographically organized neural circuits during postnatal development.
Collapse
Affiliation(s)
- Rachel Antonioli-Santos
- Federal Fluminense University, Biology Institute, Neurobiology Department, Laboratory of Neuroplasticity - Niteroi, PO Box: 100180, Brazil; Institute of Biomedical Research, Marcílio Dias Navy Hospital, Rio de Janeiro, Brazil
| | - Bruna Lanzillotta-Mattos
- Federal Fluminense University, Biology Institute, Neurobiology Department, Laboratory of Neuroplasticity - Niteroi, PO Box: 100180, Brazil
| | - Cecília Hedin-Pereira
- Federal University of Rio de Janeiro, Institute of Biomedical Sciences, Laboratory of Cellular Neuroanatomy - Rio de Janeiro, Brazil; Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil
| | - Claudio Alberto Serfaty
- Federal Fluminense University, Biology Institute, Neurobiology Department, Laboratory of Neuroplasticity - Niteroi, PO Box: 100180, Brazil.
| |
Collapse
|
32
|
Lammert DB, Middleton FA, Pan J, Olson EC, Howell BW. The de novo autism spectrum disorder RELN R2290C mutation reduces Reelin secretion and increases protein disulfide isomerase expression. J Neurochem 2017; 142:89-102. [PMID: 28419454 DOI: 10.1111/jnc.14045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 03/22/2017] [Accepted: 04/05/2017] [Indexed: 12/11/2022]
Abstract
Despite the recent identification of over 40 missense heterozygous Reelin gene (RELN) mutations in autism spectrum disorder (ASD), none of these has been functionally characterized. Reelin is an integral signaling ligand for proper brain development and post-natal synapse function - properties likely disrupted in ASD patients. We find that the R2290C mutation, which arose de novo in an affected ASD proband, and other analogous mutations in arginine-amino acid-arginine domains reduce protein secretion. Closer analysis of RELN R2290C heterozygous neurospheres reveals up-regulation of Protein Disulfide Isomerase A1, best known as an endoplasmic reticulum-chaperone protein, which has been linked to neuronal pathology. This effect is recapitulated in a heterozygous RELN mouse mutant that is characterized by defective Reelin secretion. These findings suggest that both a deficiency in Reelin signaling and pathologic impairment of Reelin secretion may contribute to ASD risk.
Collapse
Affiliation(s)
- Dawn B Lammert
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Frank A Middleton
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Jen Pan
- The Broad Institute, Stanley Center Neurobiology, Cambridge, Massachusetts, USA
| | - Eric C Olson
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, USA
| | - Brian W Howell
- Department of Neuroscience and Physiology, SUNY Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
33
|
Pohlkamp T, Wasser CR, Herz J. Functional Roles of the Interaction of APP and Lipoprotein Receptors. Front Mol Neurosci 2017; 10:54. [PMID: 28298885 PMCID: PMC5331069 DOI: 10.3389/fnmol.2017.00054] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 02/16/2017] [Indexed: 11/24/2022] Open
Abstract
The biological fates of the key initiator of Alzheimer’s disease (AD), the amyloid precursor protein (APP), and a family of lipoprotein receptors, the low-density lipoprotein (LDL) receptor-related proteins (LRPs) and their molecular roles in the neurodegenerative disease process are inseparably interwoven. Not only does APP bind tightly to the extracellular domains (ECDs) of several members of the LRP group, their intracellular portions are also connected through scaffolds like the one established by FE65 proteins and through interactions with adaptor proteins such as X11/Mint and Dab1. Moreover, the ECDs of APP and LRPs share common ligands, most notably Reelin, a regulator of neuronal migration during embryonic development and modulator of synaptic transmission in the adult brain, and Agrin, another signaling protein which is essential for the formation and maintenance of the neuromuscular junction (NMJ) and which likely also has critical, though at this time less well defined, roles for the regulation of central synapses. Furthermore, the major independent risk factors for AD, Apolipoprotein (Apo) E and ApoJ/Clusterin, are lipoprotein ligands for LRPs. Receptors and ligands mutually influence their intracellular trafficking and thereby the functions and abilities of neurons and the blood-brain-barrier to turn over and remove the pathological product of APP, the amyloid-β peptide. This article will review and summarize the molecular mechanisms that are shared by APP and LRPs and discuss their relative contributions to AD.
Collapse
Affiliation(s)
- Theresa Pohlkamp
- Department of Molecular Genetics, UT Southwestern Medical CenterDallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical CenterDallas, TX, USA
| | - Catherine R Wasser
- Department of Molecular Genetics, UT Southwestern Medical CenterDallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical CenterDallas, TX, USA
| | - Joachim Herz
- Department of Molecular Genetics, UT Southwestern Medical CenterDallas, TX, USA; Center for Translational Neurodegeneration Research, UT Southwestern Medical CenterDallas, TX, USA; Department of Neuroscience, UT Southwestern Medical CenterDallas, TX, USA; Department of Neurology and Neurotherapeutics, UT Southwestern Medical CenterDallas, TX, USA
| |
Collapse
|
34
|
Sun Y, Cheng X, Hu J, Gao Z. The Role of GluN2A in Cerebral Ischemia: Promoting Neuron Death and Survival in the Early Stage and Thereafter. Mol Neurobiol 2017; 55:1208-1216. [DOI: 10.1007/s12035-017-0395-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/09/2017] [Indexed: 01/10/2023]
|
35
|
Desikan RS, Barkovich AJ. Malformations of cortical development. Ann Neurol 2016; 80:797-810. [PMID: 27862206 PMCID: PMC5177533 DOI: 10.1002/ana.24793] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 09/26/2016] [Accepted: 09/26/2016] [Indexed: 01/05/2023]
Abstract
Malformations of cortical development (MCDs) compose a diverse range of disorders that are common causes of neurodevelopmental delay and epilepsy. With improved imaging and genetic methodologies, the underlying molecular and pathobiological characteristics of several MCDs have been recently elucidated. In this review, we discuss genetic and molecular alterations that disrupt normal cortical development, with emphasis on recent discoveries, and provide detailed radiological features of the most common and important MCDs. Ann Neurol 2016;80:797-810.
Collapse
Affiliation(s)
- Rahul S. Desikan
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - A. James Barkovich
- Neuroradiology Section, Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
36
|
Ampuero E, Jury N, Härtel S, Marzolo MP, van Zundert B. Interfering of the Reelin/ApoER2/PSD95 Signaling Axis Reactivates Dendritogenesis of Mature Hippocampal Neurons. J Cell Physiol 2016; 232:1187-1199. [PMID: 27653801 DOI: 10.1002/jcp.25605] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 09/12/2016] [Indexed: 12/21/2022]
Abstract
Reelin, an extracellular glycoprotein secreted in embryonic and adult brain, participates in neuronal migration and neuronal plasticity. Extensive evidence shows that reelin via activation of the ApoER2 and VLDLR receptors promotes dendrite and spine formation during early development. Further evidence suggests that reelin signaling is needed to maintain a stable architecture in mature neurons, but, direct evidence is lacking. During activity-dependent maturation of the neuronal circuitry, the synaptic protein PSD95 is inserted into the postsynaptic membrane to induce structural refinement and stability of spines and dendrites. Given that ApoER2 interacts with PSD95, we tested if reelin signaling interference in adult neurons reactivates the dendritic architecture. Unlike findings in developing cultures, the presently obtained in vitro and in vivo data show, for the first time, that reelin signaling interference robustly increase dendritogenesis and reduce spine density in mature hippocampal neurons. In particular, the expression of a mutant ApoER2 form (ApoER2-tailless), which is unable to interact with PSD95 and hence cannot transduce reelin signaling, resulted in robust dendritogenesis in mature hippocampal neurons in vitro. These results indicate that reelin/ApoER2/PSD95 signaling is important for neuronal structure maintenance in mature neurons. Mechanistically, obtained immunofluorescent data indicate that reelin signaling impairment reduced synaptic PSD95 levels, consequently leading to synaptic re-insertion of NR2B-NMDARs. Our findings underscore the importance of reelin in maintaining adult network stability and reveal a new mode for reactivating dendritogenesis in neurological disorders where dendritic arbor complexity is limited, such as in depression, Alzheimer's disease, and stroke. J. Cell. Physiol. 232: 1187-1199, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Estibaliz Ampuero
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | - Nur Jury
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| | - Steffen Härtel
- SCIAN-Lab, CIMT, Bomedical Neuroscience Institute (BNI), ICBM, Faculty of Medicine, University of Chile, Santiago, Chile
| | - María-Paz Marzolo
- Laboratorio de Tráfico Intracelular y Señalización, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica, Santiago, Chile
| | - Brigitte van Zundert
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
37
|
Iafrati J, Malvache A, Gonzalez Campo C, Orejarena MJ, Lassalle O, Bouamrane L, Chavis P. Multivariate synaptic and behavioral profiling reveals new developmental endophenotypes in the prefrontal cortex. Sci Rep 2016; 6:35504. [PMID: 27765946 PMCID: PMC5073243 DOI: 10.1038/srep35504] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/30/2016] [Indexed: 01/02/2023] Open
Abstract
The postnatal maturation of the prefrontal cortex (PFC) represents a period of increased vulnerability to risk factors and emergence of neuropsychiatric disorders. To disambiguate the pathophysiological mechanisms contributing to these disorders, we revisited the endophenotype approach from a developmental viewpoint. The extracellular matrix protein reelin which contributes to cellular and network plasticity, is a risk factor for several psychiatric diseases. We mapped the aggregate effect of the RELN risk allele on postnatal development of PFC functions by cross-sectional synaptic and behavioral analysis of reelin-haploinsufficient mice. Multivariate analysis of bootstrapped datasets revealed subgroups of phenotypic traits specific to each maturational epoch. The preeminence of synaptic AMPA/NMDA receptor content to pre-weaning and juvenile endophenotypes shifts to long-term potentiation and memory renewal during adolescence followed by NMDA-GluN2B synaptic content in adulthood. Strikingly, multivariate analysis shows that pharmacological rehabilitation of reelin haploinsufficient dysfunctions is mediated through induction of new endophenotypes rather than reversion to wild-type traits. By delineating previously unknown developmental endophenotypic sequences, we conceived a promising general strategy to disambiguate the molecular underpinnings of complex psychiatric disorders and for the rational design of pharmacotherapies in these disorders.
Collapse
Affiliation(s)
- Jillian Iafrati
- INSERM, 13009 Marseille, France; INMED UMR S 901, 13009 Marseille, France; Aix-Marseille Université, 13009 Marseille, France
| | - Arnaud Malvache
- INSERM, 13009 Marseille, France; INMED UMR S 901, 13009 Marseille, France; Aix-Marseille Université, 13009 Marseille, France
| | - Cecilia Gonzalez Campo
- INSERM, 13009 Marseille, France; INMED UMR S 901, 13009 Marseille, France; Aix-Marseille Université, 13009 Marseille, France
| | - M Juliana Orejarena
- INSERM, 13009 Marseille, France; INMED UMR S 901, 13009 Marseille, France; Aix-Marseille Université, 13009 Marseille, France
| | - Olivier Lassalle
- INSERM, 13009 Marseille, France; INMED UMR S 901, 13009 Marseille, France; Aix-Marseille Université, 13009 Marseille, France
| | - Lamine Bouamrane
- INSERM, 13009 Marseille, France; INMED UMR S 901, 13009 Marseille, France; Aix-Marseille Université, 13009 Marseille, France
| | - Pascale Chavis
- INSERM, 13009 Marseille, France; INMED UMR S 901, 13009 Marseille, France; Aix-Marseille Université, 13009 Marseille, France
| |
Collapse
|
38
|
Mullen BR, Ross B, Chou JW, Khankan R, Khialeeva E, Bui K, Carpenter EM. A Complex Interaction Between Reduced Reelin Expression and Prenatal Organophosphate Exposure Alters Neuronal Cell Morphology. ASN Neuro 2016; 8:8/3/1759091416656253. [PMID: 27364165 PMCID: PMC4962342 DOI: 10.1177/1759091416656253] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 05/11/2016] [Indexed: 01/09/2023] Open
Abstract
Genetic and environmental factors are both likely to contribute to neurodevelopmental disorders including schizophrenia, autism spectrum disorders, and major depressive disorders. Prior studies from our laboratory and others have demonstrated that the combinatorial effect of two factors—reduced expression of reelin protein and prenatal exposure to the organophosphate pesticide chlorpyrifos oxon—gives rise to acute biochemical effects and to morphological and behavioral phenotypes in adolescent and young adult mice. In the current study, we examine the consequences of these factors on reelin protein expression and neuronal cell morphology in adult mice. While the cell populations that express reelin in the adult brain appear unchanged in location and distribution, the levels of full length and cleaved reelin protein show persistent reductions following prenatal exposure to chlorpyrifos oxon. Cell positioning and organization in the hippocampus and cerebellum are largely normal in animals with either reduced reelin expression or prenatal exposure to chlorpyrifos oxon, but cellular complexity and dendritic spine organization is altered, with a skewed distribution of immature dendritic spines in adult animals. Paradoxically, combinatorial exposure to both factors appears to generate a rescue of the dendritic spine phenotypes, similar to the mitigation of behavioral and morphological changes observed in our prior study. Together, our observations support an interaction between reelin expression and chlorpyrifos oxon exposure that is not simply additive, suggesting a complex interplay between genetic and environmental factors in regulating brain morphology.
Collapse
Affiliation(s)
- Brian R Mullen
- Molecular, Cellular and Integrative Physiology, University of California, Los Angeles, CA, USA
| | - Brennan Ross
- Neuroscience Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Joan Wang Chou
- Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Rana Khankan
- Molecular, Cellular and Integrative Physiology, University of California, Los Angeles, CA, USA
| | - Elvira Khialeeva
- Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA
| | - Kimberly Bui
- Molecular, Cellular and Developmental Biology, University of California, Los Angeles, CA, USA
| | - Ellen M Carpenter
- Molecular, Cellular and Integrative Physiology, University of California, Los Angeles, CA, USA Neuroscience Interdepartmental Program, University of California, Los Angeles, CA, USA Molecular Biology Interdepartmental Program, University of California, Los Angeles, CA, USA Department of Psychiatry and Biobehavioral Science, University of California, Los Angeles, CA, USA
| |
Collapse
|
39
|
Crider A, Pillai A. The Neurobiological Basis for Social Affiliation in Autism Spectrum Disorder and Schizophrenia. Curr Behav Neurosci Rep 2016; 3:154-164. [DOI: 10.1007/s40473-016-0079-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
40
|
Bosch C, Muhaisen A, Pujadas L, Soriano E, Martínez A. Reelin Exerts Structural, Biochemical and Transcriptional Regulation Over Presynaptic and Postsynaptic Elements in the Adult Hippocampus. Front Cell Neurosci 2016; 10:138. [PMID: 27303269 PMCID: PMC4884741 DOI: 10.3389/fncel.2016.00138] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 05/10/2016] [Indexed: 11/13/2022] Open
Abstract
Reelin regulates neuronal positioning and synaptogenesis in the developing brain, and adult brain plasticity. Here we used transgenic mice overexpressing Reelin (Reelin-OE mice) to perform a comprehensive dissection of the effects of this protein on the structural and biochemical features of dendritic spines and axon terminals in the adult hippocampus. Electron microscopy (EM) revealed both higher density of synapses and structural complexity of both pre- and postsynaptic elements in transgenic mice than in WT mice. Dendritic spines had larger spine apparatuses, which correlated with a redistribution of Synaptopodin. Most of the changes observed in Reelin-OE mice were reversible after blockade of transgene expression, thus supporting the specificity of the observed phenotypes. Western blot and transcriptional analyses did not show major changes in the expression of pre- or postsynaptic proteins, including SNARE proteins, glutamate receptors, and scaffolding and signaling proteins. However, EM immunogold assays revealed that the NMDA receptor subunits NR2a and NR2b, and p-Cofilin showed a redistribution from synaptic to extrasynaptic pools. Taken together with previous studies, the present results suggest that Reelin regulates the structural and biochemical properties of adult hippocampal synapses by increasing their density and morphological complexity and by modifying the distribution and trafficking of major glutamatergic components.
Collapse
Affiliation(s)
- Carles Bosch
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of BarcelonaBarcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), MadridSpain; Vall d'Hebron Institut de RecercaBarcelona, Spain
| | - Ashraf Muhaisen
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of BarcelonaBarcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), MadridSpain; Vall d'Hebron Institut de RecercaBarcelona, Spain; Institute of Neurosciences, University of BarcelonaBarcelona, Spain
| | - Lluís Pujadas
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of BarcelonaBarcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), MadridSpain; Vall d'Hebron Institut de RecercaBarcelona, Spain
| | - Eduardo Soriano
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of BarcelonaBarcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), MadridSpain; Vall d'Hebron Institut de RecercaBarcelona, Spain; Institute of Neurosciences, University of BarcelonaBarcelona, Spain; Institució Catalana de Recerca i Estudis Avançats AcademiaBarcelona, Spain
| | - Albert Martínez
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona Barcelona, Spain
| |
Collapse
|
41
|
Lee GH, D'Arcangelo G. New Insights into Reelin-Mediated Signaling Pathways. Front Cell Neurosci 2016; 10:122. [PMID: 27242434 PMCID: PMC4860420 DOI: 10.3389/fncel.2016.00122] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/27/2016] [Indexed: 11/21/2022] Open
Abstract
Reelin, a multifunctional extracellular protein that is important for mammalian brain development and function, is secreted by different cell types in the prenatal or postnatal brain. The spatiotemporal regulation of Reelin expression and distribution during development relates to its multifaceted function in the brain. Prenatally Reelin controls neuronal radial migration and proper positioning in cortical layers, whereas postnatally Reelin promotes neuronal maturation, synaptic formation and plasticity. The molecular mechanisms underlying the distinct biological functions of Reelin during and after brain development involve unique and overlapping signaling pathways that are activated following Reelin binding to its cell surface receptors. Distinct Reelin ligand isoforms, such as the full-length protein or fragments generated by proteolytic cleavage differentially affect the activity of downstream signaling pathways. In this review, we discuss recent advances in our understanding of the signaling transduction pathways activated by Reelin that regulate different aspects of brain development and function. A core signaling machinery, including ApoER2/VLDLR receptors, Src/Fyn kinases, and the adaptor protein Dab1, participates in all known aspects of Reelin biology. However, distinct downstream mechanisms, such as the Crk/Rap1 pathway and cell adhesion molecules, play crucial roles in the control of neuronal migration, whereas the PI3K/Akt/mTOR pathway appears to be more important for dendrite and spine development. Finally, the NMDA receptor (NMDAR) and an unidentified receptor contribute to the activation of the MEK/Erk1/2 pathway leading to the upregulation of genes involved in synaptic plasticity and learning. This knowledge may provide new insight into neurodevelopmental or neurodegenerative disorders that are associated with Reelin dysfunction.
Collapse
Affiliation(s)
- Gum Hwa Lee
- College of Pharmacy, Chosun University Gwangju, South Korea
| | - Gabriella D'Arcangelo
- Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey Piscataway, NJ, USA
| |
Collapse
|
42
|
Identification of RELN variation p.Thr3192Ser in a Chinese family with schizophrenia. Sci Rep 2016; 6:24327. [PMID: 27071546 PMCID: PMC4829830 DOI: 10.1038/srep24327] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 03/24/2016] [Indexed: 12/11/2022] Open
Abstract
Schizophrenia (SCZ) is a serious psychiatric disease with strong heritability. Its complexity is reflected by extensive genetic heterogeneity and much of the genetic liability remains unaccounted for. We applied a combined strategy involving detection of copy number variants (CNVs), whole-genome mapping, and exome sequencing to identify the genetic basis of autosomal-dominant SCZ in a Chinese family. To rule out pathogenic CNVs, we first performed Illumina single nucleotide polymorphism (SNP) array analysis on samples from two patients and one psychiatrically healthy family member, but no pathogenic CNVs were detected. In order to further narrow down the susceptible region, we conducted genome-wide linkage analysis and mapped the disease locus to chromosome 7q21.13-22.3, with a maximum multipoint logarithm of odds score of 2.144. Whole-exome sequencing was then carried out with samples from three affected individuals and one unaffected individual in the family. A missense variation c.9575 C > G (p.Thr3192Ser) was identified in RELN, which is known as a risk gene for SCZ, located on chromosome 7q22, in the pedigree. This rare variant, as a highly penetrant risk variant, co-segregated with the phenotype. Our results provide genetic evidence that RELN may be one of pathogenic gene in SCZ.
Collapse
|
43
|
Guidotti A, Grayson DR, Caruncho HJ. Epigenetic RELN Dysfunction in Schizophrenia and Related Neuropsychiatric Disorders. Front Cell Neurosci 2016; 10:89. [PMID: 27092053 PMCID: PMC4820443 DOI: 10.3389/fncel.2016.00089] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 03/21/2016] [Indexed: 01/02/2023] Open
Abstract
REELIN (RELN) is a large (420 kDa) glycoprotein that in adulthood is mostly synthesized in GABAergic neurons of corticolimbic structures. Upon secretion in the extracellular matrix (ECM), RELN binds to VLDL, APOE2, and α3β2 Integrin receptors located on dendritic shafts and spines of postsynaptic pyramidal neurons. Reduced levels of RELN expression in the adult brain induce cognitive impairment and dendritic spine density deficits. RELN supplementation recovers these deficits suggesting a trophic action for RELN in synaptic plasticity. We and others have shown that altered RELN expression in schizophrenia (SZ) and bipolar (BP) disorder patients is difficult to reconcile with classical Mendelian genetic disorders and it is instead plausible to associate these disorders with altered epigenetic homeostasis. Support for the contribution of altered epigenetic mechanisms in the down-regulation of RELN expression in corticolimbic structures of psychotic patients includes the concomitant increase of DNA-methyltransferases and the increased levels of the methyl donor S-adenosylmethionine (SAM). It is hypothesized that these conditions lead to RELN promoter hypermethylation and a reduction in RELN protein amounts in psychotic patients. The decreased synthesis and release of RELN from GABAergic corticolimbic neurons could serve as a model to elucidate the epigenetic pathophysiological mechanisms acting at pyramidal neuron dendrites that regulate synaptic plasticity and cognition in psychotic and non-psychotic subjects.
Collapse
Affiliation(s)
- Alessandro Guidotti
- Department of Psychiatry, The Psychiatric Institute, College of Medicine, University of Illinois at Chicago Chicago, IL, USA
| | - Dennis R Grayson
- Department of Psychiatry, The Psychiatric Institute, College of Medicine, University of Illinois at Chicago Chicago, IL, USA
| | - Hector J Caruncho
- College of Pharmacy and Nutrition, University of Saskatchewan Saskatoon, SK, Canada
| |
Collapse
|
44
|
Lammert DB, Howell BW. RELN Mutations in Autism Spectrum Disorder. Front Cell Neurosci 2016; 10:84. [PMID: 27064498 PMCID: PMC4814460 DOI: 10.3389/fncel.2016.00084] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 03/18/2016] [Indexed: 11/13/2022] Open
Abstract
RELN encodes a large, secreted glycoprotein integral to proper neuronal positioning during development and regulation of synaptic function postnatally. Rare, homozygous, null mutations lead to lissencephaly with cerebellar hypoplasia (LCH), accompanied by developmental delay and epilepsy. Until recently, little was known about the frequency or consequences of heterozygous mutations. Several lines of evidence from multiple studies now implicate heterozygous mutations in RELN in autism spectrum disorders (ASD). RELN maps to the AUTS1 locus on 7q22, and at this time over 40 distinct mutations have been identified that would alter the protein sequence, four of which are de novo. The RELN mutations that are most clearly consequential are those that are predicted to inactivate the signaling function of the encoded protein and those that fall in a highly conserved RXR motif found at the core of the 16 Reelin subrepeats. Despite the growing evidence of RELN dysfunction in ASD, it appears that these mutations in isolation are insufficient and that secondary genetic or environmental factors are likely required for a diagnosis.
Collapse
Affiliation(s)
- Dawn B Lammert
- Department of Neuroscience and Physiology, SUNY Upstate Medical School Syracuse, NY, USA
| | - Brian W Howell
- Department of Neuroscience and Physiology, SUNY Upstate Medical School Syracuse, NY, USA
| |
Collapse
|
45
|
Negrón-Oyarzo I, Lara-Vásquez A, Palacios-García I, Fuentealba P, Aboitiz F. Schizophrenia and reelin: a model based on prenatal stress to study epigenetics, brain development and behavior. Biol Res 2016; 49:16. [PMID: 26968981 PMCID: PMC4787713 DOI: 10.1186/s40659-016-0076-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 02/22/2016] [Indexed: 11/20/2022] Open
Abstract
Schizophrenia is a severe psychiatric disorder that results in a significant disability for the patient. The disorder is characterized by impairment of the adaptive orchestration of actions, a cognitive function that is mainly dependent on the prefrontal cortex. This behavioral deficit, together with cellular and neurophysiological alterations in the prefrontal cortex, as well as reduced density of GABAergic cells and aberrant oscillatory activity, all indicate structural and functional deficits of the prefrontal cortex in schizophrenia. Among the several risk factors for the development of schizophrenia, stress during the prenatal period has been identified as crucial. Thus, it is proposed that prenatal stress induces neurodevelopmental alterations in the prefrontal cortex that are expressed as cognitive impairment observed in schizophrenia. However, the precise mechanisms that link prenatal stress with the impairment of prefrontal cortex function is largely unknown. Reelin is an extracellular matrix protein involved in the development of cortical neural connectivity at embryonic stages, and in synaptic plasticity at postnatal stages. Interestingly, down-regulation of reelin expression has been associated with epigenetic changes in the reelin gene of the prefrontal cortex of schizophrenic patients. We recently showed that, similar to schizophrenic patients, prenatal stress induces down-expression of reelin associated with the methylation of its promoter in the rodent prefrontal cortex. These alterations were paralleled with altered prefrontal cortex functional connectivity and impairment in prefrontal cortex-dependent behavioral tasks. Therefore, considering molecular, cellular, physiological and behavioral evidence, we propose a unifying framework that links prenatal stress and prefrontal malfunction through epigenetic alterations of the reelin gene.
Collapse
Affiliation(s)
- Ignacio Negrón-Oyarzo
- Departamento de Psiquiatría, Escuela de Medicina, and Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ariel Lara-Vásquez
- Departamento de Psiquiatría, Escuela de Medicina, and Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ismael Palacios-García
- Departamento de Psiquiatría, Escuela de Medicina, and Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Fuentealba
- Departamento de Psiquiatría, Escuela de Medicina, and Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisco Aboitiz
- Departamento de Psiquiatría, Escuela de Medicina, and Centro Interdisciplinario de Neurociencia, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
46
|
Kwon HJ, Jang WC, Lim MH. Association between RELN Gene Polymorphisms and Attention Deficit Hyperactivity Disorder in Korean Children. Psychiatry Investig 2016; 13:210-6. [PMID: 27081382 PMCID: PMC4823197 DOI: 10.4306/pi.2016.13.2.210] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Attention deficit hyperactivity disorder (ADHD) is common disorder of the school-age population. ADHD is familial and genetic studies estimate heritability at 80-90%. The aim of the present study was to investigate the association between the genetic type and alleles for RELNgene (rs736707, rs2229864, rs362746, rs362726, rs362691, rs1062831, rs607755, and rs2072403) in Korean children with ADHD. METHODS The sample consisted of 180 ADHD children and 159 control children. We diagnosed ADHD according to DSM-IV. ADHD symptoms were evaluated with Conners' Parent Rating Scales and Dupaul Parent ADHD Rating Scales. Blood samples were taken from the 339 subjects, DNA was extracted from blood lymphocytes, and PCR was performed for RELN Polymorphism. Alleles and genotype frequencies were compared using the chi-square test. We compared the allele and genotype frequencies of RELN gene polymorphism in the ADHD and control groups. RESULTS This study showed that there was a significant correlation among the frequencies of the rs736707 (OR=1.40, 95% CI=1.03-1.90, p=0.031) of alleles of RELN, but the final conclusions are not definite. CONCLUSION Follow up studies with larger patient or pure subgroups are expected. These results suggested that RELN might be related to ADHD symptoms.
Collapse
Affiliation(s)
- Ho Jang Kwon
- Environmental Health Center, Dankook Medical Hospital, Cheonan, Republic of Korea
- Department of Preventive Medicine, College of Medicine, Dankook University, Cheonan, Republic of Korea
| | - Won-Cheol Jang
- Department of Chemistry, College of Natural Science, Dankook University, Cheonan, Republic of Korea
| | - Myung Ho Lim
- Environmental Health Center, Dankook Medical Hospital, Cheonan, Republic of Korea
- Department of Psychology, College of Public Welfare, Dankook University, Cheonan, Republic of Korea
| |
Collapse
|
47
|
Caruncho HJ, Brymer K, Romay-Tallón R, Mitchell MA, Rivera-Baltanás T, Botterill J, Olivares JM, Kalynchuk LE. Reelin-Related Disturbances in Depression: Implications for Translational Studies. Front Cell Neurosci 2016; 10:48. [PMID: 26941609 PMCID: PMC4766281 DOI: 10.3389/fncel.2016.00048] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/11/2016] [Indexed: 02/02/2023] Open
Abstract
The finding that reelin expression is significantly decreased in mood and psychotic disorders, together with evidence that reelin can regulate key aspects of hippocampal plasticity in the adult brain, brought our research group and others to study the possible role of reelin in the pathogenesis of depression. This review describes recent progress on this topic using an animal model of depression that makes use of repeated corticosterone (CORT) injections. This methodology produces depression-like symptoms in both rats and mice that are reversed by antidepressant treatment. We have reported that CORT causes a decrease in the number of reelin-immunopositive cells in the dentate gyrus subgranular zone (SGZ), where adult hippocampal neurogenesis takes place; that down-regulation of the number of reelin-positive cells closely parallels the development of a depression-like phenotype during repeated CORT treatment; that reelin downregulation alters the co-expression of reelin with neuronal nitric oxide synthase (nNOS); that deficits in reelin might also create imbalances in glutamatergic and GABAergic circuits within the hippocampus and other limbic structures; and that co-treatment with antidepressant drugs prevents both reelin deficits and the development of a depression-like phenotype. We also observed alterations in the pattern of membrane protein clustering in peripheral lymphocytes in animals with low levels of reelin. Importantly, we found parallel changes in membrane protein clustering in depression patients, which differentiated two subpopulations of naïve depression patients that showed a different therapeutic response to antidepressant treatment. Here, we review these findings and develop the hypothesis that restoring reelin-related function could represent a novel approach for antidepressant therapies.
Collapse
Affiliation(s)
- Hector J Caruncho
- Neuroscience Cluster, College of Pharmacy and Nutrition, University of Saskatchewan Saskatoon, SK, Canada
| | - Kyle Brymer
- Department of Psychology, University of Saskatchewan Saskatoon, SK, Canada
| | | | - Milann A Mitchell
- Department of Psychology, University of Saskatchewan Saskatoon, SK, Canada
| | - Tania Rivera-Baltanás
- Department of Psychiatry, Alvaro Cunqueiro Hospital, Biomedical Research Institute of Vigo Galicia, Spain
| | - Justin Botterill
- Department of Psychology, University of Saskatchewan Saskatoon, SK, Canada
| | - Jose M Olivares
- Department of Psychiatry, Alvaro Cunqueiro Hospital, Biomedical Research Institute of Vigo Galicia, Spain
| | - Lisa E Kalynchuk
- Department of Medicine, University of Saskatchewan Saskatoon, SK, Canada
| |
Collapse
|
48
|
The Functional and Molecular Properties, Physiological Functions, and Pathophysiological Roles of GluN2A in the Central Nervous System. Mol Neurobiol 2016; 54:1008-1021. [DOI: 10.1007/s12035-016-9715-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 01/11/2016] [Indexed: 11/25/2022]
|
49
|
Drago A, Crisafulli C, Sidoti A, Calabrò M, Serretti A. The microtubule-associated molecular pathways may be genetically disrupted in patients with Bipolar Disorder. Insights from the molecular cascades. J Affect Disord 2016; 190:429-438. [PMID: 26551401 DOI: 10.1016/j.jad.2015.10.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 09/24/2015] [Accepted: 10/10/2015] [Indexed: 01/15/2023]
Abstract
Bipolar Disorder is a severe disease characterized by pathological mood swings from major depressive episodes to manic ones and vice versa. The biological underpinnings of Bipolar Disorder have yet to be defined. As a consequence, pharmacological treatments are suboptimal. In the present paper we test the hypothesis that the molecular pathways involved with the direct targets of lithium, hold significantly more genetic variations associated with BD. A molecular pathway approach finds its rationale in the polygenic nature of the disease. The pathways were tested in a sample of ∼ 7,000 patients and controls. Data are available from the public NIMH database. The definition of the pathways was conducted according to the National Cancer Institute (http://pid.nci.nih.gov/). As a result, 3 out of the 18 tested pathways related to lithium action resisted the permutation analysis and were found to be associated with BD. These pathways were related to Reelin, Integrins and Aurora. A pool of genes selected from the ones linked with the above pathways was further investigated in order to identify the fine molecular mechanics shared by our significant pathways and also their link with lithium mechanism of action. The data obtained point out to a possible involvement of microtubule-related mechanics.
Collapse
Affiliation(s)
- Antonio Drago
- Department of Biomedical and Neuromotor Sciences - DIBINEM - University of Bologna, Bologna, Italy
| | - Concetta Crisafulli
- Department of Biomedical Science and Morphological and Functional Images, University of Messina, Via Consolare Valeria, 98125 Messina, Italy.
| | - Antonina Sidoti
- Department of Biomedical Science and Morphological and Functional Images, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Marco Calabrò
- Department of Biomedical Science and Morphological and Functional Images, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Alessandro Serretti
- Department of Biomedical and Neuromotor Sciences - DIBINEM - University of Bologna, Bologna, Italy
| |
Collapse
|
50
|
The Role of Reelin Signaling in Alzheimer’s Disease. Mol Neurobiol 2015; 53:5692-700. [DOI: 10.1007/s12035-015-9459-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 09/25/2015] [Indexed: 12/23/2022]
|