1
|
Salasova A, Nykjær A. Emerging potential of progranulin-dependent SorCS2 signaling in healthy and diseased nervous systems. Neural Regen Res 2025; 20:2591-2593. [PMID: 39503427 PMCID: PMC11801297 DOI: 10.4103/nrr.nrr-d-24-00734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 02/08/2025] Open
Affiliation(s)
- Alena Salasova
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Centre of Excellence PROMEMO, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Anders Nykjær
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Centre of Excellence PROMEMO, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
2
|
Staehr C, Login H, Melnikova EV, Bakun M, Ziemlinska E, Kisiswa L, Ardestani SB, Nolte SS, Beck HC, Hansen LMB, Postnov D, Verkhratsky A, Malik AR, Nykjaer A, Matchkov VV. SorCS2 Is Important for Astrocytic Function in Neurovascular Signaling. Acta Physiol (Oxf) 2025; 241:e70052. [PMID: 40342271 PMCID: PMC12060088 DOI: 10.1111/apha.70052] [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: 11/24/2024] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
INTRODUCTION The receptor SorCS2 is involved in the trafficking of membrane receptors and transporters. It has been implicated in brain disorders and has previously been reported to be indispensable for ionotropic glutamatergic neurotransmission in the hippocampus. AIM We aimed to study the role of SorCS2 in the control of astrocyte-neuron communication, critical for neurovascular coupling. METHODS Brain slices from P8 and 2-month-old wild-type and SorCS2 knockout (Sorcs2-/-) mice were immunostained for SorCS2, GFAP, AQP4, IB4, and CD31. Neurovascular coupling was assessed in vivo using laser speckle contrast imaging and ex vivo in live brain slices loaded with calcium-sensitive dye. Bulk and cell surface fraction proteomics was analyzed on freshly isolated and cultured astrocytes, respectively, and validated with Western blot and qPCR. RESULTS SorCS2 was strongly expressed in astrocytes, primarily in their endfeet, of P8 mice; however, it was sparsely represented in 2-month-old mice. Sorcs2-/- mice demonstrated reduced neurovascular coupling associated with a reduced astrocytic calcium response to neuronal excitation. No differences in vascularization or endothelium-dependent relaxation ex vivo between the 2-month-old groups were observed. Proteomics suggested changes in glutamatergic signaling and suppressed calcium signaling in Sorcs2-/- brains from both P8 and 2-month-old mice. The increased abundance of glutamate metabotropic receptor 3 in Sorcs2-/- astrocytes was validated by PCR and Western blot. In cultured Sorcs2-/- astrocytes, AQP4 abundance was increased in the bulk lysate but reduced in the cell surface fraction, suggesting impaired trafficking. CONCLUSION The results suggest that SorCS2 expression is important for the development of neurovascular coupling, at least in part by modulating glutamatergic and calcium signaling in astrocytes.
Collapse
Affiliation(s)
- Christian Staehr
- Department of BiomedicineAarhus UniversityAarhusDenmark
- Department of Anaesthesiology and Intensive CareAarhus University HospitalAarhusDenmark
| | - Hande Login
- Department of BiomedicineAarhus UniversityAarhusDenmark
| | | | - Magdalena Bakun
- Mass Spectrometry LaboratoryInstitute of Biochemistry and Biophysics, Polish Academy of SciencesWarsawPoland
| | - Ewelina Ziemlinska
- Cellular Neurobiology Research Group, Faculty of Biology, Institute of Developmental Biology and Biomedical SciencesUniversity of WarsawWarsawPoland
| | | | | | | | | | | | - Dmitry Postnov
- Department of Clinical Medicine, Center of Functionally Integrative NeuroscienceAarhus University Hospital SkejbyAarhusDenmark
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Department of NeurosciencesUniversity of the Basque CountryLeioaSpain
- IKERBASQUE Basque Foundation for ScienceBilbaoSpain
- Department of Forensic Analytical Toxicology, School of Forensic MedicineChina Medical UniversityShenyangChina
- International Joint Research Centre on Purinergic Signalling of Sichuan Province Chengdu University of Traditional Chinese MedicineChengduChina
| | - Anna R. Malik
- Cellular Neurobiology Research Group, Faculty of Biology, Institute of Developmental Biology and Biomedical SciencesUniversity of WarsawWarsawPoland
| | - Anders Nykjaer
- Department of BiomedicineAarhus UniversityAarhusDenmark
- PROMEMO and DANDRITEAarhus UniversityAarhusDenmark
- Department of NeurosurgeryAarhus University Hospital SkejbyAarhusDenmark
| | | |
Collapse
|
3
|
Guo M, Zhao X, Zhao X, Wang G, Ren X, Chen A, Jiang X, Zhang Y, Cheng X, Yu X, Wang H, Li F, Ning Z, Qu L. Genomic and transcriptomic analyses reveal the genetic basis of leg diseases in laying hens. Poult Sci 2025; 104:104887. [PMID: 39970519 PMCID: PMC11880710 DOI: 10.1016/j.psj.2025.104887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/25/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025] Open
Abstract
In recent years, while monitoring commercial laying hens during the brooding period, we noticed that some chicks exhibited an abnormal lameness phenomenon. Systematic tests were carried out for pathogens including, but not limited to, common diseases such as viral arthritis, mycoplasma synoviae, Marek's disease and encephalomyelitis. However, the absence of these pathogens in these chicks rules out these common diseases as a direct cause of limping and points to genetic factors. Leg disorders in laying hens pose significant welfare challenges and result in considerable economic losses within the poultry industry. To address this issue, we conducted a comprehensive genomic and transcriptomic analysis to uncover the genetic basis of leg diseases in laying hens. Our study involved 74 laying hens with leg diseases and 60 healthy controls from three different strains. Radiographic identification and resequencing were performed to analyze genetic loci and candidate genes. Transcriptomic analysis was also conducted on cartilage tissues from both groups. Four potential single nucleotide polymorphism (SNP) loci located on chromosomes 4 and 33 were identified through genome-wide association studies (GWAS). Notably, the functional candidate gene SORCS2 has been particularly implicated in the pathogenesis of leg disease. A total of 1,515 differentially expressed genes (DEGs) were identified through transcriptomic analysis. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses revealed significant enrichment in pathways such as cytoskeletal protein binding, cell cycle regulation, and muscle cell cytoskeleton organization. Notably, the calcium signaling and the extracellular matrix (ECM)-receptor interaction pathways were associated with the development of leg diseases. The integration of genomic and transcriptomic data identified key genetic loci and candidate genes associated with leg health, providing a genetic foundation for improving disease resistance in laying hens. Our findings offer valuable insights for the development of selective breeding strategies to reduce the incidence of leg disorders in laying hens, thereby enhancing their welfare and productivity.
Collapse
Affiliation(s)
- Menghan Guo
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiaoyu Zhao
- Xingrui Agricultural Stock Breeding, Baoding 072550, Hebei Province, China
| | - Xiurong Zhao
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Gang Wang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xufang Ren
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Anqi Chen
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiaoyu Jiang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yalan Zhang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xue Cheng
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiaofan Yu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Huie Wang
- Xinjiang Production and Construction Corps, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, Tarim University, Alar 843300, China
| | - Fugui Li
- Xinjiang Production and Construction Corps, Key Laboratory of Protection and Utilization of Biological Resources in Tarim Basin, Tarim University, Alar 843300, China
| | - Zhonghua Ning
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Lujiang Qu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
4
|
Ambrose A, McNiven V, Wilson D, Tempes A, Underwood M, Chau V, Schulze A, Wyszynska A, Desch K, Malik AR, Mercimek-Andrews S. Neonatal Encephalopathy: Novel Phenotypes and Genotypes Identified by Genome Sequencing. Neurol Genet 2025; 11:e200232. [PMID: 39810752 PMCID: PMC11731368 DOI: 10.1212/nxg.0000000000200232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 11/18/2024] [Indexed: 01/16/2025]
Abstract
Background and Objectives Neonatal encephalopathy (NE) is characterized by an abnormal level of consciousness with or without seizures in the neonatal period. It affects 1-6/1,000 live term newborns. We applied genome sequencing (GS) in term newborns with NE to investigate the underlying genetic causes. Methods We enrolled term newborns according to inclusion/exclusion criteria during their Neonatal Intensive Care admission. We performed GS trio and applied bioinformatic tools. We developed pipelines for manual filters. We applied in silico prediction tools, protein 3D modeling, and functional characterization to assess the pathogenicity of variants. Results Seventeen newborns fulfilled inclusion criteria. We identified 12 variants in 10 genes. We classified 4 variants in PPP2R5D, BCOR, CFL2, and SCN2A (previously established disease genes) as pathogenic/likely pathogenic; 7 variants in DST (previously established disease gene), STAB2, CELF4, SORCS2, CTNND2, and ASTN1 (5 candidate genes) as variants of uncertain significance (VUS); and one variant in STAB2 as likely benign. The CELF4 and ASTN1 copy number variants (CNVs) resulted in structural changes in protein 3D models. The functional characterization of SORCS2 VUS revealed disruption of SorCS2 dimer formation and confirmed its pathogenicity. The functional characterization of STAB2 variants updated their characterization from VUS/likely benign to benign. The CTNND2 VUS resulted in a shift in 3D protein structure. We were not able to perform protein 3D modeling and functional characterization of two DST VUS. We are not certain whether CTNND2 and DST variants may be causative of NE in our study. Discussion The diagnostic rate of research GS was 41% in our prospective study. We broaden the phenotypic spectrum of PPP2R5D-associated Hogue-Janssens syndrome 1, CFL2-associated nemaline myopathy 7, and BCOR-associated oculo-facio-cardio-dental syndrome to include NE and/or neonatal seizures. We identified 3 candidate genes (SORCS2, CELF4, ASTN1) that may cause NE. We believe that protein 3D modeling is an important tool to assess the pathogenicity of CNVs and may advance the discoveries of novel genetic diseases. However, functional characterization of missense variants is essential for discoveries of novel genetic diseases. It seems that GS can help identify more candidate genes compared with ES.
Collapse
Affiliation(s)
- Anastasia Ambrose
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Vanda McNiven
- Division of Genetics, Department of Pediatrics, McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Diane Wilson
- Division of Neonatology, Department of Pediatrics, University of Toronto, Ontario, Canada
| | | | - Mary Underwood
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Vann Chau
- Division of Neurology, Department of Pediatrics, University of Toronto, Ontario, Canada
| | - Andreas Schulze
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Ontario, Canada
| | | | - Karl Desch
- Department of Pediatrics, University of Michigan, Ann Arbor, MI
- Division of Neonatal-Perinatal Medicine, Cell and Molecular Biology Program, Department of Pediatrics, University of Michigan, Ann Arbor, MI
| | - Anna R Malik
- Faculty of Biology, University of Warsaw, Poland
| | - Saadet Mercimek-Andrews
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada; and
- Alberta Health Services, Edmonton Zone, Alberta, Canada
| |
Collapse
|
5
|
Li Y, Yang Y, Guan X, Liu Z, Pan L, Wang Y, Jia X, Yang J, Hou T. SorCS2 is involved in promoting periodontitis-induced depression-like behaviour in mice. Oral Dis 2024; 30:5408-5420. [PMID: 38568959 DOI: 10.1111/odi.14944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Emerging evidence supports the association between periodontitis and depression, although the mechanisms are unclear. This study investigated the role of SorCS2 in the pathogenesis of periodontitis-induced depression. MATERIALS AND METHODS An experimental periodontitis model was established using SorCS2 knockout mice and their wild-type littermates, and depression-like behaviour was evaluated. The expression of proBDNF signalling, neuronal activity, and glutamate-associated signalling pathways were further measured by western blotting and immunofluorescence. In addition, neuroinflammatory status, astrocytic and microglial markers, and the expression of corticosterone-related factors were measured by immunofluorescence, western blotting, and enzyme-linked immunosorbent assays. RESULTS SorCS2 deficiency alleviated periodontitis-induced depression-like behaviour in mice. Further results suggested that SorCS2 deficiency downregulated the expression of pro-BDNF and glutamate signalling and restored neuronal activities in mice with periodontitis. Neuroinflammation in the mouse hippocampus was triggered by experimental periodontitis but was not affected by SorCS2 deficiency. The levels of corticosterone and the expression of glucocorticoid receptors were also not altered. CONCLUSION Our study, for the first time, reveals the critical role of SorCS2 in the pathogenesis of periodontitis-induced depression. The underlying mechanism involves proBDNF and glutamate signalling in the hippocampus, providing a novel therapeutic target for periodontitis-associated depression.
Collapse
Affiliation(s)
- Yingxue Li
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Yao Yang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyue Guan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Zhijun Liu
- Institute of Regenerative and Reconstructive Medicine, Med-X Institute, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lifei Pan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Yuting Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Xiangbin Jia
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Jianmin Yang
- Department of Medicine, Weill Cornell Medical School, Cornell University, New York, New York, USA
| | - Tiezhou Hou
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
- Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
6
|
Chung J, Kim S, Won JH, Park H. Integrating Multimodal Neuroimaging and Genetics: A Structurally-Linked Sparse Canonical Correlation Analysis Approach. IEEE JOURNAL OF TRANSLATIONAL ENGINEERING IN HEALTH AND MEDICINE 2024; 12:659-667. [PMID: 39464624 PMCID: PMC11505868 DOI: 10.1109/jtehm.2024.3463720] [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] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/16/2024] [Accepted: 09/14/2024] [Indexed: 10/29/2024]
Abstract
Neuroimaging genetics represents a multivariate approach aimed at elucidating the intricate relationships between high-dimensional genetic variations and neuroimaging data. Predominantly, existing methodologies revolve around Sparse Canonical Correlation Analysis (SCCA), a framework we expand to 1) encompass multiple imaging modalities and 2) promote the simultaneous identification of structurally linked features across imaging modalities. The structurally linked brain regions were assessed using diffusion tensor imaging, which quantifies the presence of neuronal fibers, thereby grounding our approach in biologically well-founded prior knowledge within the SCCA model. In our proposed structurally linked SCCA framework, we leverage T1-weighted MRI and functional MRI (fMRI) time series data to delineate both the structural and functional characteristics of the brain. Genetic variations, specifically single nucleotide polymorphisms (SNPs), are also incorporated as a genetic modality. Validation of our methodology was conducted using a simulated dataset and large-scale normative data from the Human Connectome Project (HCP). Our approach demonstrated superior performance compared to existing methods on simulated data and revealed interpretable gene-imaging associations in the real dataset. Thus, our methodology lays the groundwork for elucidating the genetic underpinnings of brain structure and function, thereby providing novel insights into the field of neuroscience. Our code is available at https://github.com/mungegg.
Collapse
Affiliation(s)
- Jiwon Chung
- Department of Electrical and Computer EngineeringSungkyunkwan UniversitySuwon16419Republic of Korea
| | - Sunghun Kim
- Department of Electrical and Computer EngineeringSungkyunkwan UniversitySuwon16419Republic of Korea
| | - Ji Hye Won
- Department of Computer Engineering and Artificial IntelligencePukyong National UniversityBusan48513Republic of Korea
| | - Hyunjin Park
- Department of Electrical and Computer EngineeringSungkyunkwan UniversitySuwon16419Republic of Korea
- Center for Neuroscience Imaging ResearchInstitute for Basic ScienceSuwon16419Republic of Korea
| |
Collapse
|
7
|
Kalnytska O, Qvist P, Kunz S, Conrad T, Willnow TE, Schmidt V. SORCS2 activity in pancreatic α-cells safeguards insulin granule formation and release from glucose-stressed β-cells. iScience 2024; 27:108725. [PMID: 38226160 PMCID: PMC10788290 DOI: 10.1016/j.isci.2023.108725] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/18/2023] [Accepted: 12/11/2023] [Indexed: 01/17/2024] Open
Abstract
Sorting receptor SORCS2 is a stress-response factor protecting neurons from acute insults, such as during epilepsy. SORCS2 is also expressed in the pancreas, yet its action in this tissue remains unknown. Combining metabolic studies in SORCS2-deficient mice with ex vivo functional analyses and single-cell transcriptomics of pancreatic tissues, we identified a role for SORCS2 in protective stress response in pancreatic islets, essential to sustain insulin release. We show that SORCS2 is predominantly expressed in islet alpha cells. Loss of expression coincides with inability of these cells to produce osteopontin, a secreted factor that facilitates insulin release from stressed beta cells. In line with diminished osteopontin levels, beta cells in SORCS2-deficient islets show gene expression patterns indicative of aggravated cell stress, and exhibit defects in insulin granule maturation and a blunted glucose response. These findings corroborate a function for SORCS2 in protective stress response that extends to metabolism.
Collapse
Affiliation(s)
- Oleksandra Kalnytska
- Molecular Cardiovascular Research, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Charité – Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Per Qvist
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Séverine Kunz
- Technology Platform for Electron Microscopy, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Thomas Conrad
- Genomics Technology Platform, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
| | - Thomas E. Willnow
- Molecular Cardiovascular Research, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
- Charité – Universitätsmedizin Berlin, 10117 Berlin, Germany
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Vanessa Schmidt
- Molecular Cardiovascular Research, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| |
Collapse
|
8
|
Thomasen PB, Salasova A, Kjaer-Sorensen K, Woloszczuková L, Lavický J, Login H, Tranberg-Jensen J, Almeida S, Beel S, Kavková M, Qvist P, Kjolby M, Ovesen PL, Nolte S, Vestergaard B, Udrea AC, Nejsum LN, Chao MV, Van Damme P, Krivanek J, Dasen J, Oxvig C, Nykjaer A. SorCS2 binds progranulin to regulate motor neuron development. Cell Rep 2023; 42:113333. [PMID: 37897724 DOI: 10.1016/j.celrep.2023.113333] [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: 04/28/2022] [Revised: 07/25/2023] [Accepted: 10/09/2023] [Indexed: 10/30/2023] Open
Abstract
Motor neuron (MN) development and nerve regeneration requires orchestrated action of a vast number of molecules. Here, we identify SorCS2 as a progranulin (PGRN) receptor that is required for MN diversification and axon outgrowth in zebrafish and mice. In zebrafish, SorCS2 knockdown also affects neuromuscular junction morphology and fish motility. In mice, SorCS2 and PGRN are co-expressed by newborn MNs from embryonic day 9.5 until adulthood. Using cell-fate tracing and nerve segmentation, we find that SorCS2 deficiency perturbs cell-fate decisions of brachial MNs accompanied by innervation deficits of posterior nerves. Additionally, adult SorCS2 knockout mice display slower motor nerve regeneration. Interestingly, primitive macrophages express high levels of PGRN, and their interaction with SorCS2-positive motor axon is required during axon pathfinding. We further show that SorCS2 binds PGRN to control its secretion, signaling, and conversion into granulins. We propose that PGRN-SorCS2 signaling controls MN development and regeneration in vertebrates.
Collapse
Affiliation(s)
- Pernille Bogetofte Thomasen
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Alena Salasova
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.
| | - Kasper Kjaer-Sorensen
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Lucie Woloszczuková
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Josef Lavický
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Hande Login
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Jeppe Tranberg-Jensen
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Sergio Almeida
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Sander Beel
- Department of Neurology and Department of Neurosciences, KU Leuven and Center for Brain & Disease Research VIB, 3000 Leuven, Belgium
| | - Michaela Kavková
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Per Qvist
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Mads Kjolby
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Peter Lund Ovesen
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Stella Nolte
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Benedicte Vestergaard
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Andreea-Cornelia Udrea
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | | | - Moses V Chao
- Department of Neuroscience and Physiology, NYU Langone Health, New York, NY 10016, USA
| | - Philip Van Damme
- Department of Neurology and Department of Neurosciences, KU Leuven and Center for Brain & Disease Research VIB, 3000 Leuven, Belgium
| | - Jan Krivanek
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, 62500 Brno, Czech Republic
| | - Jeremy Dasen
- Department of Neuroscience and Physiology, NYU Langone Health, New York, NY 10016, USA
| | - Claus Oxvig
- Department of Molecular Biology and Genetics, Aarhus University, 8000 Aarhus C, Denmark
| | - Anders Nykjaer
- Danish Research Institute of Translational Neuroscience DANDRITE-Nordic EMBL Partnership for Molecular Medicine, and Center of Excellence PROMEMO, 8000 Aarhus C, Denmark; Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark.
| |
Collapse
|
9
|
Lukic B, Curik I, Drzaic I, Galić V, Shihabi M, Vostry L, Cubric-Curik V. Genomic signatures of selection, local adaptation and production type characterisation of East Adriatic sheep breeds. J Anim Sci Biotechnol 2023; 14:142. [PMID: 37932811 PMCID: PMC10626677 DOI: 10.1186/s40104-023-00936-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/04/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND The importance of sheep breeding in the Mediterranean part of the eastern Adriatic has a long tradition since its arrival during the Neolithic migrations. Sheep production system is extensive and generally carried out in traditional systems without intensive systematic breeding programmes for high uniform trait production (carcass, wool and milk yield). Therefore, eight indigenous Croatian sheep breeds from eastern Adriatic treated here as metapopulation (EAS), are generally considered as multipurpose breeds (milk, meat and wool), not specialised for a particular type of production, but known for their robustness and resistance to certain environmental conditions. Our objective was to identify genomic regions and genes that exhibit patterns of positive selection signatures, decipher their biological and productive functionality, and provide a "genomic" characterization of EAS adaptation and determine its production type. RESULTS We identified positive selection signatures in EAS using several methods based on reduced local variation, linkage disequilibrium and site frequency spectrum (eROHi, iHS, nSL and CLR). Our analyses identified numerous genomic regions and genes (e.g., desmosomal cadherin and desmoglein gene families) associated with environmental adaptation and economically important traits. Most candidate genes were related to meat/production and health/immune response traits, while some of the candidate genes discovered were important for domestication and evolutionary processes (e.g., HOXa gene family and FSIP2). These results were also confirmed by GO and QTL enrichment analysis. CONCLUSIONS Our results contribute to a better understanding of the unique adaptive genetic architecture of EAS and define its productive type, ultimately providing a new opportunity for future breeding programmes. At the same time, the numerous genes identified will improve our understanding of ruminant (sheep) robustness and resistance in the harsh and specific Mediterranean environment.
Collapse
Affiliation(s)
- Boris Lukic
- Faculty of Agrobiotechnical Sciences Osijek, J.J, Strossmayer University of Osijek, Vladimira Preloga 1, 31000, Osijek, Croatia.
| | - Ino Curik
- Department of Animal Science, Faculty of Agriculture, University of Zagreb, Svetošimunska Cesta 25, 10000, Zagreb, Croatia.
| | - Ivana Drzaic
- Department of Animal Science, Faculty of Agriculture, University of Zagreb, Svetošimunska Cesta 25, 10000, Zagreb, Croatia
| | - Vlatko Galić
- Department of Maize Breeding and Genetics, Agricultural Institute Osijek, Južno predgrađe 17, 31000, Osijek, Croatia
| | - Mario Shihabi
- Department of Animal Science, Faculty of Agriculture, University of Zagreb, Svetošimunska Cesta 25, 10000, Zagreb, Croatia
| | - Luboš Vostry
- Czech University of Life Sciences Prague, Kamýcká 129, 165 00, Praque, Czech Republic
| | - Vlatka Cubric-Curik
- Department of Animal Science, Faculty of Agriculture, University of Zagreb, Svetošimunska Cesta 25, 10000, Zagreb, Croatia
| |
Collapse
|
10
|
Klein M, Failla AV, Hermey G. Internally tagged Vps10p-domain receptors reveal uptake of the neurotrophin BDNF. J Biol Chem 2023; 299:105216. [PMID: 37660918 PMCID: PMC10540051 DOI: 10.1016/j.jbc.2023.105216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/14/2023] [Accepted: 08/18/2023] [Indexed: 09/05/2023] Open
Abstract
The Vps10p-domain (Vps10p-D) receptor family consists of Sortilin, SorLA, SorCS1, SorCS2, and SorCS3. They mediate internalization and intracellular sorting of specific cargo in various cell types, but underlying molecular determinants are incompletely understood. Deciphering the dynamic intracellular itineraries of Vps10p-D receptors is crucial for understanding their role in physiological and cytopathological processes. However, studying their spatial and temporal dynamics by live imaging has been challenging so far, as terminal tagging with fluorophores presumably impedes several of their protein interactions and thus functions. Here, we addressed the lack of appropriate tools and developed functional versions of all family members internally tagged in their ectodomains. We predict folding of the newly designed receptors by bioinformatics and show their exit from the endoplasmic reticulum. We examined their subcellular localization in immortalized cells and primary cultured neurons by immunocytochemistry and live imaging. This was, as far as known, identical to that of wt counterparts. We observed homodimerization of fluorophore-tagged SorCS2 by coimmunoprecipitation and fluorescence lifetime imaging, suggesting functional leucine-rich domains. Through ligand uptake experiments, live imaging and fluorescence lifetime imaging, we show for the first time that all Vps10p-D receptors interact with the neurotrophin brain-derived neurotrophic factor and mediate its uptake, indicating functionality of the Vps10p-Ds. In summary, we developed versions of all Vps10p-D receptors, with internal fluorophore tags that preserve several functions of the cytoplasmic and extracellular domains. These newly developed fluorophore-tagged receptors are likely to serve as powerful functional tools for accurate live studies of the individual cellular functions of Vps10p-D receptors.
Collapse
Affiliation(s)
- Marcel Klein
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | | | - Guido Hermey
- Institute for Molecular and Cellular Cognition, Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
11
|
Febbraro F, Andersen HHB, Kitt MM, Willnow TE. Spatially and temporally distinct patterns of expression for VPS10P domain receptors in human cerebral organoids. Front Cell Dev Biol 2023; 11:1229584. [PMID: 37842085 PMCID: PMC10570844 DOI: 10.3389/fcell.2023.1229584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/28/2023] [Indexed: 10/17/2023] Open
Abstract
Vacuolar protein sorting 10 protein (VPS10P) domain receptors are a unique class of intracellular sorting receptors that emerge as major risk factors associated with psychiatric and neurodegenerative diseases, including bipolar disorders, autism, schizophrenia, as well as Alzheimer's disease and frontotemporal dementia. Yet, the lack of suitable experimental models to study receptor functions in the human brain has hampered elucidation of receptor actions in brain disease. Here, we have adapted protocols using human cerebral organoids to the detailed characterization of VPS10P domain receptor expression during neural development and differentiation, including single-cell RNA sequencing. Our studies uncovered spatial and temporal patterns of expression unique to individual receptor species in the human brain. While SORL1 expression is abundant in stem cells and SORCS1 peaks in neural progenitors at onset of neurogenesis, SORT1 and SORCS2 show increasing expression with maturation of neuronal and non-neuronal cell types, arguing for distinct functions in development versus the adult brain. In neurons, subcellular localization also distinguishes between types of receptor species, either mainly localized to the cell soma (SORL1 and SORT1) or also to neuronal projections (SORCS1 and SORCS2), suggesting divergent functions in protein sorting between Golgi and the endo-lysosomal system or along axonal and dendritic tracks. Taken together, our findings provide an important resource on temporal, spatial, and subcellular patterns of VPS10P domain receptor expression in cerebral organoids for further elucidation of receptor (dys) functions causative of behavioral and cognitive defects of the human brain.
Collapse
Affiliation(s)
- Fabia Febbraro
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Meagan M. Kitt
- Max Delbrueck Center for Molecular Medicine, Charité—Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas E. Willnow
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Max Delbrueck Center for Molecular Medicine, Charité—Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
12
|
Skeldal S, Voss LF, Lende J, Pedersen SB, Mølgaard S, Kaas M, Demange P, Bentsen AH, Fuglsang M, Sander MR, Buttenschøn H, Gustafsen C, Madsen P, Glerup S. Alternative splicing regulates adaptor protein binding, trafficking, and activity of the Vps10p domain receptor SorCS2 in neuronal development. J Biol Chem 2023; 299:105102. [PMID: 37507021 PMCID: PMC10463258 DOI: 10.1016/j.jbc.2023.105102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/12/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023] Open
Abstract
The Vps10p domain receptor SorCS2 is crucial for the development and function of the nervous system and essential for brain-derived neurotrophic factor (BDNF)-induced changes in neuronal morphology and plasticity. SorCS2 regulates the subcellular trafficking of the BDNF signaling receptor TrkB as well as selected neurotransmitter receptors in a manner that is dependent on the SorCS2 intracellular domain (ICD). However, the cellular machinery and adaptor protein (AP) interactions that regulate receptor trafficking via the SorCS2 ICD are unknown. We here identify four splice variants of human SorCS2 differing in the insertion of an acidic cluster motif and/or a serine residue within the ICD. We show that each variant undergoes posttranslational proteolytic processing into a one- or two-chain receptor, giving rise to eight protein isoforms, the expression of which differs between neuronal and nonneuronal tissues and is affected by cellular stressors. We found that the only variants without the serine were able to rescue BDNF-induced branching of SorCS2 knockout hippocampal neurons, while variants without the acidic cluster showed increased interactions with clathrin-associated APs AP-1, AP-2, and AP-3. Using yeast two-hybrid screens, we further discovered that all variants bound dynein light chain Tctex-type 3; however, only variants with an acidic cluster motif bound kinesin light chain 1. Accordingly, splice variants showed markedly different trafficking properties and localized to different subcellular compartments. Taken together, our findings demonstrate the existence of eight functional SorCS2 isoforms with differential capacity for interactions with cytosolic ligands dynein light chain Tctex-type 3 and kinesin light chain 1, which potentially allows cell-type specific SorCS2 trafficking and BDNF signaling.
Collapse
Affiliation(s)
- Sune Skeldal
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | | | - Jonas Lende
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | | | - Simon Mølgaard
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Mathias Kaas
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Perline Demange
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Andreas Høiberg Bentsen
- Department of Surgical Gastroenterology and Transplantation, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Marie Fuglsang
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | | | - Henriette Buttenschøn
- NIDO | Centre for Research and Education, Gødstrup Hospital, Herning, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | | | - Peder Madsen
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark.
| |
Collapse
|
13
|
Kuś J, Saramowicz K, Czerniawska M, Wiese W, Siwecka N, Rozpędek-Kamińska W, Kucharska-Lusina A, Strzelecki D, Majsterek I. Molecular Mechanisms Underlying NMDARs Dysfunction and Their Role in ADHD Pathogenesis. Int J Mol Sci 2023; 24:12983. [PMID: 37629164 PMCID: PMC10454781 DOI: 10.3390/ijms241612983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is one of the most common neurodevelopmental disorders, although the aetiology of ADHD is not yet understood. One proposed theory for developing ADHD is N-methyl-D-aspartate receptors (NMDARs) dysfunction. NMDARs are involved in regulating synaptic plasticity and memory function in the brain. Abnormal expression or polymorphism of some genes associated with ADHD results in NMDAR dysfunction. Correspondingly, NMDAR malfunction in animal models results in ADHD-like symptoms, such as impulsivity and hyperactivity. Currently, there are no drugs for ADHD that specifically target NMDARs. However, NMDAR-stabilizing drugs have shown promise in improving ADHD symptoms with fewer side effects than the currently most widely used psychostimulant in ADHD treatment, methylphenidate. In this review, we outline the molecular and genetic basis of NMDAR malfunction and how it affects the course of ADHD. We also present new therapeutic options related to treating ADHD by targeting NMDAR.
Collapse
Affiliation(s)
- Justyna Kuś
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Kamil Saramowicz
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Maria Czerniawska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Wojciech Wiese
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Natalia Siwecka
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Wioletta Rozpędek-Kamińska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Aleksandra Kucharska-Lusina
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz, Czechoslowacka 8/10, 92-216 Lodz, Poland;
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (J.K.); (K.S.); (M.C.); (W.W.); (N.S.); (W.R.-K.); (A.K.-L.)
| |
Collapse
|
14
|
Li Y, Guan X, He Y, Jia X, Pan L, Wang Y, Han Y, Zhao R, Yang J, Hou T. ProBDNF signaling is involved in periodontitis-induced depression-like behavior in mouse hippocampus. Int Immunopharmacol 2023; 116:109767. [PMID: 36738676 DOI: 10.1016/j.intimp.2023.109767] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/15/2023] [Accepted: 01/19/2023] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Increasing evidence supports the association between periodontitis and depression. However, the specific mechanisms remain to be further elucidated. The present study aimed to mechanistically investigate the regional roles of proBDNF (the precursor of brain-derived neurotrophic factor) in periodontitis induced depression-like behavior in mice. METHODS Experimental periodontitis model was established by periodontal injection of Porphyromonas gingivalis lipopolysaccharide (Pg-LPS) in 8-week-old male Bdnf-HA/HA mice for 3 weeks. The depression-like behaviors, spontaneous exploratory activity and the level of anxiety were assessed by behavior tests. The activation of microglia and astrocytes, as well as the expression of Interleukin (IL)-1β and Tumor necrosis factor (TNF)-α in the hippocampus, prefrontal cortex, and cortex were further assessed by immunofluorescence and western blots. The levels of IL-1β in blood serum and expression of occludin as well as claudin5 in the hippocampus, prefrontal cortex, and cortex were further determined by enzyme-linked immunosorbent assay and western blot. Finally, the expression of proBDNF, its receptors, and mature BDNF (mBDNF), as well as neuronal activity were measured by western blots and immunofluorescence. RESULTS Pg-LPS successfully induced periodontitis in mice and caused obvious depression-like behavior. Furthermore, we observed an increased activation of astrocytes and microglia, as well as a significant increase in expression of IL-1β and TNF-α in the hippocampus of mice treated with Pg-LPS, with elevated level of IL-1β in serum and decreased expression of occludin and claudin5 in the hippocampus. Importantly, we found that the levels of proBDNF and its receptors, SorCS2 and p75NTR, were increased significantly; however, the level of mBDNF was decreased, therefor leading to greater ratio of proBDNF/mBDNF. In addition, we also detected decreased neuronal activity in the hippocampus of mice treated with Pg-LPS. CONCLUSIONS Our results indicate that Pg-LPS-induced periodontitis could cause depression-like behaviors in mice, and the proBDNF signaling is involved in the process.
Collapse
Affiliation(s)
- Yingxue Li
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xiaoyue Guan
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yani He
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Xiangbin Jia
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Lifei Pan
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yuting Wang
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Yue Han
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Rui Zhao
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Jianmin Yang
- Department of Medicine, Weill Cornell Medical School, Cornell University, New York, NY 10065, USA.
| | - Tiezhou Hou
- Key laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Department of Cariology and Endodontics, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| |
Collapse
|
15
|
Lakhssassi K, Sarto MP, Lahoz B, Alabart JL, Folch J, Serrano M, Calvo JH. Blood transcriptome of Rasa Aragonesa rams with different sexual behavior phenotype reveals CRYL1 and SORCS2 as genes associated with this trait. J Anim Sci 2023; 101:skad098. [PMID: 36996265 PMCID: PMC10118393 DOI: 10.1093/jas/skad098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 03/28/2023] [Indexed: 04/01/2023] Open
Abstract
Reproductive fitness of rams is seasonal, showing the highest libido during short days coinciding with the ovarian cyclicity resumption in the ewe. However, the remarkable variation in sexual behavior between rams impair farm efficiency and profitability. Intending to identify in vivo sexual behavior biomarkers that may aid farmers to select active rams, transcriptome profiling of blood was carried out by analyzing samples from 6 sexually active (A) and 6 nonactive (NA) Rasa Aragonesa rams using RNA-Seq technique. A total of 14,078 genes were expressed in blood but only four genes were differentially expressed (FDR < 0.10) in the A vs. NA rams comparison. The genes, acrosin inhibitor 1 (ENSOARG00020023278) and SORCS2, were upregulated (log2FC > 1) in active rams, whereas the CRYL1 and immunoglobulin lambda-1 light chain isoform X47 (ENSOARG00020025518) genes were downregulated (log2FC < -1) in this same group. Gene set Enrichment Analysis (GSEA) identified 428 signaling pathways, predominantly related to biological processes. The lysosome pathway (GO:0005764) was the most enriched, and may affect fertility and sexual behavior, given the crucial role played by lysosomes in steroidogenesis, being the SORCS2 gene related to this signaling pathway. Furthermore, the enriched positive regulation of ERK1 and ERK2 cascade (GO:0070374) pathway is associated with reproductive phenotypes such as fertility via modulation of hypothalamic regulation and GnRH-mediated production of pituitary gonadotropins. Furthermore, external side of plasma membrane (GO:0009897), fibrillar center (GO:0001650), focal adhesion (GO:0005925), and lamellipodium (GO:0030027) pathways were also enriched, suggesting that some molecules of these pathways might also be involved in rams' sexual behavior. These results provide new clues for understanding the molecular regulation of sexual behavior in rams. Further investigations will be needed to confirm the functions of SORCS2 and CRYL1 in relation to sexual behavior.
Collapse
Affiliation(s)
- Kenza Lakhssassi
- Agrifood Research and Technology Centre of Aragon-IA2, 50059 Zaragoza, Spain
- INRA Instituts, 6356 Rabat, Morocco
| | - María Pilar Sarto
- Agrifood Research and Technology Centre of Aragon-IA2, 50059 Zaragoza, Spain
| | - Belén Lahoz
- Agrifood Research and Technology Centre of Aragon-IA2, 50059 Zaragoza, Spain
| | - José Luis Alabart
- Agrifood Research and Technology Centre of Aragon-IA2, 50059 Zaragoza, Spain
| | - José Folch
- Agrifood Research and Technology Centre of Aragon-IA2, 50059 Zaragoza, Spain
| | - Malena Serrano
- Department of Animal Breeding and Genetics, INIA-CSIC, 28040 Madrid, Spain
| | - Jorge Hugo Calvo
- Agrifood Research and Technology Centre of Aragon-IA2, 50059 Zaragoza, Spain
- ARAID, 50018 Zaragoza, Spain
| |
Collapse
|
16
|
Gospodinova KO, Olsen D, Kaas M, Anderson SM, Phillips J, Walker RM, Bermingham ML, Payne AL, Giannopoulos P, Pandya D, Spires-Jones TL, Abbott CM, Porteous DJ, Glerup S, Evans KL. Loss of SORCS2 is Associated with Neuronal DNA Double-Strand Breaks. Cell Mol Neurobiol 2023; 43:237-249. [PMID: 34741697 PMCID: PMC9813074 DOI: 10.1007/s10571-021-01163-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 10/29/2021] [Indexed: 01/09/2023]
Abstract
SORCS2 is one of five proteins that constitute the Vps10p-domain receptor family. Members of this family play important roles in cellular processes linked to neuronal survival, differentiation and function. Genetic and functional studies implicate SORCS2 in cognitive function, as well as in neurodegenerative and psychiatric disorders. DNA damage and DNA repair deficits are linked to ageing and neurodegeneration, and transient neuronal DNA double-strand breaks (DSBs) also occur as a result of neuronal activity. Here, we report a novel role for SORCS2 in DSB formation. We show that SorCS2 loss is associated with elevated DSB levels in the mouse dentate gyrus and that knocking out SORCS2 in a human neuronal cell line increased Topoisomerase IIβ-dependent DSB formation and reduced neuronal viability. Neuronal stimulation had no impact on levels of DNA breaks in vitro, suggesting that the observed differences may not be the result of aberrant neuronal activity in these cells. Our findings are consistent with studies linking the VPS10 receptors and DNA damage to neurodegenerative conditions.
Collapse
Affiliation(s)
- Katerina O. Gospodinova
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Ditte Olsen
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Mathias Kaas
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Susan M. Anderson
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Jonathan Phillips
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Rosie M. Walker
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK ,Present Address: University of Edinburgh, Chancellor’s Building, 49, Edinburgh, EH16 4SB UK
| | - Mairead L. Bermingham
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Abigail L. Payne
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Panagiotis Giannopoulos
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Divya Pandya
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Tara L. Spires-Jones
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, University of Edinburgh, Edinburgh, EH8 9XD UK
| | - Catherine M. Abbott
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - David J. Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| | - Kathryn L. Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, EH4 2XU UK
| |
Collapse
|
17
|
Salasova A, Monti G, Andersen OM, Nykjaer A. Finding memo: versatile interactions of the VPS10p-Domain receptors in Alzheimer’s disease. Mol Neurodegener 2022; 17:74. [PMID: 36397124 PMCID: PMC9673319 DOI: 10.1186/s13024-022-00576-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 10/17/2022] [Indexed: 11/19/2022] Open
Abstract
The family of VPS10p-Domain (D) receptors comprises five members named SorLA, Sortilin, SorCS1, SorCS2 and SorCS3. While their physiological roles remain incompletely resolved, they have been recognized for their signaling engagements and trafficking abilities, navigating a number of molecules between endosome, Golgi compartments, and the cell surface. Strikingly, recent studies connected all the VPS10p-D receptors to Alzheimer’s disease (AD) development. In addition, they have been also associated with diseases comorbid with AD such as diabetes mellitus and major depressive disorder. This systematic review elaborates on genetic, functional, and mechanistic insights into how dysfunction in VPS10p-D receptors may contribute to AD etiology, AD onset diversity, and AD comorbidities. Starting with their functions in controlling cellular trafficking of amyloid precursor protein and the metabolism of the amyloid beta peptide, we present and exemplify how these receptors, despite being structurally similar, regulate various and distinct cellular events involved in AD. This includes a plethora of signaling crosstalks that impact on neuronal survival, neuronal wiring, neuronal polarity, and synaptic plasticity. Signaling activities of the VPS10p-D receptors are especially linked, but not limited to, the regulation of neuronal fitness and apoptosis via their physical interaction with pro- and mature neurotrophins and their receptors. By compiling the functional versatility of VPS10p-D receptors and their interactions with AD-related pathways, we aim to further propel the AD research towards VPS10p-D receptor family, knowledge that may lead to new diagnostic markers and therapeutic strategies for AD patients.
Collapse
|
18
|
Artemieva LE, Mineev KS, Arseniev AS, Goncharuk SA. Expression, purification and characterization of SORCS2 intracellular domain for structural studies. Protein Expr Purif 2022; 193:106058. [PMID: 35114376 DOI: 10.1016/j.pep.2022.106058] [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: 09/28/2021] [Revised: 12/21/2021] [Accepted: 01/28/2022] [Indexed: 10/19/2022]
Abstract
Neurotrophin signaling pathways are one of the major cascades in neuronal development and involved in many key processes including proliferation, differentiation, apoptosis, synaptic plasticity, axonal growth. In addition to the main classes of neurotrophin receptors, Trk and P75NTR, there are many auxiliary proteins, which can also bind neurotrophins and regulate the signaling pathways. The versatility of interactions between them could explain multiple and completely opposite biological outcomes such as cell survival or apoptosis. Membrane protein SorCS2, a vacuolar protein sorting 10 protein-domain receptor, interacts with P75NTR and controls the activity of Trk receptors. The abnormal functioning of SorCS2 is associated with neurodegenerative diseases, such as Alzheimer's and Huntington's disease. But the mechanism of SorCS2 activation and basis of the interaction with P75NTR has remained elusive. Herein, we describe two efficient approaches for the intracellular domain of the SorCS2 production employing bacterial and cell-free expression systems, as well as purification and refolding protocols. Finally, we characterized the purified protein by DLS and NMR and demonstrated that the protein sample is suitable for structural studies.
Collapse
Affiliation(s)
- L E Artemieva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - K S Mineev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - A S Arseniev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - S A Goncharuk
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia.
| |
Collapse
|
19
|
Wang J, Anastasia A, Bains H, Giza JI, Clossey DG, Deng J, Neubert TA, Rice WJ, Lee FS, Hempstead BL, Bracken C. Zinc induced structural changes in the intrinsically disordered BDNF Met prodomain confer synaptic elimination. Metallomics 2021; 12:1208-1219. [PMID: 32744273 DOI: 10.1039/d0mt00108b] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human brain derived neurotrophic factor (BDNF) encodes a protein product consisting of a C-terminal mature domain (mature BDNF) and an N-terminal prodomain, which is an intrinsically disordered protein. A common single nucleotide polymorphism in humans results in a methionine substitution for valine at position 66 of the prodomain, and is associated with memory deficits, depression and anxiety disorders. The BDNF Met66 prodomain, but not the Val66 prodomain, promotes rapid structural remodeling of hippocampal neurons' growth cones and dendritic spines by interacting directly with the SorCS2 receptor. While it has been reported that the Met66 and Val66 prodomains exhibit only modest differences in structural propensities in the apo state, here we show that Val66 and Met66 prodomains differentially bind zinc (Zn). Zn2+ binds with higher affinity and more broadly impacts residues on the Met66 prodomain compared to the Val66 prodomain as shown by NMR and ITC. Zn2+ binding to the Met66 and Val66 prodomains results in distinct conformational and macroscopic differences observed by NMR, light scattering and cryoEM. To determine if Zn2+ mediated conformational change in the Met66 prodomain is required for biological effect, we mutated His40, a Zn2+ binding site, and observed a loss of Met66 prodomain bioactivity. As the His40 site is distant from the known region of the prodomain involved in receptor binding, we suggest that Met66 prodomain bioactivity involves His40 mediated stabilization of the multimeric structure. Our results point to the necessity of a Zn2+-mediated higher order molecular assembly of the Met66 prodomain to mediate neuronal remodeling.
Collapse
Affiliation(s)
- Jing Wang
- Weill Cornell Medicine, Department of Biochemistry, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Greening DW, Notaras M, Chen M, Xu R, Smith JD, Cheng L, Simpson RJ, Hill AF, van den Buuse M. Chronic methamphetamine interacts with BDNF Val66Met to remodel psychosis pathways in the mesocorticolimbic proteome. Mol Psychiatry 2021; 26:4431-4447. [PMID: 31822818 DOI: 10.1038/s41380-019-0617-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 11/17/2019] [Accepted: 11/21/2019] [Indexed: 02/06/2023]
Abstract
Methamphetamine (Meth) abuse has reached epidemic proportions in many countries and can induce psychotic episodes mimicking the clinical profile of schizophrenia. Brain-derived neurotrophic factor (BDNF) is implicated in both Meth effects and schizophrenia. We therefore studied the long-term effects of chronic Meth exposure in transgenic mice engineered to harbor the human BDNFVal66Met polymorphism expressed via endogenous mouse promoters. These mice were chronically treated with an escalating Meth regime during late adolescence. At least 4 weeks later, all hBDNFVal66Met Meth-treated mice exhibited sensitization confirming persistent behavioral effects of Meth. We used high-resolution quantitative mass spectrometry-based proteomics to biochemically map the long-term effects of Meth within the brain, resulting in the unbiased detection of 4808 proteins across the mesocorticolimbic circuitry. Meth differentially altered dopamine signaling markers (e.g., Dat, Comt, and Th) between hBDNFVal/Val and hBDNFMet/Met mice, implicating involvement of BDNF in Meth-induced reprogramming of the mesolimbic proteome. Targeted analysis of 336 schizophrenia-risk genes, as well as 82 growth factor cascade markers, similarly revealed that hBDNFVal66Met genotype gated the recruitment of these factors by Meth in a region-specific manner. Cumulatively, these data represent the first comprehensive analysis of the long-term effects of chronic Meth exposure within the mesocorticolimbic circuitry. In addition, these data reveal that long-term Meth-induced brain changes are strongly dependent upon BDNF genetic variation, illustrating how drug-induced psychosis may be modulated at the molecular level by a single genetic locus.
Collapse
Affiliation(s)
- David W Greening
- Baker Heart and Diabetes Institute, Molecular Proteomics, Melbourne, VIC, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Michael Notaras
- Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Maoshan Chen
- Australian Centre for Blood Diseases (ACBD), Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Rong Xu
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Joel D Smith
- Biological Research Unit, Racing Analytical Services Ltd, Flemington, VIC, Australia
| | - Lesley Cheng
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Richard J Simpson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Andrew F Hill
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, VIC, Australia. .,Department of Pharmacology, University of Melbourne, Melbourne, VIC, Australia. .,College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia.
| |
Collapse
|
21
|
Covaceuszach S, Peche L, Konarev P, Lamba D. A combined evolutionary and structural approach to disclose the primary structural determinants essential for proneurotrophins biological functions. Comput Struct Biotechnol J 2021; 19:2891-2904. [PMID: 34094000 PMCID: PMC8144349 DOI: 10.1016/j.csbj.2021.05.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/03/2021] [Accepted: 05/03/2021] [Indexed: 12/24/2022] Open
Abstract
The neurotrophins, i.e., Nerve Growth Factor (NGF), Brain-Derived Neurotrophic Factor (BDNF), Neurotrophin 3 (NT3) and Neurotrophin 4 (NT4), are known to play a range of crucial functions in the developing and adult peripheral and central nervous systems. Initially synthesized as precursors, i.e., proneurotrophins (proNTs), that are cleaved to release C-terminal mature forms, they act through two types of receptors, the specific Trk receptors (Tropomyosin-related kinases) and the pan-neurotrophin receptor p75NTR, to initiate survival and differentiative responses. Recently, all the proNTs but proNT4 have been demonstrated to be not just inactive precursors, but signaling ligands that mediate opposing actions in fundamental aspects of the nervous system with respect to the mature counterparts through dual-receptor complexes formation with a member of the VPS10 family and p75NTR. Despite the functional relevance, the molecular determinants underpinning the interactions between the pro-domains and their receptors are still elusive probably due to their intrinsically disordered nature. Here we present an evolutionary approach coupled to an experimental study aiming to uncover the structural and dynamical basis of the biological function displayed by proNGF, proBDNF and proNT3 but missing in proNT4. A bioinformatic analysis allowed to elucidate the functional adaptability of the proNTs family in vertebrates, identifying conserved key structural features. The combined biochemical and SAXS experiments shed lights on the structure and dynamic behavior of the human proNTs in solution, giving insights on the evolutionary conserved structural motifs, essential for the multifaceted roles of proNTs in physiological as well as in pathological contexts.
Collapse
Affiliation(s)
- S. Covaceuszach
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Trieste, Italy
| | - L.Y. Peche
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Trieste, Italy
| | - P.V. Konarev
- A.V. Shubnikov Institute of Crystallography of Federal Scientific Research Centre “Crystallography and Photonics” of Russian Academy of Sciences, Moscow, Russia
| | - D. Lamba
- Istituto di Cristallografia, Consiglio Nazionale delle Ricerche, Trieste, Italy
- Interuniversity Consortium “Biostructures and Biosystems National Institute”, Roma, Italy
| |
Collapse
|
22
|
Lanshakov DA, Sukhareva EV, Bulygina VV, Bannova AV, Shaburova EV, Kalinina TS. Single neonatal dexamethasone administration has long-lasting outcome on depressive-like behaviour, Bdnf, Nt-3, p75ngfr and sorting receptors (SorCS1-3) stress reactive expression. Sci Rep 2021; 11:8092. [PMID: 33854153 PMCID: PMC8046778 DOI: 10.1038/s41598-021-87652-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 03/31/2021] [Indexed: 12/22/2022] Open
Abstract
Elevated glucocorticoid level in the early postnatal period is associated with glucocorticoid therapy prescribed at preterm delivery most often has severe long-lasting neurodevelopmental and behavioural effects. Detailed molecular mechanisms of such programming action of antenatal glucocorticoids on behaviour are still poorly understood. To address this question we studied neurotrophins: Bdnf, Nt-3, Ngf and their receptors: p75ngfr, Sorcs3 expression changes after subcutaneous dexamethasone (DEX) 0.2 mg/kg injection to P2 rat pups. Neurotrophins expression level was studied in the hippocampus (HPC). Disturbances in these brain regions have been implicated in the emergence of multiple psychopathologies. p75ngfr and Sorcs3 expression was studied in the brainstem—region where monoamine neurons are located. Immunohistochemically P75NTR protein level changes after DEX were investigated in the brainstem Locus Coereleus norepinephrine neurons (NE). In the first hours after DEX administration elevation of neurotrophins expression in HPC and decline of receptor’s expression in the NE brainstem neurons were observed. Another critical time point during maturation is adolescence. Impact of elevated glucocorticoid level in the neonatal period and unpredictable stress (CMUS) at the end of adolescence on depressive-like behaviour was studied. Single neonatal DEX injection leads to decrease in depressive-like behaviour, observed in FST, independently from chronic stress. Neonatal DEX administration decreased Ntf3 and SorCS1 expression in the brainstem. Also Bdnf mRNA level in the brainstem of these animals didn’t decrease after FST. CMUS at the end of adolescence changed p75ngfr and SorCS3 expression in the brainstem in the animals that received single neonatal DEX administration.
Collapse
Affiliation(s)
- D A Lanshakov
- Laboratory of Postgenomics Neurobiology, Institute of Cytology and Genetics, Russian Academy of Science, Novosibirsk, Russian Federation, 630090.
| | - E V Sukhareva
- Functional Neurogenomics Laboratory, Institute of Cytology and Genetics, Russian Academy of Science, Novosibirsk, Russian Federation, 630090.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russian Federation, 630090
| | - V V Bulygina
- Functional Neurogenomics Laboratory, Institute of Cytology and Genetics, Russian Academy of Science, Novosibirsk, Russian Federation, 630090
| | - A V Bannova
- Functional Neurogenomics Laboratory, Institute of Cytology and Genetics, Russian Academy of Science, Novosibirsk, Russian Federation, 630090
| | - E V Shaburova
- Functional Neurogenomics Laboratory, Institute of Cytology and Genetics, Russian Academy of Science, Novosibirsk, Russian Federation, 630090.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russian Federation, 630090
| | - T S Kalinina
- Functional Neurogenomics Laboratory, Institute of Cytology and Genetics, Russian Academy of Science, Novosibirsk, Russian Federation, 630090.,Department of Natural Sciences, Novosibirsk State University, Novosibirsk, Russian Federation, 630090
| |
Collapse
|
23
|
Yang J, Ma Q, Dincheva I, Giza J, Jing D, Marinic T, Milner TA, Rajadhyaksha A, Lee FS, Hempstead BL. SorCS2 is required for social memory and trafficking of the NMDA receptor. Mol Psychiatry 2021; 26:927-940. [PMID: 31988435 DOI: 10.1038/s41380-020-0650-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 11/21/2019] [Accepted: 12/02/2019] [Indexed: 11/09/2022]
Abstract
Social memory processing requires functional CA2 neurons, however the specific mechanisms that regulate their activity are poorly understood. Here, we document that SorCS2, a member of the family of the Vps10 family of sorting receptors, is highly expressed in pyramidal neurons of CA2, as well as ventral CA1, a circuit implicated in social memory. SorCS2 specifically localizes to the postsynaptic density and endosomes within dendritic spines of CA2 neurons. We have discovered that SorCS2 is a selective regulator of NMDA receptor surface trafficking in hippocampal neurons, without altering AMPA receptor trafficking. In addition, SorCS2 regulates dendritic spine density in CA2 neurons where SorCS2 expression is enriched, but not in dorsal CA1 neurons, which normally express very low levels of this protein. To specifically test the role of SorCS2 in behavior, we generated a novel SorCS2-deficient mouse, and identify a significant social memory deficit, with no change in sociability, olfaction, anxiety, or several hippocampal-dependent behaviors. Mutations in sorCS2 have been associated with bipolar disease, schizophrenia, and attention deficient-hyperactivity disorder, and abnormalities in social memory are core components of these neuropsychiatric conditions. Thus, our findings provide a new mechanism for social memory formation, through regulating synaptic receptor trafficking in pyramidal neurons by SorCS2.
Collapse
Affiliation(s)
- Jianmin Yang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, PR China. .,Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY, 10065, USA.
| | - Qian Ma
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Iva Dincheva
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Joanna Giza
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, 10065, USA.,Department of Science, Borough of Manhattan Community College, The City University of New York, 199 Chambers Street N699J, New York, NY, 10007, USA
| | - Deqiang Jing
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Tina Marinic
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Teresa A Milner
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.,Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Avenue, New York, NY, 10065, USA
| | | | - Francis S Lee
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Barbara L Hempstead
- Division of Hematology and Medical Oncology, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
24
|
Olsen D, Wellner N, Kaas M, de Jong IEM, Sotty F, Didriksen M, Glerup S, Nykjaer A. Altered dopaminergic firing pattern and novelty response underlie ADHD-like behavior of SorCS2-deficient mice. Transl Psychiatry 2021; 11:74. [PMID: 33495438 PMCID: PMC7835366 DOI: 10.1038/s41398-021-01199-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 12/12/2020] [Accepted: 01/05/2021] [Indexed: 01/03/2023] Open
Abstract
Attention deficit hyperactivity disorder (ADHD) is the most frequently diagnosed neurodevelopmental disorder worldwide. Affected individuals present with hyperactivity, inattention, and cognitive deficits and display a characteristic paradoxical response to drugs affecting the dopaminergic system. However, the underlying pathophysiology of ADHD and how this relates to dopaminergic transmission remains to be fully understood. Sorcs2-/- mice uniquely recapitulate symptoms reminiscent of ADHD in humans. Here, we show that lack of SorCS2 in mice results in lower sucrose intake, indicating general reward deficits. Using in-vivo recordings, we further find that dopaminergic transmission in the ventral tegmental area (VTA) is shifted towards a more regular firing pattern with marked reductions in the relative occurrence of irregular firing in Sorcs2-/- mice. This was paralleled by abnormal acute behavioral responses to dopamine receptor agonists, suggesting fundamental differences in dopaminergic circuits and indicating a perturbation in the balance between the activities of the postsynaptic dopamine receptor DRD1 and the presynaptic inhibitory autoreceptor DRD2. Interestingly, the hyperactivity and drug response of Sorcs2-/- mice were markedly affected by novelty. Taken together, our findings show how loss of a candidate ADHD-risk gene has marked effects on dopaminergic circuit function and the behavioral response to the environment.
Collapse
Affiliation(s)
- Ditte Olsen
- grid.7048.b0000 0001 1956 2722Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000 Aarhus C, Denmark ,grid.424580.f0000 0004 0476 7612Neurodegeneration and Biologics, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark ,grid.7048.b0000 0001 1956 2722Present Address: Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000 Aarhus C, Denmark
| | - Niels Wellner
- grid.7048.b0000 0001 1956 2722Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000 Aarhus C, Denmark ,grid.7048.b0000 0001 1956 2722Danish Research Institute of Translational Neuroscience DANDRITE Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000 Aarhus C, Denmark
| | - Mathias Kaas
- grid.7048.b0000 0001 1956 2722Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000 Aarhus C, Denmark
| | - Inge E. M. de Jong
- grid.424580.f0000 0004 0476 7612Neurodegeneration and Biologics, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark
| | - Florence Sotty
- grid.424580.f0000 0004 0476 7612Neurodegeneration and Biologics, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark
| | - Michael Didriksen
- grid.424580.f0000 0004 0476 7612Neurodegeneration and Biologics, H. Lundbeck A/S, Ottiliavej 9, DK-2500 Valby, Denmark
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000, Aarhus C, Denmark.
| | - Anders Nykjaer
- Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000, Aarhus C, Denmark. .,Danish Research Institute of Translational Neuroscience DANDRITE Nordic-EMBL Partnership, Department of Biomedicine, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000, Aarhus C, Denmark. .,The Danish National Research Foundation Center PROMEMO, Aarhus University, Hoegh-Guldbergsgade 10, DK-8000, Aarhus C, Denmark. .,Department of Neurosurgery, Skejby University Hospital, Palle Juul-Jensens Blvd. 99, DK-8200, Aarhus N, Denmark.
| |
Collapse
|
25
|
VPS10P Domain Receptors: Sorting Out Brain Health and Disease. Trends Neurosci 2020; 43:870-885. [DOI: 10.1016/j.tins.2020.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/23/2020] [Accepted: 08/11/2020] [Indexed: 12/13/2022]
|
26
|
Malik AR, Szydlowska K, Nizinska K, Asaro A, van Vliet EA, Popp O, Dittmar G, Fritsche-Guenther R, Kirwan JA, Nykjaer A, Lukasiuk K, Aronica E, Willnow TE. SorCS2 Controls Functional Expression of Amino Acid Transporter EAAT3 and Protects Neurons from Oxidative Stress and Epilepsy-Induced Pathology. Cell Rep 2020; 26:2792-2804.e6. [PMID: 30840898 PMCID: PMC6410498 DOI: 10.1016/j.celrep.2019.02.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 01/20/2019] [Accepted: 02/07/2019] [Indexed: 01/05/2023] Open
Abstract
VPS10P domain receptors emerge as central regulators of intracellular protein sorting in neurons with relevance for various brain pathologies. Here, we identified a role for the family member SorCS2 in protection of neurons from oxidative stress and epilepsy-induced cell death. We show that SorCS2 acts as sorting receptor that sustains cell surface expression of the neuronal amino acid transporter EAAT3 to facilitate import of cysteine, required for synthesis of the reactive oxygen species scavenger glutathione. Lack of SorCS2 causes depletion of EAAT3 from the plasma membrane and impairs neuronal cysteine uptake. As a consequence, SorCS2-deficient mice exhibit oxidative brain damage that coincides with enhanced neuronal cell death and increased mortality during epilepsy. Our findings highlight a protective role for SorCS2 in neuronal stress response and provide a possible explanation for upregulation of this receptor seen in surviving neurons of the human epileptic brain.
Collapse
Affiliation(s)
- Anna R Malik
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany.
| | - Kinga Szydlowska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Karolina Nizinska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Antonino Asaro
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Erwin A van Vliet
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, 1105AZ Amsterdam, the Netherlands; Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH, Amsterdam, the Netherlands
| | - Oliver Popp
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Gunnar Dittmar
- Department of Oncology, Luxembourg Institute of Health, 1445 Strassen, Luxembourg
| | - Raphaela Fritsche-Guenther
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany; Berlin Institute of Health Metabolomics Platform, 10178 Berlin, Germany
| | - Jennifer A Kirwan
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany; Berlin Institute of Health Metabolomics Platform, 10178 Berlin, Germany
| | - Anders Nykjaer
- MIND Center, Danish Research Institute of Translational Neuroscience - DANDRITE, The Danish Research Foundation Center PROMEMO, Departments of Biomedicine, Aarhus University, and Neurosurgery, Aarhus University Hospital, 8000C Aarhus, Denmark
| | - Katarzyna Lukasiuk
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, 1105AZ Amsterdam, the Netherlands; Stichting Epilepsie Instellingen Nederland (SEIN), 2103 SW Heemstede, the Netherlands
| | - Thomas E Willnow
- Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany.
| |
Collapse
|
27
|
Huang K, Crist AM, Patel NR, Blanks A, Carter K, Cleaver O, Meadows SM. Annexin A3 is necessary for parallel artery-vein alignment in the mouse retina. Dev Dyn 2020; 249:666-678. [PMID: 32020697 PMCID: PMC7995330 DOI: 10.1002/dvdy.154] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/31/2019] [Accepted: 01/23/2020] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Annexin A3 (Anxa3) is a member of the calcium-regulated, cell membrane-binding family of annexin proteins. We previously confirmed that Anxa3 is expressed in the endothelial lineage in vertebrates and that loss of anxa3 in Xenopus laevis leads to embryonic blood vessel defects. However, the biological function of Anxa3 in mammals is completely unknown. In order to investigate Anxa3 vascular function in mammals, we generated an endothelial cell-specific Anxa3 conditional knockout mouse model (Anxa3f/f ;Tie2-Cre). RESULTS Anxa3f/f ;Tie2-Cre mice are born at Mendelian ratios and display morphologically normal blood vessels during development. However, loss of Anxa3 leads to artery-vein (AV) misalignment characterized by atypical AV crossovers in the postnatal and adult retina. CONCLUSIONS Anxa3 is not essential for embryonic blood vessel formation but is required for proper parallel AV alignment in the murine retina. AV crossovers associated with Anxa3f/f ;Tie2-Cre mice are similar to AV intersections observed in patients with branch retinal vein occlusion (BRVO), although we did not observe occluded vessels. This new Anxa3 mouse model may provide a basis for understanding AV crossover formation associated with BRVO.
Collapse
Affiliation(s)
- Katie Huang
- Cell and Molecular Biology Department, Tulane University, New Orleans, Louisiana
| | - Angela M. Crist
- Cell and Molecular Biology Department, Tulane University, New Orleans, Louisiana
| | - Nehal R. Patel
- Cell and Molecular Biology Department, Tulane University, New Orleans, Louisiana
| | - Avery Blanks
- Cell and Molecular Biology Department, Tulane University, New Orleans, Louisiana
| | - Kelsey Carter
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ondine Cleaver
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Stryder M. Meadows
- Cell and Molecular Biology Department, Tulane University, New Orleans, Louisiana
| |
Collapse
|
28
|
Abstract
The brain-derived neurotrophic factor (BDNF) is a secretory growth factor that promotes neuronal proliferation and survival, synaptic plasticity and long-term potentiation in the central nervous system. Brain-derived neurotrophic factor biosynthesis and secretion are chrono-topically regulated processes at the cellular level, accounting for specific localizations and functions. Given its role in regulating brain development and activity, BDNF represents a potentially relevant gene for schizophrenia, and indeed BDNF and its non-synonymous functional variant, rs6265 (C → T, Val → Met) have been widely studied in psychiatric genetics. Human and animal studies have indicated that brain-derived neurotrophic factor is relevant for schizophrenia-related phenotypes, and that: (1) fine-tuned regulation of brain-derived neurotrophic factor secretion and activity is necessary to guarantee brain optimal development and functioning; (2) the Val → Met substitution is associated with impaired activity-dependent secretion of brain-derived neurotrophic factor; (3) disruption of brain-derived neurotrophic factor signaling is associated with altered synaptic plasticity and neurodevelopment. However, genome-wide association studies failed to associate the BDNF locus with schizophrenia, even though a sub-threshold association exists. Here, we will review studies focused on the relationship between the genetic variation of BDNF and schizophrenia, trying to fill the gap between genetic risk per se and insights from molecular biology. A deeper understanding of brain-derived neurotrophic factor biology and of the epigenetic regulation of brain-derived neurotrophic factor and its interactome during development may help clarifying the potential role of this gene in schizophrenia, thus informing development of brain-derived neurotrophic factor-based strategies of prevention and treatment of this disorder.
Collapse
|
29
|
Wang Z, Zhong XL, Xu Y, He J, Liu ZH, Nai AT, Niu L, Luo SS, Yang H, Zeng JY, He SY, Chen X, Wan W, Cao WY. Irradiation increases brain-derived neurotrophic factor precursor signaling in the mouse hippocampus. Neurobiol Learn Mem 2020; 171:107186. [PMID: 32084558 DOI: 10.1016/j.nlm.2020.107186] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 02/05/2020] [Accepted: 02/10/2020] [Indexed: 01/06/2023]
Affiliation(s)
- Zhen Wang
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China
| | - Xiao Lin Zhong
- Institute of Clinical Medicine, The First Affiliated Hospital of University of South China, 421001 Hengyang, Hunan, China
| | - Yang Xu
- Institute of Neuroscience, Medical College, University of South China, 421001 Hengyang, Hunan, China
| | - Jie He
- Department of Pathology, Medical College, University of South China, 421001 Hengyang, Hunan, China
| | - Zheng Hai Liu
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China
| | - Ai Tao Nai
- Department of Radiation Oncology, The First Affiliated Hospital of University of South China, 421001 Hengyang, Hunan, China
| | - Lei Niu
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China
| | - Shi Shi Luo
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China
| | - Hui Yang
- Department of Pathology, Medical College, University of South China, 421001 Hengyang, Hunan, China
| | - Jia Yu Zeng
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China
| | - Shu Ya He
- School of Public Health, University of South China, 421001 Hengyang, Hunan, China
| | - Xi Chen
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China
| | - Wei Wan
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China; Key Laboratory of Brain Science Research & Transformation In Tropical Environment of Hainan Province, Hainan Medical University, 571199 Haikou, China.
| | - Wen Yu Cao
- Clinical Anatomy & Reproductive Medicine Application Institute, School of Medicine, University of South China, 421001 Hengyang, Hunan, China.
| |
Collapse
|
30
|
Malik AR, Lips J, Gorniak-Walas M, Broekaart DWM, Asaro A, Kuffner MTC, Hoffmann CJ, Kikhia M, Dopatka M, Boehm-Sturm P, Mueller S, Dirnagl U, Aronica E, Harms C, Willnow TE. SorCS2 facilitates release of endostatin from astrocytes and controls post-stroke angiogenesis. Glia 2020; 68:1304-1316. [PMID: 31898841 DOI: 10.1002/glia.23778] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 01/26/2023]
Abstract
SorCS2 is an intracellular sorting receptor of the VPS10P domain receptor gene family recently implicated in oxidative stress response. Here, we interrogated the relevance of stress-related activities of SorCS2 in the brain by exploring its role in ischemic stroke in mouse models and in patients. Although primarily seen in neurons in the healthy brain, expression of SorCS2 was massively induced in astrocytes surrounding the ischemic core in mice following stroke. Post-stroke induction was likely a result of increased levels of transforming growth factor β1 in damaged brain tissue, inducing Sorcs2 gene transcription in astrocytes but not neurons. Induced astrocytic expression of SorCS2 was also seen in stroke patients, substantiating the clinical relevance of this observation. In astrocytes in vitro and in the mouse brain in vivo, SorCS2 specifically controlled release of endostatin, a factor linked to post-stroke angiogenesis. The ability of astrocytes to release endostatin acutely after stroke was lost in mice deficient for SorCS2, resulting in a blunted endostatin response which coincided with impaired vascularization of the ischemic brain. Our findings identified activated astrocytes as a source for endostatin in modulation of post-stroke angiogenesis, and the importance of the sorting receptor SorCS2 in this brain stress response.
Collapse
Affiliation(s)
- Anna R Malik
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany.,Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Janet Lips
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany.,Berlin Institute of Health, QUEST Centre for Transforming Biomedical Research, Berlin, Germany
| | | | - Diede W M Broekaart
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Antonino Asaro
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Melanie T C Kuffner
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany
| | - Christian J Hoffmann
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany
| | - Majed Kikhia
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Einstein Center for Neurosciences Berlin, Berlin, Germany
| | - Monika Dopatka
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany
| | - Philipp Boehm-Sturm
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Charité Core Facility 7T Experimental MRIs, Berlin, Germany
| | - Susanne Mueller
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Charité Core Facility 7T Experimental MRIs, Berlin, Germany
| | - Ulrich Dirnagl
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany.,Berlin Institute of Health, QUEST Centre for Transforming Biomedical Research, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Einstein Center for Neurosciences Berlin, Berlin, Germany.,German Centre for Neurodegenerative Diseases, Berlin, Germany
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, the Netherlands.,Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, The Netherlands
| | - Christoph Harms
- Department of Experimental Neurology, Charité-Universitätsmedizin Berlin and Berlin Institute of Health, Neurocure Cluster of Excellence, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Center for Stroke Research Berlin, Berlin, Germany.,Berlin Institute of Health, QUEST Centre for Transforming Biomedical Research, Berlin, Germany.,Charité-Universitätsmedizin Berlin, Einstein Center for Neurosciences Berlin, Berlin, Germany
| | | |
Collapse
|
31
|
Rohde PD, Jensen IR, Sarup PM, Ørsted M, Demontis D, Sørensen P, Kristensen TN. Genetic Signatures of Drug Response Variability in Drosophila melanogaster. Genetics 2019; 213:633-650. [PMID: 31455722 PMCID: PMC6781897 DOI: 10.1534/genetics.119.302381] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/26/2019] [Indexed: 12/27/2022] Open
Abstract
Knowledge of the genetic basis underlying variation in response to environmental exposures or treatments is important in many research areas. For example, knowing the set of causal genetic variants for drug responses could revolutionize personalized medicine. We used Drosophila melanogaster to investigate the genetic signature underlying behavioral variability in response to methylphenidate (MPH), a drug used in the treatment of attention-deficit/hyperactivity disorder. We exposed a wild-type D. melanogaster population to MPH and a control treatment, and observed an increase in locomotor activity in MPH-exposed individuals. Whole-genome transcriptomic analyses revealed that the behavioral response to MPH was associated with abundant gene expression alterations. To confirm these patterns in a different genetic background and to further advance knowledge on the genetic signature of drug response variability, we used a system of inbred lines, the Drosophila Genetic Reference Panel (DGRP). Based on the DGRP, we showed that the behavioral response to MPH was strongly genotype-dependent. Using an integrative genomic approach, we incorporated known gene interactions into the genomic analyses of the DGRP, and identified putative candidate genes for variability in drug response. We successfully validated 71% of the investigated candidate genes by gene expression knockdown. Furthermore, we showed that MPH has cross-generational behavioral and transcriptomic effects. Our findings establish a foundation for understanding the genetic mechanisms driving genotype-specific responses to medical treatment, and highlight the opportunities that integrative genomic approaches have in optimizing medical treatment of complex diseases.
Collapse
Affiliation(s)
- Palle Duun Rohde
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, 8000 Aarhus C, Denmark
- Center for Integrative Sequencing, Aarhus University, 8000, Denmark
| | - Iben Ravnborg Jensen
- Section for Biology and Environmental Science, Department of Chemistry and Bioscience, Aalborg University, 9220, Denmark
| | - Pernille Merete Sarup
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
| | - Michael Ørsted
- Section for Biology and Environmental Science, Department of Chemistry and Bioscience, Aalborg University, 9220, Denmark
| | - Ditte Demontis
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, 8000 Aarhus C, Denmark
- Center for Integrative Sequencing, Aarhus University, 8000, Denmark
- Department of Biomedicine, Aarhus University, 8000, Denmark
| | - Peter Sørensen
- Center for Quantitative Genetics and Genomics, Department of Molecular Biology and Genetics, Aarhus University, 8830 Tjele, Denmark
| | - Torsten Nygaard Kristensen
- Section for Biology and Environmental Science, Department of Chemistry and Bioscience, Aalborg University, 9220, Denmark
- Section for Genetics, Ecology and Evolution, Department of Bioscience, Aarhus University, 8000, Denmark
| |
Collapse
|
32
|
Fast-diffusing p75 NTR monomers support apoptosis and growth cone collapse by neurotrophin ligands. Proc Natl Acad Sci U S A 2019; 116:21563-21572. [PMID: 31515449 PMCID: PMC6815156 DOI: 10.1073/pnas.1902790116] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Neurotrophins (NTs) are homodimeric growth factors displaying fundamental roles in the nervous system. Their activity stems from binding and activation of 3 different receptor types in nervous cell membranes. The p75 NT receptor (p75NTR) was the first to be discovered in 1986; nevertheless, for the numerous structural and functional facets so far reported, its activation mechanisms have remained elusive. Here, we demonstrate that its pleiotropic functions are regulated by different redistributions of the receptors, which crucially depend on the available NT and on the involved subcellular compartment but are unrelated to its oligomerization state. Single-particle studies proved receptors to be monomers with a fast-diffusive behavior in the membrane with, at most, transient self-interactions on the millisecond time scale. The p75 neurotrophin (NT) receptor (p75NTR) plays a crucial role in balancing survival-versus-death decisions in the nervous system. Yet, despite 2 decades of structural and biochemical studies, a comprehensive, accepted model for p75NTR activation by NT ligands is still missing. Here, we present a single-molecule study of membrane p75NTR in living cells, demonstrating that the vast majority of receptors are monomers before and after NT activation. Interestingly, the stoichiometry and diffusion properties of the wild-type (wt) p75NTR are almost identical to those of a receptor mutant lacking residues previously believed to induce oligomerization. The wt p75NTR and mutated (mut) p75NTR differ in their partitioning in cholesterol-rich membrane regions upon nerve growth factor (NGF) stimulation: We argue that this is the origin of the ability of wt p75NTR , but not of mut p75NTR, to mediate immature NT (proNT)-induced apoptosis. Both p75NTR forms support proNT-induced growth cone retraction: We show that receptor surface accumulation is the driving force for cone collapse. Overall, our data unveil the multifaceted activity of the p75NTR monomer and let us provide a coherent interpretative frame of existing conflicting data in the literature.
Collapse
|
33
|
Olsen D, Kaas M, Lundhede J, Molgaard S, Nykjær A, Kjolby M, Østergaard SD, Glerup S. Reduced Alcohol Seeking and Withdrawal Symptoms in Mice Lacking the BDNF Receptor SorCS2. Front Pharmacol 2019; 10:499. [PMID: 31156431 PMCID: PMC6533533 DOI: 10.3389/fphar.2019.00499] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 04/18/2019] [Indexed: 11/13/2022] Open
Abstract
Alcohol use disorder (AUD) is characterized by repetitive and uncontrolled intake of alcohol with severe consequences for affected individuals, their families and society as a whole. Numerous studies have implicated brain-derived neurotrophic factor (BDNF) activity in the neurobiology underlying AUD. The BDNF signaling mechanism is complex and depends on two receptor systems, TrkB and p75NTR, which appear to have opposite effects on alcohol seeking behavior in animal models. We recently discovered that the sortilin-related receptor SorCS2 forms complexes with both TrkB and p75NTR and is important for BDNF activity in the developing and adult CNS. Moreover, the SORCS2 gene was recently identified as the top association signal for severity of alcohol withdrawal symptoms. Hence, we speculated that SorCS2 deficient mice would have an altered response to alcohol. The role of SorCS2 in the acute and adapted response to alcohol was therefore investigated by comparing SorCS2 knockout (Sorcs2-/- ) mice to wild type (WT) mice in three paradigms modeling alcohol sensitivity and consumption; alcohol-induced conditioned place preference, two-bottle choice test as well as the behavioral response to alcohol withdrawal. We found that, when compared to the WT mice, (I) Sorcs2-/- mice displayed complete lack of alcohol-induced place preference, (II) when given free choice between water and alcohol, Sorcs2-/- mice consumed less alcohol, and (III) Sorcs2-/- mice showed no handling-induced convulsion in response to alcohol withdrawal following extended alcohol exposure. Taken together, these results show that lack of the alcohol withdrawal risk gene Sorcs2 results in abnormal behavioral response to alcohol in mice. Consequently, SorCS2 may play an important role in the molecular pathways underlying AUD and complications associated with alcohol withdrawal.
Collapse
Affiliation(s)
- Ditte Olsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Mathias Kaas
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jesper Lundhede
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Simon Molgaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Anders Nykjær
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Nordic EMBL Partnership for Molecular Medicine, DANDRITE - Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark.,Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark.,The Danish National Research Foundation Center PROMEMO, Aarhus University, Aarhus, Denmark
| | - Mads Kjolby
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Nordic EMBL Partnership for Molecular Medicine, DANDRITE - Danish Research Institute of Translational Neuroscience, Aarhus University, Aarhus, Denmark.,Danish Diabetes Academy, Novo Nordisk Foundation, Aarhus, Denmark
| | - Søren D Østergaard
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark.,Department of Affective Disorders, Aarhus University Hospital - Psychiatry, Aarhus, Denmark.,The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
34
|
De Vincenti AP, Ríos AS, Paratcha G, Ledda F. Mechanisms That Modulate and Diversify BDNF Functions: Implications for Hippocampal Synaptic Plasticity. Front Cell Neurosci 2019; 13:135. [PMID: 31024262 PMCID: PMC6465932 DOI: 10.3389/fncel.2019.00135] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 03/19/2019] [Indexed: 11/16/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a neurotrophin that has pleiotropic effects on neuronal morphology and synaptic plasticity that underlie hippocampal circuit development and cognition. Recent advances established that BDNF function is controlled and diversified by molecular and cellular mechanisms including trafficking and subcellular compartmentalization of different Bdnf mRNA species, pre- vs. postsynaptic release of BDNF, control of BDNF signaling by tropomyosin receptor kinase B (TrkB) receptor interactors and conversion of pro-BDNF to mature BDNF and BDNF-propeptide. Defects in these regulatory mechanisms affect dendritic spine formation and morphology of pyramidal neurons as well as synaptic integration of newborn granule cells (GCs) into preexisting circuits of mature hippocampus, compromising the cognitive function. Here, we review recent findings describing novel dynamic mechanisms that diversify and locally control the function of BDNF in hippocampal neurons.
Collapse
Affiliation(s)
- Ana Paula De Vincenti
- División de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Antonella S Ríos
- División de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina.,Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Gustavo Paratcha
- División de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Fernanda Ledda
- División de Neurociencia Molecular y Celular, Instituto de Biología Celular y Neurociencias, Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina.,Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires, CONICET, Buenos Aires, Argentina
| |
Collapse
|
35
|
Martin LJ, Pilipenko V, Benson DW. Role of Segregation for Variant Discovery in Multiplex Families Ascertained by Probands With Left Sided Cardiovascular Malformations. Front Genet 2019; 9:729. [PMID: 30687393 PMCID: PMC6336695 DOI: 10.3389/fgene.2018.00729] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 12/22/2018] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular malformations (CVM) are common birth defects (incidence of 2-5/100 live births). Although a genetic basis is established, in most cases the cause remains unknown. Analysis of whole exome sequencing (WES) in left sided CVM case and trio series has identified large numbers of potential variants but evidence of causality has remained elusive except in a small percentage of cases. We sought to determine whether variant segregation in families would aid in novel gene discovery. The objective was to compare conventional and co-segregation approaches for WES in multiplex families. WES was performed on 52 individuals from 4 multiplex families ascertained by probands with hypoplastic left heart syndrome (HLHS). We identified rare variants with informatics support (RVIS, minor allele frequency ≤0.01 and Combined Annotation Dependent Depletion score ≥20) in probands. Non-RVIS variants did not meet these criteria. Family specific two point logarithm of the odds (LOD) scores identified co-segregating variants (C-SV) using a dominant model and 80% penetrance. In families, 702 RVIS in 668 genes were identified, but only 1 RVIS was also a C-SV (LOD ≥ 1). On the other hand, there were 109 non-RVIS variants with LOD ≥ 1. Among 110 C-SV, 97% were common (MAF > 1%). These results suggest that conventional variant identification methods focused on RVIS, miss most C-SV. For diseases such as left sided CVM, which exhibit strong familial transmission, co-segregation can identify novel candidates.
Collapse
Affiliation(s)
- Lisa J Martin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati School of Medicine, Cincinnati, OH, United States
| | - Valentina Pilipenko
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - D Woodrow Benson
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
36
|
Smith AH, Ovesen PL, Skeldal S, Yeo S, Jensen KP, Olsen D, Diazgranados N, Zhao H, Farrer LA, Goldman D, Glerup S, Kranzler HR, Nykjær A, Gelernter J. Risk Locus Identification Ties Alcohol Withdrawal Symptoms to SORCS2. Alcohol Clin Exp Res 2018; 42:2337-2348. [PMID: 30252935 PMCID: PMC6317871 DOI: 10.1111/acer.13890] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 09/06/2018] [Indexed: 01/11/2023]
Abstract
BACKGROUND Efforts to promote the cessation of harmful alcohol use are hindered by the affective and physiological components of alcohol withdrawal (AW), which can include life-threatening seizures. Although previous studies of AW and relapse have highlighted the detrimental role of stress, little is known about genetic risk factors. METHODS We conducted a genome-wide association study of AW symptom count in uniformly assessed subjects with histories of serious AW, followed by additional genotyping in independent AW subjects. RESULTS The top association signal for AW severity was in sortilin family neurotrophin receptor gene SORCS2 on chromosome 4 (European American meta-analysis n = 1,478, p = 4.3 × 10-9 ). There were no genome-wide significant findings in African Americans (n = 1,231). Bioinformatic analyses were conducted using publicly available high-throughput transcriptomic and epigenomic data sets, showing that in humans SORCS2 is most highly expressed in the nervous system. The identified SORCS2 risk haplotype is predicted to disrupt a stress hormone-modulated regulatory element that has tissue-specific activity in human hippocampus. We used human neural lineage cells to demonstrate in vitro a causal relationship between stress hormone levels and SORCS2 expression, and show that SORCS2 levels in culture are increased upon ethanol exposure and withdrawal. CONCLUSIONS Taken together, these findings indicate that the pathophysiology of withdrawal may involve the effects of stress hormones on neurotrophic factor signaling. Further investigation of these pathways could produce new approaches to managing the aversive consequences of abrupt alcohol cessation.
Collapse
Affiliation(s)
- Andrew H. Smith
- Interdepartmental Neuroscience Program and Medical Scientist Training Program, Yale School of Medicine
- Division of Human Genetics, Department of Psychiatry, VA CT Healthcare Center and Yale School of Medicine
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Peter L. Ovesen
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE - Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Sune Skeldal
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE - Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Seungeun Yeo
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism
| | - Kevin P. Jensen
- Division of Human Genetics, Department of Psychiatry, VA CT Healthcare Center and Yale School of Medicine
| | - Ditte Olsen
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE - Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Nancy Diazgranados
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health
| | - Lindsay A. Farrer
- Departments of Medicine (Biomedical Genetics), Neurology, and Ophthalmology, School of Medicine, and Departments of Biostatistics and Epidemiology, School of Public Health, Boston University, Boston, MA 02118, USA
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| | - Simon Glerup
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE - Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Henry R. Kranzler
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania and Corporal Michael J. Crescenz VAMC, Philadelphia, Pennsylvania 19104, USA
| | - Anders Nykjær
- The Lundbeck Foundation Research Center MIND, Danish Research Institute of Translational Neuroscience DANDRITE - Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, DK-8000 Aarhus C, Denmark
- Department of Neuroscience, Mayo Clinic, Jacksonville 32224, Florida, USA
| | - Joel Gelernter
- Division of Human Genetics, Department of Psychiatry, VA CT Healthcare Center and Yale School of Medicine
- Departments of Genetics and Neuroscience, Yale School of Medicine, Yale University, New Haven, Connecticut 06510, USA
| |
Collapse
|
37
|
Structural insights into SorCS2-Nerve Growth Factor complex formation. Nat Commun 2018; 9:2979. [PMID: 30061605 PMCID: PMC6065357 DOI: 10.1038/s41467-018-05405-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 07/04/2018] [Indexed: 01/09/2023] Open
Abstract
Signaling of SorCS receptors by proneurotrophin ligands regulates neuronal plasticity, induces apoptosis and is associated with mental disorders. The detailed structure of SorCS2 and its extracellular specificity are unresolved. Here we report crystal structures of the SorCS2–NGF complex and unliganded SorCS2 ectodomain, revealing cross-braced SorCS2 homodimers with two NGF dimers bound in a 2:4 stoichiometry. Five out of six SorCS2 domains directly contribute to dimer formation and a C-terminal membrane proximal unreported domain, with an RNA recognition motif fold, locks the dimer in an intermolecular head-to-tail interaction. The complex structure shows an altered SorCS2 conformation indicating substantial structural plasticity. Both NGF dimer chains interact exclusively with the top face of a SorCS2 β-propeller. Biophysical experiments reveal that NGF, proNGF, and proBDNF bind at this site on SorCS2. Taken together, our data reveal a structurally flexible SorCS2 receptor that employs the large β-propeller as a ligand binding platform. The Sortilin-related CNS-expressed receptor 2 (SorCS2)–proneurotrophin signaling system regulates neuronal plasticity and its dysfunction is linked to schizophrenia. Here the authors present the structures of the SorCS2 ectodomain alone and in complex with Nerve Growth Factor, which provides insights into SorCS2 ligand binding and signaling.
Collapse
|
38
|
Giza JI, Kim J, Meyer HC, Anastasia A, Dincheva I, Zheng CI, Lopez K, Bains H, Yang J, Bracken C, Liston C, Jing D, Hempstead BL, Lee FS. The BDNF Val66Met Prodomain Disassembles Dendritic Spines Altering Fear Extinction Circuitry and Behavior. Neuron 2018; 99:163-178.e6. [PMID: 29909994 DOI: 10.1016/j.neuron.2018.05.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/18/2018] [Accepted: 05/15/2018] [Indexed: 11/16/2022]
Abstract
A human variant in the BDNF gene (Val66Met; rs6265) is associated with impaired fear extinction. Using super-resolution imaging, we demonstrate that the BDNF Met prodomain disassembles dendritic spines and eliminates synapses in hippocampal neurons. In vivo, ventral CA1 (vCA1) hippocampal neurons undergo similar morphological changes dependent on their transient co-expression of a SorCS2/p75NTR receptor complex during peri-adolescence. BDNF Met prodomain infusion into the vCA1 during this developmental time frame reduces dendritic spine density and prelimbic (PL) projections, impairing cued fear extinction. Adolescent BdnfMet/Met mice display similar spine and PL innervation deficits. Using fiber photometry, we found that, in wild-type mice, vCA1 neurons projecting to the PL encode extinction by enhancing neural activity in threat anticipation and rapidly subsiding their response. This adaptation is absent in BDNFMet/Met mice. We conclude that the BDNF Met prodomain renders vCA1-PL projection neurons underdeveloped, preventing their capacity for subsequent circuit modulation necessary for fear extinction. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Joanna I Giza
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jihye Kim
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Heidi C Meyer
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Agustin Anastasia
- Instituto de Investigación Médica Mercedes y Martín Ferreyra (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
| | - Iva Dincheva
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
| | - Crystal I Zheng
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Katherine Lopez
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Henrietta Bains
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Jianmin Yang
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA; Key Laboratory of Shaanxi Province Department for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, PR China
| | - Clay Bracken
- Department of Biochemistry, Weill Cornell Medicine, New York, NY 10065, USA
| | - Conor Liston
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA
| | - Deqiang Jing
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Francis S Lee
- Department of Psychiatry, Weill Cornell Medicine, New York, NY 10065, USA; Sackler Institute for Developmental Psychobiology, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
39
|
Højland Knudsen C, Ásgrímsdóttir ES, Rahimi K, Gill KP, Frandsen S, Hvolbøl Buchholdt S, Chen M, Kjems J, Febbraro F, Denham M. A Modified Monomeric Red Fluorescent Protein Reporter for Assessing CRISPR Activity. Front Cell Dev Biol 2018; 6:54. [PMID: 29868584 PMCID: PMC5962823 DOI: 10.3389/fcell.2018.00054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Accepted: 04/25/2018] [Indexed: 01/01/2023] Open
Abstract
Gene editing in human embryonic stem cells (hESCs) has been significantly enhanced by the discovery and development of CRISPR Cas9, a programmable nuclease system that can introduce targeted double-stranded breaks. The system relies on the optimal selection of a sgRNA sequence with low off-targets and high efficiency. We designed an improved monomeric red fluorescent protein reporter, GEmCherry2, for assessing CRISPR Cas9 activity and for optimizing sgRNA. By incorporating an out-of-frame sequence to the N-terminal of the red fluorescent protein mCherry, we created a visual tool for assessing the indel frequency after cutting with CRISPR Cas9. When a sgRNA-Cas9 construct is co-transfected with a corresponding GEmCherry2 construct, single nucleotide indels can move the GEmCherry2 sequence back in-frame and allow quantification and comparison of the efficiency of different sgRNA target sites by measuring red fluorescence. With this GEmCherry2 assay, we compared four target sites in the safe harbor AAVS1 locus and found significant differences in target site activity. We verified the activity using TIDE, which ranked our target sites in a similar order as the GEmCherry2 system. We also identified an AAV short inverted terminal repeat sequence within the Cas9 construct that, upon removal significantly improved transient transfection and expression in hESCs. Moreover, using GEmCherry2, we designed a sgRNA to target SORCS2 in hESCs and successfully introduced indels into the coding sequence of SORCS2.
Collapse
Affiliation(s)
- Camilla Højland Knudsen
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Emilía S Ásgrímsdóttir
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Karim Rahimi
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark
| | - Katherine P Gill
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Søs Frandsen
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.,Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Susanne Hvolbøl Buchholdt
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Muwan Chen
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jørgen Kjems
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,Interdisciplinary Nanoscience Center, Aarhus University, Aarhus, Denmark
| | - Fabia Febbraro
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Mark Denham
- Danish Research Institute of Translational Neuroscience - DANDRITE, Nordic-EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark.,Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
40
|
Buttenschøn HN, Elfving B, Nielsen M, Skeldal S, Kaas M, Mors O, Glerup S. Exploring the sortilin related receptor, SorLA, in depression. J Affect Disord 2018; 232:260-267. [PMID: 29499509 DOI: 10.1016/j.jad.2018.02.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 01/29/2018] [Accepted: 02/16/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND Studies of individual biomarkers for depression have shown insufficient sensitivity and specificity for clinical use, and most likely combinations of biomarkers may provide a better signature. The sorting-related receptor with A-type repeats (SorLA) is a well-studied pathogenic factor for Alzheimer's. SorLA belongs to the Vps10p domain receptor family, which also encompasses sortilin and SorCS1-3. All family members have been implicated in neurological and mental disorders. Notably, the SORCS3 gene is genome-wide significantly associated with depression and serum protein levels of sortilin are reduced in depressed individuals. SorLA regulates the activity of neurotrophic factors and cytokines and we hence speculated that SorLA might be implicated in depression. METHODS Serum SorLA levels were measured in two well-defined clinical samples using ELISA. Generalized linear models were used in the statistical analyses. RESULTS We identified a multivariate model to discriminate depressed individuals from healthy controls. Interestingly, the model consisted of serum SorLA levels and additional four predictors: previous depressive episode, stressful life events, serum levels of sortilin and VEGF. However, as an isolated factor, we observed no significant difference in SorLA levels between 140 depressed individuals and 140 healthy controls. Nevertheless, we observed a significant increase in SorLA levels following 12 weeks of treatment with nortriptyline, but not escitalopram. LIMITATIONS The number of biomarkers included in the multivariate model for depression and lack of replication limit our study. CONCLUSIONS Our results suggest SorLA as one of five factors that in combination may support the depression diagnosis, but not as an individual biomarker for depression or treatment response.
Collapse
Affiliation(s)
- Henriette N Buttenschøn
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark; The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark.
| | - Betina Elfving
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
| | - Marit Nielsen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Denmark
| | - Sune Skeldal
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Mathias Kaas
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark
| | - Ole Mors
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark; Psychosis Research Unit, Aarhus University Hospital, Risskov, Denmark
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark; The Lundbeck Foundation Research Center, MIND, Aarhus University, Denmark
| |
Collapse
|
41
|
Parrish DC, Francis Stuart SD, Olivas A, Wang L, Nykjaer A, Ripplinger CM, Habecker BA. Transient denervation of viable myocardium after myocardial infarction does not alter arrhythmia susceptibility. Am J Physiol Heart Circ Physiol 2018; 314:H415-H423. [PMID: 29101167 PMCID: PMC5899257 DOI: 10.1152/ajpheart.00300.2017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 10/30/2017] [Accepted: 10/30/2017] [Indexed: 01/14/2023]
Abstract
Cardiac sympathetic nerves stimulate heart rate and force of contraction. Myocardial infarction (MI) leads to the loss of sympathetic nerves within the heart, and clinical studies have indicated that sympathetic denervation is a risk factor for arrhythmias and cardiac arrest. Two distinct types of denervation have been identified in the mouse heart after MI caused by ischemia-reperfusion: transient denervation of peri-infarct myocardium and sustained denervation of the infarct. Sustained denervation is linked to increased arrhythmia risk, but it is not known whether acute nerve loss in peri-infarct myocardium also contributes to arrhythmia risk. Peri-infarct sympathetic denervation requires the p75 neurotrophin receptor (p75NTR), but removal of p75NTR alters the pattern of sympathetic innervation in the heart and increases spontaneous arrhythmias. Therefore, we targeted the p75NTR coreceptor sortilin and the p75NTR-induced protease tumor necrosis factor-α-converting enzyme/A disintegrin and metalloproteinase domain 17 (TACE/ADAM17) to selectively block peri-infarct denervation. Sympathetic nerve density was quantified using immunohistochemistry for tyrosine hydroxylase. Genetic deletion of sortilin had no effect on the timing or extent of axon degeneration, but inhibition of TACE/ADAM17 with the protease inhibitor marimastat prevented the loss of axons from viable myocardium. We then asked whether retention of nerves in peri-infarct myocardium had an impact on cardiac electrophysiology 3 days after MI using ex vivo optical mapping of transmembrane potential and intracellular Ca2+. Preventing acute denervation of viable myocardium after MI did not significantly alter cardiac electrophysiology or Ca2+ handling, suggesting that transient denervation at this early time point has minimal impact on arrhythmia risk. NEW & NOTEWORTHY Sympathetic denervation after myocardial infarction is a risk factor for arrhythmias. We asked whether transient loss of nerves in viable myocardium contributed to arrhythmia risk. We found that targeting protease activity could prevent acute peri-infarct denervation but that it did not significantly alter cardiac electrophysiology or Ca2+ handling 3 days after myocardial infarction.
Collapse
MESH Headings
- ADAM17 Protein/metabolism
- Action Potentials
- Adaptor Proteins, Vesicular Transport/metabolism
- Animals
- Arrhythmias, Cardiac/etiology
- Arrhythmias, Cardiac/metabolism
- Arrhythmias, Cardiac/pathology
- Arrhythmias, Cardiac/physiopathology
- Calcium Signaling
- Disease Models, Animal
- Heart/innervation
- Heart Rate
- Isolated Heart Preparation
- Mice, Inbred C57BL
- Mice, Knockout
- Myocardial Infarction/complications
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardial Infarction/physiopathology
- Myocardium/metabolism
- Myocardium/pathology
- Receptors, Nerve Growth Factor/deficiency
- Receptors, Nerve Growth Factor/genetics
- Sympathetic Nervous System/metabolism
- Sympathetic Nervous System/physiopathology
- Time Factors
- Tissue Survival
Collapse
Affiliation(s)
- Diana C Parrish
- Department of Physiology and Pharmacology, Oregon Health and Science University , Portland, Oregon
| | | | - Antoinette Olivas
- Department of Physiology and Pharmacology, Oregon Health and Science University , Portland, Oregon
| | - Lianguo Wang
- Department of Pharmacology, University of California , Davis, California
| | - Anders Nykjaer
- Department of Biomedicine-Medical Biochemistry, Aarhus University , Aarhus , Denmark
| | | | - Beth A Habecker
- Department of Physiology and Pharmacology, Oregon Health and Science University , Portland, Oregon
- Department of Medicine and Knight Cardiovascular Institute, Oregon Health and Science University , Portland, Oregon
| |
Collapse
|
42
|
Boggild S, Molgaard S, Glerup S, Nyengaard JR. Highly segregated localization of the functionally related vps10p receptors sortilin and SorCS2 during neurodevelopment. J Comp Neurol 2018; 526:1267-1286. [PMID: 29405286 DOI: 10.1002/cne.24403] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 01/20/2018] [Accepted: 01/22/2018] [Indexed: 12/11/2022]
Abstract
Nervous system development is a precisely orchestrated series of events requiring a multitude of intrinsic and extrinsic cues. Sortilin and SorCS2 are members of the Vps10p receptor family with complementary influence on some of these cues including the neurotrophins (NTs). However, the developmental time points where sortilin and SorCS2 exert their activities in conjunction or independently still remain unclear. In this study we present the characterization of the spatiotemporal expression pattern of sortilin and SorCS2 in the developing murine nervous system. Sortilin is highly expressed in the fetal nervous system with expression localized to distinct cell populations. Expression was high in neurons of the cortical plate and developing allocortex, as well as subpallial structures. Furthermore, the neuroepithelium lining the ventricles and the choroid plexus showed high expression of sortilin, together with the developing retina, spinal ganglia, and sympathetic ganglia. In contrast, SorCS2 was confined in a marked degree to the thalamus and, at E13.5, the floor plate from midbrain rostrally to spinal cord caudally. SorCS2 was also found in the ventricular zones of the ventral hippocampus and nucleus accumbens areas, in the meninges and in Schwann cells. Hence, sortilin and SorCS2 are extensively present in several distinct anatomical areas in the developing nervous system and are rarely co-expressed. Possible functions of sortilin and SorCS2 pertain to NT signaling, axon guidance and beyond. The present data will form the basis for hypotheses and study designs for unravelling the functions of sortilin and SorCS2 during the establishment of neuronal structures and connections.
Collapse
Affiliation(s)
- Simon Boggild
- Department of Clinical Medicine, Aarhus University, MIND Centre, Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus C, 8000, Denmark.,MIND Centre, Department of Biomedicine, Aarhus University, Ole Worms Allé 3, Aarhus C, 8000, Denmark
| | - Simon Molgaard
- Department of Clinical Medicine, Aarhus University, MIND Centre, Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus C, 8000, Denmark.,MIND Centre, Department of Biomedicine, Aarhus University, Ole Worms Allé 3, Aarhus C, 8000, Denmark
| | - Simon Glerup
- MIND Centre, Department of Biomedicine, Aarhus University, Ole Worms Allé 3, Aarhus C, 8000, Denmark
| | - Jens Randel Nyengaard
- Department of Clinical Medicine, Aarhus University, MIND Centre, Core Center for Molecular Morphology, Section for Stereology and Microscopy, Aarhus C, 8000, Denmark.,Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus C, 8000, Denmark
| |
Collapse
|
43
|
Uemura T, Shiroshima T, Maeda A, Yasumura M, Shimada T, Fukata Y, Fukata M, Yoshida T. In situ screening for postsynaptic cell adhesion molecules during synapse formation. J Biochem 2017; 162:295-302. [PMID: 28449070 DOI: 10.1093/jb/mvx030] [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: 04/06/2017] [Accepted: 04/13/2017] [Indexed: 01/01/2023] Open
Abstract
Neuronal synapse formation is regulated by pre- and postsynaptic cell adhesion molecules. Presynaptic neurexins (NRXNs) and receptor protein tyrosine phosphatases (RPTPs; PTPδ, PTPσ and LAR in mammals) can induce postsynaptic differentiation through the interaction with various postsynaptic cell adhesion molecules. Here, we developed a novel in situ screening method to identify postsynaptic membranous proteins involved in synaptogenesis. Magnetic beads coated with the extracellular domains of NRXN1β(-S4) and PTPδ-A6 variants preferentially induced excitatory postsynaptic differentiation on the beads' surface when co-cultured with cortical neurons. After inducing postsynaptic sites on these beads, protein complexes including NRXN1β(-S4)/PTPδ-A6 and their ligands on the neuronal membrane were chemically cross-linked and purified using a magnetic separator. Liquid chromatography-tandem mass spectrometry analysis of the complexes revealed two types of postsynaptic ligands for NRXN1β(-S4) and PTPδ-A6, one has an activity to induce presynaptic differentiation in a trans manner, whereas the other has no such activity. These results suggest that synapse formation is regulated by the interplay between presynaptic NRXN/PTPδ and their postsynaptic ligands with functionally different impacts on pre- and postsynaptic differentiation. Thus, our in situ screening method for identifying synapse-organizing complexes will help to understand the molecular basis for elaborate neuronal networks.
Collapse
Affiliation(s)
- Takeshi Uemura
- Department of Molecular and Cellular Physiology, Institute of Medicine, Academic Assembly, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.,Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.,CREST, JST, 4-1-8 Hon-cho, Kawaguchi, Saitama 332-0012, Japan
| | - Tomoko Shiroshima
- Department of Molecular Neurobiology and Pharmacology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0073, Japan
| | - Asami Maeda
- Department of Molecular Neurobiology and Pharmacology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0073, Japan
| | - Misato Yasumura
- Department of Anatomy and Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takashi Shimada
- Technology Research Laboratory, SHIMADZU Corporation, 380-1 Horiyamashita, Hadano, Kanagawa 259-1304, Japan
| | - Yuko Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| | - Masaki Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan
| | - Tomoyuki Yoshida
- Department of Molecular Neuroscience, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama, Toyama 930-0194, Japan.,PRESTO, JST, 4-1-8 Hon-cho, Kawaguchi, Saitama 332-0012, Japan
| |
Collapse
|
44
|
Januliene D, Manavalan A, Ovesen PL, Pedersen KM, Thirup S, Nykjær A, Moeller A. Hidden Twins: SorCS Neuroreceptors Form Stable Dimers. J Mol Biol 2017; 429:2907-2917. [PMID: 28827148 DOI: 10.1016/j.jmb.2017.08.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 08/09/2017] [Accepted: 08/11/2017] [Indexed: 11/26/2022]
Abstract
SorCS1, SorCS2 and SorCS3 belong to the Vps10p-domain family of multiligand receptors. Genetic and functional studies have linked SorCS receptors to psychiatric disorders, Alzheimer's disease and type 2 diabetes, demonstrating critical roles in neuronal functionality and metabolic control. Surprisingly, their structural composition has so far not been studied. Here we have characterized SorCS1, SorCS2 and SorCS3 using biochemical methods and electron microscopy. We found that their purified extracellular domains co-exist in stable dimeric and monomeric populations. This was supported by co-immunoprecipitation experiments, where membrane-bound dimers were successfully pulled down from cell lysate. While dimers were virtually unbreakable, dimerization of the monomeric population was promoted through enzymatic deglycosylation. We conclude that post-translational modifications, specifically the degree and pattern of glycosylation, regulate the oligomeric state of the protein. Hence, cells may dictate ligand specificity by controlling the ratio between monomers and dimers and, therefore, regulate the multiple functions of SorCS receptors.
Collapse
Affiliation(s)
- Dovile Januliene
- Department of Structural Biology, Max Planck Institute of Biophysics, Max-von-Laue-Straße 3, 60438 Frankfurt am Main, Germany; DANDRITE, iNANO, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark
| | | | - Peter Lund Ovesen
- DANDRITE, Department of Biomedicine, Aarhus University, Ole Worms Allé 3, 8000 Aarhus C, Denmark
| | - Karen-Marie Pedersen
- DANDRITE, Department of Biomedicine, Aarhus University, Ole Worms Allé 3, 8000 Aarhus C, Denmark
| | - Søren Thirup
- MIND Centre, Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10C, 8000 Aarhus C, Denmark
| | - Anders Nykjær
- Mayo Clinic, 4500 San Pablo Rd S, Jacksonville, FL 32224, USA; DANDRITE, Department of Biomedicine, Aarhus University, Ole Worms Allé 3, 8000 Aarhus C, Denmark
| | - Arne Moeller
- Department of Structural Biology, Max Planck Institute of Biophysics, Max-von-Laue-Straße 3, 60438 Frankfurt am Main, Germany; DANDRITE, iNANO, Aarhus University, Gustav Wieds Vej 14, 8000 Aarhus C, Denmark.
| |
Collapse
|
45
|
Notaras MJ, Hill RA, Gogos JA, van den Buuse M. BDNF Val66Met Genotype Interacts With a History of Simulated Stress Exposure to Regulate Sensorimotor Gating and Startle Reactivity. Schizophr Bull 2017; 43:665-672. [PMID: 27262112 PMCID: PMC5464110 DOI: 10.1093/schbul/sbw077] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Reduced expression of Brain-Derived Neurotrophic Factor (BDNF) has been implicated in the pathophysiology of schizophrenia. The BDNF Val66Met polymorphism, which results in deficient activity-dependent secretion of BDNF, is associated with clinical features of schizophrenia. We investigated the effect of this polymorphism on Prepulse Inhibition (PPI), a translational model of sensorimotor gating which is disrupted in schizophrenia. We utilized humanized BDNFVal66Met (hBDNFVal66Met) mice which have been modified to carry the Val66Met polymorphism, as well as express humanized BDNF in vivo. We also studied the long-term effect of chronic corticosterone (CORT) exposure in these animals as a model of history of stress. PPI was assessed at 30ms and 100ms interstimulus intervals (ISI). Analysis of PPI at the commonly used 100ms ISI identified that, irrespective of CORT treatment, the hBDNFVal/Met genotype was associated with significantly reduced PPI. In contrast, PPI was not different between hBDNFMet/Met and hBDNFVal/Val genotype mice. At the 30ms ISI, CORT treatment selectively disrupted sensorimotor gating of hBDNFVal/Met heterozygote mice but not hBDNFVal/Val or hBDNFMet/Met mice. Analysis of startle reactivity revealed that chronic CORT reduced startle reactivity of hBDNFVal/Val male mice by 51%. However, this was independent of the effect of CORT on PPI. In summary, we provide evidence of a distinct BDNFVal66Met heterozygote-specific phenotype using the sensorimotor gating endophenotype of schizophrenia. These data have important implications for clinical studies where, if possible, the BDNFVal/Met heterozygote genotype should be distinguished from the BDNFMet/Met genotype.
Collapse
Affiliation(s)
- Michael J. Notaras
- Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, Melbourne, Australia;,Psychoneuroendocrinology Laboratory, Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Rachel A. Hill
- Psychoneuroendocrinology Laboratory, Florey Institute of Neuroscience and Mental Health, Melbourne, Australia
| | - Joseph A. Gogos
- Departments of Biophysics and Neuroscience, Columbia University, New York, NY
| | - Maarten van den Buuse
- Behavioural Neuroscience Laboratory, Florey Institute of Neuroscience and Mental Health, Melbourne, Australia;,School of Psychology and Public Health, La Trobe University, Melbourne, Australia;,The College of Public Health, Medical and Veterinary Sciences, James Cook University, Queensland, Australia
| |
Collapse
|
46
|
PROneurotrophins and CONSequences. Mol Neurobiol 2017; 55:2934-2951. [DOI: 10.1007/s12035-017-0505-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 03/21/2017] [Indexed: 01/12/2023]
|
47
|
Tsai SJ. Role of neurotrophic factors in attention deficit hyperactivity disorder. Cytokine Growth Factor Rev 2017; 34:35-41. [DOI: 10.1016/j.cytogfr.2016.11.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 11/15/2016] [Accepted: 11/25/2016] [Indexed: 12/20/2022]
|
48
|
Forge A, Taylor RR, Dawson SJ, Lovett M, Jagger DJ. Disruption of SorCS2 reveals differences in the regulation of stereociliary bundle formation between hair cell types in the inner ear. PLoS Genet 2017; 13:e1006692. [PMID: 28346477 PMCID: PMC5386298 DOI: 10.1371/journal.pgen.1006692] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 04/10/2017] [Accepted: 03/14/2017] [Indexed: 12/24/2022] Open
Abstract
Behavioural anomalies suggesting an inner ear disorder were observed in a colony of transgenic mice. Affected animals were profoundly deaf. Severe hair bundle defects were identified in all outer and inner hair cells (OHC, IHC) in the cochlea and in hair cells of vestibular macular organs, but hair cells in cristae were essentially unaffected. Evidence suggested the disorder was likely due to gene disruption by a randomly inserted transgene construct. Whole-genome sequencing identified interruption of the SorCS2 (Sortilin-related VPS-10 domain containing protein) locus. Real-time-qPCR demonstrated disrupted expression of SorCS2 RNA in cochlear tissue from affected mice and this was confirmed by SorCS2 immuno-labelling. In all affected hair cells, stereocilia were shorter than normal, but abnormalities of bundle morphology and organisation differed between hair cell types. Bundles on OHC were grossly misshapen with significantly fewer stereocilia than normal. However, stereocilia were organised in rows of increasing height. Bundles on IHC contained significantly more stereocilia than normal with some longer stereocilia towards the centre, or with minimal height differentials. In early postnatal mice, kinocilia (primary cilia) of IHC and of OHC were initially located towards the lateral edge of the hair cell surface but often became surrounded by stereocilia as bundle shape and apical surface contour changed. In macular organs the kinocilium was positioned in the centre of the cell surface throughout maturation. There was disruption of the signalling pathway controlling intrinsic hair cell apical asymmetry. LGN and Gαi3 were largely absent, and atypical Protein Kinase C (aPKC) lost its asymmetric distribution. The results suggest that SorCS2 plays a role upstream of the intrinsic polarity pathway and that there are differences between hair cell types in the deployment of the machinery that generates a precisely organised hair bundle.
Collapse
MESH Headings
- Age Factors
- Animals
- Gene Expression Regulation
- Hair Cells, Auditory, Inner/metabolism
- Hair Cells, Auditory, Inner/pathology
- Hearing Loss/genetics
- Hearing Loss/metabolism
- Hearing Loss/physiopathology
- Immunohistochemistry
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Microscopy, Confocal
- Microscopy, Electron, Scanning
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Organ of Corti/metabolism
- Organ of Corti/physiopathology
- Organ of Corti/ultrastructure
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Stereocilia/genetics
- Stereocilia/metabolism
- Stereocilia/pathology
Collapse
Affiliation(s)
- Andrew Forge
- UCL Ear Institute, University College London, London, United Kingdom
- * E-mail:
| | - Ruth R. Taylor
- UCL Ear Institute, University College London, London, United Kingdom
| | - Sally J. Dawson
- UCL Ear Institute, University College London, London, United Kingdom
| | - Michael Lovett
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Daniel J. Jagger
- UCL Ear Institute, University College London, London, United Kingdom
| |
Collapse
|
49
|
Zanin JP, Unsain N, Anastasia A. Growth factors and hormones pro-peptides: the unexpected adventures of the BDNF prodomain. J Neurochem 2017; 141:330-340. [PMID: 28218971 DOI: 10.1111/jnc.13993] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 02/08/2017] [Accepted: 02/13/2017] [Indexed: 12/26/2022]
Abstract
Most growth factors and hormones are synthesized as pre-pro-proteins which are processed to the biologically active mature protein. The pre- and prodomains are cleaved from the precursor protein in the secretory pathway or, in some cases, extracellularly. The canonical functions of these prodomains are to assist in folding and stabilization of the mature domain, to direct intra and extracellular localization, to facilitate storage, and to regulate bioavailability of their mature counterpart. Recently, exciting evidence has revealed that prodomains of certain growth factors, after cleaved from the precursor pro-protein, can act as independent active signaling molecules. In this review, we discuss the various classical functions of prodomains, and the biological consequences of these pro-peptides acting as ligands. We will focus our attention on the brain-derived neurotrophic factor prodomain (pBDNF), which has been recently described as a novel secreted ligand influencing neuronal morphology and physiology.
Collapse
Affiliation(s)
- Juan Pablo Zanin
- Department of Biological Sciences, Rutgers University, Newark, New Jersey, USA
| | - Nicolás Unsain
- Instituto de Investigación Médica Mercedes y Martin Ferreyra, (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
| | - Agustin Anastasia
- Instituto de Investigación Médica Mercedes y Martin Ferreyra, (INIMEC-CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
| |
Collapse
|
50
|
Differences in the Biological Functions of BDNF and proBDNF in the Central Nervous System. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s11055-017-0391-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|