1
|
Valle-Bautista R, Olivera-Acevedo M, Horta-Brussolo VR, Díaz NF, Ávila-González D, Molina-Hernández A. From songbird to humans: The multifaceted roles of FOXP2 in speech and motor learning. Neurosci Biobehav Rev 2024; 167:105936. [PMID: 39510218 DOI: 10.1016/j.neubiorev.2024.105936] [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: 06/10/2024] [Revised: 09/24/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024]
Abstract
Motor learning involves a complex network of brain structures and is crucial for tasks like speech. The cerebral cortex, subcortical nuclei, and cerebellum are involved in motor learning and vocalization. Vocal learning has been demonstrated across species. However, it is a task that should be further studied and reevaluated, particularly in species considered non-vocal learners, to potentially uncover new insights. FOXP2, a transcription factor, has been implicated in speech learning and execution. Several variants have been involved in speech and cognitive impairments; the most studied is the R553H, found in the KE family, where more than half of the members show verbal dyspraxia. Brain FOXP2 expression shows consistent patterns across species in regions associated with motor learning and execution. Animal models expressing mutated FOXP2 showed impaired motor learning and vocalization. Genes regulated by FOXP2 are related to neural differentiation, connectivity, and synaptic plasticity, indicating its role in brain development and function. This review explores the intricate relationship between FOXP2, motor learning, and speech in an anatomical and functional context.
Collapse
Affiliation(s)
- Rocío Valle-Bautista
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales 800, Miguel Hidalgo, Ciudad de México 11000, Mexico
| | - Monserrath Olivera-Acevedo
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales 800, Miguel Hidalgo, Ciudad de México 11000, Mexico; Instituto Tecnológico de Monterrey Campus Ciudad de México, Escuela de Medicina y Ciencias de la Salud, Colombia
| | - Victoria Regina Horta-Brussolo
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales 800, Miguel Hidalgo, Ciudad de México 11000, Mexico; Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, CINVESTAV-IPN, Ciudad de México, México
| | - Néstor Fabián Díaz
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales 800, Miguel Hidalgo, Ciudad de México 11000, Mexico
| | - Daniela Ávila-González
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales 800, Miguel Hidalgo, Ciudad de México 11000, Mexico
| | - Anayansi Molina-Hernández
- Departamento de Fisiología y Desarrollo Celular, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Montes Urales 800, Miguel Hidalgo, Ciudad de México 11000, Mexico.
| |
Collapse
|
2
|
Gergues MM, Lalani LK, Kheirbek MA. Identifying dysfunctional cell types and circuits in animal models for psychiatric disorders with calcium imaging. Neuropsychopharmacology 2024; 50:274-284. [PMID: 39122815 PMCID: PMC11525937 DOI: 10.1038/s41386-024-01942-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/30/2024] [Accepted: 07/09/2024] [Indexed: 08/12/2024]
Abstract
A central goal of neuroscience is to understand how the brain transforms external stimuli and internal bodily signals into patterns of activity that underlie cognition, emotional states, and behavior. Understanding how these patterns of activity may be disrupted in mental illness is crucial for developing novel therapeutics. It is well appreciated that psychiatric disorders are complex, circuit-based disorders that arise from dysfunctional activity patterns generated in discrete cell types and their connections. Recent advances in large-scale, cell-type specific calcium imaging approaches have shed new light on the cellular, circuit, and network-level dysfunction in animal models for psychiatric disorders. Here, we highlight a series of recent findings over the last ~10 years from in vivo calcium imaging studies that show how aberrant patterns of activity in discrete cell types and circuits may underlie behavioral deficits in animal models for several psychiatric disorders, including depression, anxiety, autism spectrum disorders, and schizophrenia. These advances in calcium imaging in pre-clinical models demonstrate the power of cell-type-specific imaging tools in understanding the underlying dysfunction in cell types, activity patterns, and neural circuits that may contribute to disease and provide new blueprints for developing more targeted therapeutics and treatment strategies.
Collapse
Affiliation(s)
- Mark M Gergues
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Lahin K Lalani
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Mazen A Kheirbek
- Neuroscience Graduate Program, University of California San Francisco, San Francisco, CA, USA.
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, USA.
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA, USA.
- Center for Integrative Neuroscience, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
3
|
Zhu YQ, Zhou XQ, Yang M, Horill S, Wang ZY, Yang JJ, Pan YB, Zhou XK. Case report: Recurring potassium channel complex autoimmunity-related neuropathic pain. Front Immunol 2024; 15:1390171. [PMID: 39524448 PMCID: PMC11543456 DOI: 10.3389/fimmu.2024.1390171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Voltage-gated potassium channel (VGKC) complex autoimmunity associated with nerve hyperexcitability is an uncommon clinical spectrum. It is mostly characterized by limbic encephalitis, continuous neuromyotonia, and dysautonomia. Pain, however, has rarely been reported as the first symptom. The present study describes a case of persistent neuropathic pain as the only symptom associated with a positive serum contactin-associated protein-like 2 (CASPR2) auto-antibody in a 41-year-old female patient. Her pain was completely relieved with steroids and intravenous immunoglobulin (IVIG) therapy. Nevertheless, the pain recurred 1 year later, consistent with an immunofluorescence titer of the CASPR2 antibody. Our case shows that neuropathic pain may occur as the first and only manifestation of a VGKC complex autoimmunity disorder. VGKC antibody titers might be an indication of pain severity. Steroids coupled with IVIG are effective, but relapse may still occur.
Collapse
Affiliation(s)
- Yun-Qing Zhu
- The First Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Xin-Qiao Zhou
- Department of Anesthesia and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Min Yang
- Department of Anesthesiology and Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Smita Horill
- The First Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Zhong-Yun Wang
- Department of Anesthesia and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yin-Bing Pan
- Department of Anesthesia and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| | - Xiao-Kai Zhou
- Department of Anesthesia and Perioperative Medicine, The First Affiliated Hospital with Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Koster AK, Yarishkin O, Dubin AE, Kefauver JM, Pak RA, Cravatt BF, Patapoutian A. Chemical mapping of the surface interactome of PIEZO1 identifies CADM1 as a modulator of channel inactivation. Proc Natl Acad Sci U S A 2024; 121:e2415934121. [PMID: 39356664 PMCID: PMC11474052 DOI: 10.1073/pnas.2415934121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
The propeller-shaped blades of the PIEZO1 and PIEZO2 ion channels partition into the plasma membrane and respond to indentation or stretching of the lipid bilayer, thus converting mechanical forces into signals that can be interpreted by cells, in the form of calcium flux and changes in membrane potential. While PIEZO channels participate in diverse physiological processes, from sensing the shear stress of blood flow in the vasculature to detecting touch through mechanoreceptors in the skin, the molecular details that enable these mechanosensors to tune their responses over a vast dynamic range of forces remain largely uncharacterized. To survey the molecular landscape surrounding PIEZO channels at the cell surface, we employed a mass spectrometry-based proteomic approach to capture and identify extracellularly exposed proteins in the vicinity of PIEZO1. This PIEZO1-proximal interactome was enriched in surface proteins localized to cell junctions and signaling hubs within the plasma membrane. Functional screening of these interaction candidates by calcium imaging and electrophysiology in an overexpression system identified the adhesion molecule CADM1/SynCAM that slows the inactivation kinetics of PIEZO1 with little effect on PIEZO2. Conversely, we found that CADM1 knockdown accelerates inactivation of endogenous PIEZO1 in Neuro-2a cells. Systematic deletion of CADM1 domains indicates that the transmembrane region is critical for the observed effects on PIEZO1, suggesting that modulation of inactivation is mediated by interactions in or near the lipid bilayer.
Collapse
Affiliation(s)
- Anna K. Koster
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
- Department of Chemistry, Scripps Research, La Jolla, CA92037
| | - Oleg Yarishkin
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | - Adrienne E. Dubin
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | - Jennifer M. Kefauver
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | - Ryan A. Pak
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| | | | - Ardem Patapoutian
- HHMI, Scripps Research, La JollaCA92037
- Department of Neuroscience, Scripps Research, La Jolla, CA92037
| |
Collapse
|
5
|
Bastien BL, Haury WR, Smisko WR, Hart MP. nlr-1/CNTNAP regulates dopamine circuit structure and foraging behaviors in C. elegans. Commun Biol 2024; 7:1248. [PMID: 39358459 PMCID: PMC11447218 DOI: 10.1038/s42003-024-06936-6] [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: 01/10/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
The neurexin superfamily, consisting of neurexins and Casprs, play important roles in the development, maintenance, function, and plasticity of neuronal circuits. Caspr/CNTNAP genes are linked to alterations in neuronal circuits and associated with neurodevelopmental and neurodegenerative conditions. Casprs are implicated in multiple neuronal signaling pathways, including dopamine; however, the molecular mechanisms by which Casprs differentially alter specific signaling pathways and downstream behaviors are unclear. We find that the C. elegans Caspr nlr-1 functions in neurons to control foraging behaviors, acting in distinct monoamine neurons to modulate locomotor activity in the presence or absence of food. nlr-1 functions in dopamine neurons to reduce activity in the absence of food, similar to the role of dopamine, and regulates dopamine signaling through D2-like receptors. Furthermore, nlr-1 contributes to proper morphology and presynaptic structure of dopamine neurons, dopamine receptor expression and localization, and the behavioral response to dopamine. We find that nlr-1 similarly regulates another dopamine-dependent behavior, the basal slowing response. Therefore, spatial manipulation of a broadly expressed neuronal gene is sufficient to alter neural circuits and behavior and uncovers important functions masked by global manipulation, highlighting the importance of genetic variation and mechanisms that impact spatial expression of genes to behavior.
Collapse
Affiliation(s)
- Brandon L Bastien
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - William R Haury
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - William R Smisko
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Michael P Hart
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
6
|
Quinot V, Rostasy K, Höftberger R. Antibody-Mediated Nodo- and Paranodopathies. J Clin Med 2024; 13:5721. [PMID: 39407781 PMCID: PMC11477122 DOI: 10.3390/jcm13195721] [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: 08/07/2024] [Revised: 09/12/2024] [Accepted: 09/22/2024] [Indexed: 10/20/2024] Open
Abstract
The recent discovery of pathogenic antibodies targeting cell adhesion molecules of the node of Ranvier has prompted efforts to develop a new classification for a subset of antibody-mediated peripheral neuropathies. These autoimmune nodo- and paranodopathies encompass epitopes such as neurofascin 155, neurofascin 186, contactin-1, and contactin-associated protein 1, with a high likelihood of involving additional yet unidentified proteins. So far, the investigation of this subset of patients was primarily focused on adults, with only rare reports of pediatric cases. Low awareness among pediatricians and insufficient availability of appropriate diagnostic methods in many laboratories may mask a higher pediatric incidence than currently observed. Diagnosis is made by transfected cell-based assays and ELISA to characterize the specific target antigen and antibody subclass that provides insight into the pathophysiology. Clinical features often resemble those of CIDP or GBS in adults, whilst in pediatric patients, although rare, an atypical CIDP phenotype has predominantly been reported. Yet, in contrast to classical immune-mediated neuropathies, the clinical course is usually rapidly progressive, and response to classical first-line therapy often poor. Although electrophysiological signs of demyelination are observed, segmental demyelination and inflammation are not present on pathological examination. Rather, few neuropathological reports demonstrate features of axonal neuropathy without signs of true de- or remyelination. This review aims to summarize recent findings on such nodo- and paranodoneuropathies, shining light on features of these disorders in pediatric patients, a still little-explored field with only a few reports currently present.
Collapse
Affiliation(s)
- Valérie Quinot
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Kevin Rostasy
- Department of Pediatric Neurology, Children’s Hospital Datteln, University Witten/Herdecke, 45711 Datteln, Germany;
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria;
| |
Collapse
|
7
|
Nawrocka WI, Cheng S, Hao B, Rosen MC, Cortés E, Baltrusaitis EE, Aziz Z, Kovács IA, Özkan E. Nematode Extracellular Protein Interactome Expands Connections between Signaling Pathways. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602367. [PMID: 39026773 PMCID: PMC11257444 DOI: 10.1101/2024.07.08.602367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Multicellularity was accompanied by the emergence of new classes of cell surface and secreted proteins. The nematode C. elegans is a favorable model to study cell surface interactomes, given its well-defined and stereotyped cell types and intercellular contacts. Here we report our C. elegans extracellular interactome dataset, the largest yet for an invertebrate. Most of these interactions were unknown, despite recent datasets for flies and humans, as our collection contains a larger selection of protein families. We uncover new interactions for all four major axon guidance pathways, including ectodomain interactions between three of the pathways. We demonstrate that a protein family known to maintain axon locations are secreted receptors for insulins. We reveal novel interactions of cystine-knot proteins with putative signaling receptors, which may extend the study of neurotrophins and growth-factor-mediated functions to nematodes. Finally, our dataset provides insights into human disease mechanisms and how extracellular interactions may help establish connectomes.
Collapse
Affiliation(s)
- Wioletta I. Nawrocka
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
- Institute for Neuroscience, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Shouqiang Cheng
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
- Institute for Neuroscience, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Bingjie Hao
- Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208, USA
| | - Matthew C. Rosen
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
- Institute for Neuroscience, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Elena Cortés
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
- Institute for Neuroscience, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Elana E. Baltrusaitis
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
- Institute for Neuroscience, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - Zainab Aziz
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
- Institute for Neuroscience, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| | - István A. Kovács
- Department of Physics and Astronomy, Northwestern University, Evanston, IL 60208, USA
- Northwestern Institute on Complex Systems, Northwestern University, Evanston, IL 60208, USA
- Department of Engineering Sciences and Applied Mathematics, Northwestern University, Evanston, IL 60208, USA
| | - Engin Özkan
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA
- Institute for Neuroscience, The University of Chicago, Chicago, IL 60637, USA
- Institute for Biophysical Dynamics, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
8
|
Cording KR, Tu EM, Wang H, Agopyan-Miu AHCW, Bateup HS. Cntnap2 loss drives striatal neuron hyperexcitability and behavioral inflexibility. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593387. [PMID: 38766169 PMCID: PMC11100810 DOI: 10.1101/2024.05.09.593387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by two major diagnostic criteria - persistent deficits in social communication and interaction, and the presence of restricted, repetitive patterns of behavior (RRBs). Evidence from both human and animal model studies of ASD suggest that alteration of striatal circuits, which mediate motor learning, action selection, and habit formation, may contribute to the manifestation of RRBs. CNTNAP2 is a syndromic ASD risk gene, and loss of function of Cntnap2 in mice is associated with RRBs. How loss of Cntnap2 impacts striatal neuron function is largely unknown. In this study, we utilized Cntnap2-/- mice to test whether altered striatal neuron activity contributes to aberrant motor behaviors relevant to ASD. We find that Cntnap2-/- mice exhibit increased cortical drive of striatal projection neurons (SPNs), with the most pronounced effects in direct pathway SPNs. This enhanced drive is likely due to increased intrinsic excitability of SPNs, which make them more responsive to cortical inputs. We also find that Cntnap2-/- mice exhibit spontaneous repetitive behaviors, increased motor routine learning, and cognitive inflexibility. Increased corticostriatal drive, in particular of the direct pathway, may contribute to the acquisition of repetitive, inflexible behaviors in Cntnap2 mice.
Collapse
Affiliation(s)
- Katherine R. Cording
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA USA
| | - Emilie M. Tu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | - Hongli Wang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| | | | - Helen S. Bateup
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA USA
| |
Collapse
|
9
|
Greguletz P, Plötz M, Baade-Büttner C, Bien CG, Eisenhut K, Geis C, Handreka R, Klausewitz J, Körtvelyessy P, Kovac S, Kraft A, Lewerenz J, Malter M, Nagel M, von Podewils F, Prüß H, Rada A, Rau J, Rauer S, Rößling R, Seifert-Held T, Siebenbrodt K, Sühs KW, Tauber SC, Thaler F, Wagner J, Wickel J, Leypoldt F, Rittner HL, Sommer C, Villmann C, Doppler K. Different pain phenotypes are associated with anti-Caspr2 autoantibodies. J Neurol 2024; 271:2736-2744. [PMID: 38386048 PMCID: PMC11055745 DOI: 10.1007/s00415-024-12224-4] [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: 12/12/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/23/2024]
Abstract
Autoantibodies against contactin-associated protein 2 (Caspr2) not only induce limbic autoimmune encephalitis but are also associated with pain conditions. Here, we analyzed clinical data on pain in a large cohort of patients included into the German Network for Research in Autoimmune Encephalitis. Out of 102 patients in our cohort, pain was a frequent symptom (36% of all patients), often severe (63.6% of the patients with pain) and/or even the major symptom (55.6% of the patients with pain). Pain phenotypes differed between patients. Cluster analysis revealed two major phenotypes including mostly distal-symmetric burning pain and widespread pain with myalgia and cramps. Almost all patients had IgG4 autoantibodies and some additional IgG1, 2, and/or 3 autoantibodies, but IgG subclasses, titers, and presence or absence of intrathecal synthesis were not associated with the occurrence of pain. However, certain pre-existing risk factors for chronic pain like diabetes mellitus, peripheral neuropathy, or preexisting chronic back pain tended to occur more frequently in patients with anti-Caspr2 autoantibodies and pain. Our data show that pain is a relevant symptom in patients with anti-Caspr2 autoantibodies and support the idea of decreased algesic thresholds leading to pain. Testing for anti-Caspr2 autoantibodies needs to be considered in patients with various pain phenotypes.
Collapse
Affiliation(s)
- Patrik Greguletz
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Str. 11, 97080, Würzburg, Germany
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Maria Plötz
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Str. 11, 97080, Würzburg, Germany
| | - Carolin Baade-Büttner
- Section Translational Neuroimmunology, Department for Neurology, Jena University Hospital, Jena, Germany
| | - Christian G Bien
- Department of Epileptology (Krankenhaus Mara), Medical School, Bielefeld University, Campus Bielefeld-Bethel, Bielefeld, Germany
- Laboratory Krone, Bad Salzuflen, Germany
| | - Katharina Eisenhut
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
- Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christian Geis
- Section Translational Neuroimmunology, Department for Neurology, Jena University Hospital, Jena, Germany
| | | | - Jaqueline Klausewitz
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Peter Körtvelyessy
- Department of Neurology, University Hospital Magdeburg, Magdeburg, Germany
- Department of Neurology and Experimental Neurology, Charité Berlin, and German Center for Neurodegenerative Diseases (DZNE),, Berlin, Germany
| | - Stjepana Kovac
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Andrea Kraft
- Department of Neurology, Martha-Maria Hospital Halle, Halle, Germany
| | - Jan Lewerenz
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Michael Malter
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Michael Nagel
- Department of Neurology, Klinikum Osnabrück, Osnabrück, Germany
| | - Felix von Podewils
- Department of Neurology, University Hospital Greifswald, Greifswald, Germany
| | - Harald Prüß
- Department of Neurology and Experimental Neurology, Charité Berlin, and German Center for Neurodegenerative Diseases (DZNE),, Berlin, Germany
| | - Anna Rada
- Department of Epileptology (Krankenhaus Mara), Medical School, Bielefeld University, Campus Bielefeld-Bethel, Bielefeld, Germany
| | - Johanna Rau
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Sebastian Rauer
- Department of Neurology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Rosa Rößling
- Department of Neurology and Experimental Neurology, Charité Berlin, and German Center for Neurodegenerative Diseases (DZNE),, Berlin, Germany
| | - Thomas Seifert-Held
- Department of Neurology, Medical University of Graz, Graz, Austria
- Department of Neurology, Hospital Murtal, Knittelfeld, Austria
| | - Kai Siebenbrodt
- Department of Neurology, University Hospital Frankfurt, Frankfurt, Germany
| | | | - Simone C Tauber
- Department of Neurology, RWTH University Hospital Aachen, Aachen, Germany
| | - Franziska Thaler
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität Munich, Munich, Germany
- Biomedical Center (BMC), Medical Faculty, Ludwig-Maximilians-Universität Munich, Martinsried, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Judith Wagner
- Department of Neurology, Kepler University Hospital Linz, Linz, Austria
- Department of Neurology, Evangelisches Klinikum Gelsenkirchen, Academic Hospital University Essen-Duisburg, Gelsenkirchen, Germany
| | - Jonathan Wickel
- Section Translational Neuroimmunology, Department for Neurology, Jena University Hospital, Jena, Germany
| | - Frank Leypoldt
- Department of Neurology, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Heike L Rittner
- Department of Anesthesiology, Intensive Care, Emergency Medicine and Pain Medicine, Centre for Interdisciplinary Pain Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Claudia Sommer
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Str. 11, 97080, Würzburg, Germany
| | - Carmen Villmann
- Institute of Clinical Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Kathrin Doppler
- Department of Neurology, University Hospital Würzburg, Josef-Schneider-Str. 11, 97080, Würzburg, Germany.
| |
Collapse
|
10
|
Zhang Q, Xing M, Bao Z, Xu L, Bai Y, Chen W, Pan W, Cai F, Wang Q, Guo S, Zhang J, Wang Z, Wu Y, Zhang Y, Li JD, Song W. Contactin-associated protein-like 2 (CNTNAP2) mutations impair the essential α-secretase cleavages, leading to autism-like phenotypes. Signal Transduct Target Ther 2024; 9:51. [PMID: 38424048 PMCID: PMC10904759 DOI: 10.1038/s41392-024-01768-6] [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: 10/03/2023] [Revised: 01/22/2024] [Accepted: 02/07/2024] [Indexed: 03/02/2024] Open
Abstract
Mutations in the Contactin-associated protein-like 2 (CNTNAP2) gene are associated with autism spectrum disorder (ASD), and ectodomain shedding of the CNTNAP2 protein plays a role in its function. However, key enzymes involved in the C-terminal cleavage of CNTNAP2 remain largely unknown, and the effect of ASD-associated mutations on this process and its role in ASD pathogenesis remain elusive. In this report we showed that CNTNAP2 undergoes sequential cleavages by furin, ADAM10/17-dependent α-secretase and presenilin-dependent γ-secretase. We identified that the cleavage sites of ADAM10 and ADAM17 in CNTNAP2 locate at its C-terminal residue I79 and L96, and the main α-cleavage product C79 by ADAM10 is required for the subsequent γ-secretase cleavage to generate CNTNAP2 intracellular domain (CICD). ASD-associated CNTNAP2 mutations impair the α-cleavage to generate C79, and the inhibition leads to ASD-like repetitive and social behavior abnormalities in the Cntnap2-I1254T knock-in mice. Finally, exogenous expression of C79 improves autism-like phenotypes in the Cntnap2-I1254T knock-in and Cntnap2-/- knockout mice. This data demonstrates that the α-secretase is essential for CNTNAP2 processing and its function. Our study indicates that inhibition of the cleavage by pathogenic mutations underlies ASD pathogenesis, and upregulation of its C-terminal fragments could have therapeutical potentials for ASD treatment.
Collapse
Affiliation(s)
- Qing Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Wenzhou Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Mengen Xing
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Wenzhou Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Zhengkai Bao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Wenzhou Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Lu Xu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Wenzhou Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yang Bai
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Wenzhou Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wanqi Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Wenzhou Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Wenhao Pan
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Wenzhou Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Fang Cai
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Wenzhou Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Qunxian Wang
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Shipeng Guo
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Jing Zhang
- Center for Medical Genetics, Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Zhe Wang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yili Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Wenzhou Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yun Zhang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Jia-Da Li
- Center for Medical Genetics, Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China.
| | - Weihong Song
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Zhejiang Key Laboratory of Alzheimer's Disease, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Wenzhou Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- Chongqing Key Laboratory of Translational Medical Research in Cognitive Development and Learning and Memory Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China.
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
11
|
Guise AJ, Misal SA, Carson R, Chu JH, Boekweg H, Van Der Watt D, Welsh NC, Truong T, Liang Y, Xu S, Benedetto G, Gagnon J, Payne SH, Plowey ED, Kelly RT. TDP-43-stratified single-cell proteomics of postmortem human spinal motor neurons reveals protein dynamics in amyotrophic lateral sclerosis. Cell Rep 2024; 43:113636. [PMID: 38183652 PMCID: PMC10926001 DOI: 10.1016/j.celrep.2023.113636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 11/02/2023] [Accepted: 12/14/2023] [Indexed: 01/08/2024] Open
Abstract
A limitation of conventional bulk-tissue proteome studies in amyotrophic lateral sclerosis (ALS) is the confounding of motor neuron (MN) signals by admixed non-MN proteins. Here, we leverage laser capture microdissection and nanoPOTS single-cell mass spectrometry-based proteomics to query changes in protein expression in single MNs from postmortem ALS and control tissues. In a follow-up analysis, we examine the impact of stratification of MNs based on cytoplasmic transactive response DNA-binding protein 43 (TDP-43)+ inclusion pathology on the profiles of 2,238 proteins. We report extensive overlap in differentially abundant proteins identified in ALS MNs with or without overt TDP-43 pathology, suggesting early and sustained dysregulation of cellular respiration, mRNA splicing, translation, and vesicular transport in ALS. Together, these data provide insights into proteome-level changes associated with TDP-43 proteinopathy and begin to demonstrate the utility of pathology-stratified trace sample proteomics for understanding single-cell protein dynamics in human neurologic diseases.
Collapse
Affiliation(s)
| | - Santosh A Misal
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Richard Carson
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | | | - Hannah Boekweg
- Biology Department, Brigham Young University, Provo, UT 84602, USA
| | | | | | - Thy Truong
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Yiran Liang
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | | | | | | | - Samuel H Payne
- Biology Department, Brigham Young University, Provo, UT 84602, USA
| | | | - Ryan T Kelly
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
12
|
Griffin EN, Jucius T, Sim SE, Harris BS, Heinz S, Ackerman SL. RREB1 regulates neuronal proteostasis and the microtubule network. SCIENCE ADVANCES 2024; 10:eadh3929. [PMID: 38198538 PMCID: PMC10780896 DOI: 10.1126/sciadv.adh3929] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024]
Abstract
Transcription factors play vital roles in neuron development; however, little is known about the role of these proteins in maintaining neuronal homeostasis. Here, we show that the transcription factor RREB1 (Ras-responsive element-binding protein 1) is essential for neuron survival in the mammalian brain. A spontaneous mouse mutation causing loss of a nervous system-enriched Rreb1 transcript is associated with progressive loss of cerebellar Purkinje cells and ataxia. Analysis of chromatin immunoprecipitation and sequencing, along with RNA sequencing data revealed dysregulation of RREB1 targets associated with the microtubule cytoskeleton. In agreement with the known role of microtubules in dendritic development, dendritic complexity was disrupted in Rreb1-deficient neurons. Analysis of sequencing data also suggested that RREB1 plays a role in the endomembrane system. Mutant Purkinje cells had fewer numbers of autophagosomes and lysosomes and contained P62- and ubiquitin-positive inclusions. Together, these studies demonstrate that RREB1 functions to maintain the microtubule network and proteostasis in mammalian neurons.
Collapse
Affiliation(s)
- Emily N. Griffin
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Thomas Jucius
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Su-Eon Sim
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - Sven Heinz
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Susan L. Ackerman
- Howard Hughes Medical Institute, Department of Cellular and Molecular Medicine, Department of Neurobiology, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
13
|
Quinot V, Höftberger R. Pathogenesis and immunopathology of paraneoplastic disorders. HANDBOOK OF CLINICAL NEUROLOGY 2024; 200:33-54. [PMID: 38494287 DOI: 10.1016/b978-0-12-823912-4.00027-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Paraneoplastic neurologic syndromes (PNS) represent a rare group of immune-mediated complications associated with an underlying tumor. Ectopic protein expression in neoplastic cells or an aberrant immune regulation in the course of hematooncologic diseases or thymomas trigger an autoimmune response that may affect any part of the central and/or peripheral nervous system. Recent advances in drug therapies as well as novel animal models and neuropathologic studies have led to further insights on the immune pathomechanisms of PNS. Although the syndromes share common paths in pathogenesis, they may differ in the disease course, prognosis, and therapy targets, depending on the localization and type of antibody epitope. Neuropathologic hallmarks of PNS associated with antibodies directed against intracellular epitopes are characterized by T cell-dominated inflammation, reactive gliosis including microglial nodules, and neuronal degeneration. By contrast, the neuropathology of cell surface antibody-mediated PNS strongly depends on the targeted antigen and varies from B cell/plasma cell-dominated inflammation and well-preserved neurons together with a reduced expression of the target antigen in anti-NMDAR encephalitis to irreversible Purkinje cell loss in anti-P/Q-type VGCC antibody-associated paraneoplastic cerebellar degeneration. The understanding of different pathomechanisms in PNS is important because they strongly correspond with therapy response and prognosis, and should guide treatment decisions.
Collapse
Affiliation(s)
- Valérie Quinot
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Romana Höftberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
14
|
Sourisse JM, Bonzi LC, Semmelhack J, Schunter C. Warming affects routine swimming activity and novel odour response in larval zebrafish. Sci Rep 2023; 13:21075. [PMID: 38030737 PMCID: PMC10687225 DOI: 10.1038/s41598-023-48287-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/24/2023] [Indexed: 12/01/2023] Open
Abstract
Temperature is a primary factor affecting the physiology of ectothermic animals and global warming of water bodies may therefore impact aquatic life. Understanding the effects of near-future predicted temperature changes on the behaviour and underlying molecular mechanisms of aquatic animals is of particular importance, since behaviour mediates survival. In this study, we investigate the effects of developmental temperature on locomotory behaviour and olfactory learning in the zebrafish, Danio rerio. We exposed zebrafish from embryonic stage to either control (28 °C) or elevated temperature (30 °C) for seven days. Overall, warming reduced routine swimming activity and caused upregulation of metabolism and neuron development genes. When exposed to olfactory cues, namely catfish cue, a non-alarming but novel odour, and conspecifics alarming cue, warming differently affected the larvae response to the two cues. An increase in locomotory activity and a large transcriptional reprogramming was observed at elevated temperature in response to novel odour, with upregulation of cell signalling, neuron development and neuron functioning genes. As this response was coupled with the downregulation of genes involved in protein translation and ATP metabolism, novel odour recognition in future-predicted thermal conditions would require energetic trade-offs between expensive baseline processes and responsive functions. To evaluate their learning abilities at both temperatures, larvae were conditioned with a mixture of conspecifics alarm cue and catfish cue. Regardless of temperature, no behavioural nor gene expression changes were detected, reinforcing our findings that warming mainly affects zebrafish molecular response to novel odours. Overall, our results show that future thermal conditions will likely impact developing stages, causing trade-offs following novel olfactory detection in the environment.
Collapse
Affiliation(s)
- Jade M Sourisse
- The Swire Institute of Marine Science, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong SAR, China
| | - Lucrezia C Bonzi
- The Swire Institute of Marine Science, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong SAR, China
| | - Julie Semmelhack
- The Division of Life Science, Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clearwater Bay, Kowloon, Hong Kong SAR, China
| | - Celia Schunter
- The Swire Institute of Marine Science, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
15
|
St George-Hyslop F, Haneklaus M, Kivisild T, Livesey FJ. Loss of CNTNAP2 Alters Human Cortical Excitatory Neuron Differentiation and Neural Network Development. Biol Psychiatry 2023; 94:780-791. [PMID: 37001843 DOI: 10.1016/j.biopsych.2023.03.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 05/14/2023]
Abstract
BACKGROUND Loss-of-function mutations in the contactin-associated protein-like 2 (CNTNAP2) gene are causal for neurodevelopmental disorders, including autism, schizophrenia, epilepsy, and intellectual disability. CNTNAP2 encodes CASPR2, a single-pass transmembrane protein that belongs to the neurexin family of cell adhesion molecules. These proteins have a variety of functions in developing neurons, including connecting presynaptic and postsynaptic neurons, and mediating signaling across the synapse. METHODS To study the effect of loss of CNTNAP2 function on human cerebral cortex development, and how this contributes to the pathogenesis of neurodevelopmental disorders, we generated human induced pluripotent stem cells from one neurotypical control donor null for full-length CNTNAP2, modeling cortical development from neurogenesis through to neural network formation in vitro. RESULTS CNTNAP2 is particularly highly expressed in the first two populations of early-born excitatory cortical neurons, and loss of CNTNAP2 shifted the relative proportions of these two neuronal types. Live imaging of excitatory neuronal growth showed that loss of CNTNAP2 reduced neurite branching and overall neuronal complexity. At the network level, developing cortical excitatory networks null for CNTNAP2 had complex changes in activity compared with isogenic controls: an initial period of relatively reduced activity compared with isogenic controls, followed by a lengthy period of hyperexcitability, and then a further switch to reduced activity. CONCLUSIONS Complete loss of CNTNAP2 contributes to the pathogenesis of neurodevelopmental disorders through complex changes in several aspects of human cerebral cortex excitatory neuron development that culminate in aberrant neural network formation and function.
Collapse
Affiliation(s)
- Frances St George-Hyslop
- University College London Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, University College London, London, United Kingdom
| | - Moritz Haneklaus
- University College London Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, University College London, London, United Kingdom
| | - Toomas Kivisild
- Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, Estonia; Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Frederick J Livesey
- University College London Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, University College London, London, United Kingdom.
| |
Collapse
|
16
|
Robinson BG, Oster BA, Robertson K, Kaltschmidt JA. Loss of ASD-related molecule Cntnap2 affects colonic motility in mice. Front Neurosci 2023; 17:1287057. [PMID: 38027494 PMCID: PMC10665486 DOI: 10.3389/fnins.2023.1287057] [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: 09/01/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Gastrointestinal (GI) symptoms are highly prevalent among individuals with autism spectrum disorder (ASD), but the molecular link between ASD and GI dysfunction remains poorly understood. The enteric nervous system (ENS) is critical for normal GI motility and has been shown to be altered in mouse models of ASD and other neurological disorders. Contactin-associated protein-like 2 (Cntnap2) is an ASD-related synaptic cell-adhesion molecule important for sensory processing. In this study, we examine the role of Cntnap2 in GI motility by characterizing Cntnap2's expression in the ENS and assessing GI function in Cntnap2 mutant mice. We find Cntnap2 expression predominately in enteric sensory neurons. We further assess in vivo and ex vivo GI motility in Cntnap2 mutants and show altered transit time and colonic motility patterns. The overall organization of the ENS appears undisturbed. Our results suggest that Cntnap2 plays a role in GI function and may provide a molecular link between ASD and GI dysfunction.
Collapse
Affiliation(s)
- Beatriz G. Robinson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, United States
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA, United States
| | - Beau A. Oster
- Nevada ENDURE Program, University of Nevada, Reno, Reno, NV, United States
| | - Keiramarie Robertson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, United States
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA, United States
| | - Julia A. Kaltschmidt
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, United States
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
17
|
Ciancone-Chama AG, Bonaldo V, Biasini E, Bozzi Y, Balasco L. Gene Expression Profiling in Trigeminal Ganglia from Cntnap2 -/- and Shank3b -/- Mouse Models of Autism Spectrum Disorder. Neuroscience 2023; 531:75-85. [PMID: 37699442 DOI: 10.1016/j.neuroscience.2023.08.028] [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: 01/19/2023] [Revised: 08/19/2023] [Accepted: 08/22/2023] [Indexed: 09/14/2023]
Abstract
Sensory difficulties represent a crucial issue in the life of autistic individuals. The diagnostic and statistical manual of mental disorders describes both hyper- and hypo-responsiveness to sensory stimulation as a criterion for the diagnosis autism spectrum disorders (ASD). Among the sensory domain affected in ASD, altered responses to tactile stimulation represent the most commonly reported sensory deficits. Although tactile abnormalities have been reported in monogenic cohorts of patients and genetic mouse models of ASD, the underlying mechanisms are still unknown. Traditionally, autism research has focused on the central nervous system as the target to infer the neurobiological bases of such tactile abnormalities. Nonetheless, the peripheral nervous system represents the initial site of processing of sensory information and a potential site of dysfunction in the sensory cascade. Here we investigated the gene expression deregulation in the trigeminal ganglion (which directly receives tactile information from whiskers) in two genetic models of syndromic autism (Shank3b and Cntnap2 mutant mice) at both adult and juvenile ages. We found several neuronal and non-neuronal markers involved in inhibitory, excitatory, neuroinflammatory and sensory neurotransmission to be differentially regulated within the trigeminal ganglia of both adult and juvenile Shank3b and Cntnap2 mutant mice. These results may help in disentangling the multifaced complexity of sensory abnormalities in autism and open avenues for the development of peripherally targeted treatments for tactile sensory deficits exhibited in ASD.
Collapse
Affiliation(s)
- Alessandra G Ciancone-Chama
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy
| | - Valerio Bonaldo
- Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Via Sommarive 9, 38123 Povo, TN, Italy
| | - Emiliano Biasini
- Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Via Sommarive 9, 38123 Povo, TN, Italy
| | - Yuri Bozzi
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy; CNR Neuroscience Institute, via Moruzzi 1, 56124 Pisa, Italy.
| | - Luigi Balasco
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy.
| |
Collapse
|
18
|
Khoodoruth MAS, Ahammad F, Khan YS, Mohammad F. The shared genetic risk factors between Tourette syndrome and obsessive-compulsive disorder. Front Neurol 2023; 14:1283572. [PMID: 37905190 PMCID: PMC10613519 DOI: 10.3389/fneur.2023.1283572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 09/25/2023] [Indexed: 11/02/2023] Open
Abstract
Tourette syndrome (TS) and obsessive-compulsive disorder (OCD) are two neuropsychiatric disorders that frequently co-occur. Previous evidence suggests a shared genetic diathesis underlying the comorbidity of TS and OCD. This review aims to comprehensively summarize the current literature on the genetic factors linked with TS and its comorbidities, with a focus on OCD. Family studies, linkage analysis, cytogenetic studies, and genome-wide association studies (GWAS) have played a pivotal role in identifying common and rare genetic variants connected with TS and OCD. Although the genetic framework of TS and OCD is complex and multifactorial, several susceptibility loci and candidate genes have been identified that might play a crucial role in the pathogenesis of both disorders. Additionally, post-infectious environmental elements have also been proposed to contribute to the development of TS-OCD, although the dynamics between genetic and environmental factors is not yet fully understood. International collaborations and studies with well-defined phenotypes will be crucial in the future to further elucidate the genetic basis of TS and OCD and to develop targeted therapeutic strategies for individuals suffering from these debilitating conditions.
Collapse
Affiliation(s)
- Mohamed Adil Shah Khoodoruth
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Child and Adolescent Mental Health Service, Hamad Medical Corporation, Doha, Qatar
| | - Foysal Ahammad
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Yasser Saeed Khan
- Child and Adolescent Mental Health Service, Hamad Medical Corporation, Doha, Qatar
| | - Farhan Mohammad
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
19
|
Robinson BG, Oster BA, Robertson K, Kaltschmidt JA. Loss of ASD-Related Molecule Cntnap2 Affects Colonic Motility in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.17.537221. [PMID: 37131706 PMCID: PMC10153124 DOI: 10.1101/2023.04.17.537221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Gastrointestinal (GI) symptoms are highly prevalent among individuals with autism spectrum disorder (ASD), but the molecular link between ASD and GI dysfunction remains poorly understood. The enteric nervous system (ENS) is critical for normal GI motility and has been shown to be altered in mouse models of ASD and other neurological disorders. Contactin-associated protein-like 2 (Cntnap2) is an ASD-related synaptic cell-adhesion molecule important for sensory processing. In this study, we examine the role of Cntnap2 in GI motility by characterizing Cntnap2's expression in the ENS and assessing GI function in Cntnap2 mutant mice. We find Cntnap2 expression predominately in enteric sensory neurons. We further assess in-vivo and ex-vivo GI motility in Cntnap2 mutants and show altered transit time and colonic motility patterns. The overall organization of the ENS appears undisturbed. Our results suggest that Cntnap2 plays a role in GI function and may provide a molecular link between ASD and GI dysfunction.
Collapse
Affiliation(s)
- Beatriz G. Robinson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Beau A. Oster
- Nevada ENDURE Program, University of Nevada, Reno, Reno, NV 89557, USA
| | - Keiramarie Robertson
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
- Neurosciences IDP Graduate Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Julia A. Kaltschmidt
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA 94305, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
20
|
Müller-Miny L, Sauer R, Schulte-Mecklenbeck A, Gross CC, Kovac S, Schilling M, Beuker C, Wiendl H, Meyer zu Hörste G. Contactin-associated protein 2 autoantibodies can be associated with multifocal motor-like neuropathy: a case report. Ther Adv Neurol Disord 2023; 16:17562864231189323. [PMID: 37599705 PMCID: PMC10434843 DOI: 10.1177/17562864231189323] [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: 04/13/2023] [Accepted: 07/03/2023] [Indexed: 08/22/2023] Open
Abstract
Autoantibodies against contactin-associated protein 2 (CASPR2) are usually associated with autoimmune encephalitis and neuromyotonia. Their association with inflammatory neuropathies has been described in case reports albeit all with distal symmetric manifestation. Here, we report a patient who developed distal arm paresis, dominantly of the right arm, over the course of 1 year. Electroneurography showed a conduction block of motor nerve conduction, nerve ultrasonography a swelling of the right median and ulnar nerve and flow cytometry an increase in natural killer (NK cells) in the blood and natural killer T (NKT) cells in the cerebrospinal fluid (CSF), therefore indicating a multifocal motor neuropathy-like (MMN-like) phenotype. CASPR2 autoantibodies were detected in serum and CSF. Through immunotherapy with intravenous immunoglobulins the patient showed clinical and neurographic improvement. We therefore describe the first association of CASPR2 autoantibodies with a MMN-like clinical manifestation, extending the spectrum of CASPR2-associated diseases.
Collapse
Affiliation(s)
- Louisa Müller-Miny
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Raoul Sauer
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Andreas Schulte-Mecklenbeck
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Catharina C. Gross
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Stjepana Kovac
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Matthias Schilling
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Carolin Beuker
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Gerd Meyer zu Hörste
- Department of Neurology with Institute of Translational Neurology, University of Münster, Albert-Schweitzer-Campus 1, Building A1, Münster 48149, Germany
| |
Collapse
|
21
|
Dawson MS, Gordon-Fleet K, Yan L, Tardos V, He H, Mui K, Nawani S, Asgarian Z, Catani M, Fernandes C, Drescher U. Sexual dimorphism in the social behaviour of Cntnap2-null mice correlates with disrupted synaptic connectivity and increased microglial activity in the anterior cingulate cortex. Commun Biol 2023; 6:846. [PMID: 37582968 PMCID: PMC10427688 DOI: 10.1038/s42003-023-05215-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 08/04/2023] [Indexed: 08/17/2023] Open
Abstract
A biological understanding of the apparent sex bias in autism is lacking. Here we have identified Cntnap2 KO mice as a model system to help better understand this dimorphism. Using this model, we observed social deficits in juvenile male KO mice only. These male-specific social deficits correlated with reduced spine densities of Layer 2/3 and Layer 5 pyramidal neurons in the Anterior Cingulate Cortex, a forebrain region prominently associated with the control of social behaviour. Furthermore, in male KO mice, microglia showed an increased activated morphology and phagocytosis of synaptic structures compared to WT mice, whereas no differences were seen in female KO and WT mice. Our data suggest that sexually dimorphic microglial activity may be involved in the aetiology of ASD, disrupting the development of neural circuits that control social behaviour by overpruning synapses at a developmentally critical period.
Collapse
Affiliation(s)
- Matt S Dawson
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Kevin Gordon-Fleet
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Lingxin Yan
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Vera Tardos
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Huanying He
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Kwong Mui
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
| | - Smriti Nawani
- Social, Genetic & Developmental Psychiatry Centre, IoPPN, King's College London, London, SE1 1UL, UK
| | - Zeinab Asgarian
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK
- Molecular Therapeutics Lab, University College London, Research Department of Targeted Intervention, London, W1W 7TY, UK
| | - Marco Catani
- NatBrainLab, Departments of Neuroimaging Sciences and Forensic and Neurodevelopmental Sciences, IoPPN, King's College London, London, SE1 1UL, UK
| | - Cathy Fernandes
- Social, Genetic & Developmental Psychiatry Centre, IoPPN, King's College London, London, SE1 1UL, UK
- MRC Centre for Neurodevelopmental Disorders, IoPPN, King's College London, London, SE1 1UL, UK
| | - Uwe Drescher
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, SE1 1UL, UK.
- MRC Centre for Neurodevelopmental Disorders, IoPPN, King's College London, London, SE1 1UL, UK.
| |
Collapse
|
22
|
D'Onofrio G, Accogli A, Severino M, Caliskan H, Kokotović T, Blazekovic A, Jercic KG, Markovic S, Zigman T, Goran K, Barišić N, Duranovic V, Ban A, Borovecki F, Ramadža DP, Barić I, Fazeli W, Herkenrath P, Marini C, Vittorini R, Gowda V, Bouman A, Rocca C, Alkhawaja IA, Murtaza BN, Rehman MMU, Al Alam C, Nader G, Mancardi MM, Giacomini T, Srivastava S, Alvi JR, Tomoum H, Matricardi S, Iacomino M, Riva A, Scala M, Madia F, Pistorio A, Salpietro V, Minetti C, Rivière JB, Srour M, Efthymiou S, Maroofian R, Houlden H, Vernes SC, Zara F, Striano P, Nagy V. Genotype-phenotype correlation in contactin-associated protein-like 2 (CNTNAP-2) developmental disorder. Hum Genet 2023; 142:909-925. [PMID: 37183190 PMCID: PMC10329570 DOI: 10.1007/s00439-023-02552-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 04/03/2023] [Indexed: 05/16/2023]
Abstract
Contactin-associated protein-like 2 (CNTNAP2) gene encodes for CASPR2, a presynaptic type 1 transmembrane protein, involved in cell-cell adhesion and synaptic interactions. Biallelic CNTNAP2 loss has been associated with "Pitt-Hopkins-like syndrome-1" (MIM#610042), while the pathogenic role of heterozygous variants remains controversial. We report 22 novel patients harboring mono- (n = 2) and bi-allelic (n = 20) CNTNAP2 variants and carried out a literature review to characterize the genotype-phenotype correlation. Patients (M:F 14:8) were aged between 3 and 19 years and affected by global developmental delay (GDD) (n = 21), moderate to profound intellectual disability (n = 17) and epilepsy (n = 21). Seizures mainly started in the first two years of life (median 22.5 months). Antiseizure medications were successful in controlling the seizures in about two-thirds of the patients. Autism spectrum disorder (ASD) and/or other neuropsychiatric comorbidities were present in nine patients (40.9%). Nonspecific midline brain anomalies were noted in most patients while focal signal abnormalities in the temporal lobes were noted in three subjects. Genotype-phenotype correlation was performed by also including 50 previously published patients (15 mono- and 35 bi-allelic variants). Overall, GDD (p < 0.0001), epilepsy (p < 0.0001), hyporeflexia (p = 0.012), ASD (p = 0.009), language impairment (p = 0.020) and severe cognitive impairment (p = 0.031) were significantly associated with the presence of biallelic versus monoallelic variants. We have defined the main features associated with biallelic CNTNAP2 variants, as severe cognitive impairment, epilepsy and behavioral abnormalities. We propose CASPR2-deficiency neurodevelopmental disorder as an exclusively recessive disease while the contribution of heterozygous variants is less likely to follow an autosomal dominant inheritance pattern.
Collapse
Affiliation(s)
- Gianluca D'Onofrio
- Department of Neurosciences Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Via Gerolamo Gaslini 5, 16147, Genoa, Italy
| | - Andrea Accogli
- Division of Medical Genetics, Department of Specialized Medicine, Montreal Children's Hospital, McGill University Health Centre (MUHC), Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | | | - Haluk Caliskan
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
| | - Tomislav Kokotović
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Antonela Blazekovic
- Department for Functional Genomics, Center for Translational and Clinical Research, University of Zagreb School of Medicine, Zagreb University Hospital Center, Zagreb, Croatia
| | - Kristina Gotovac Jercic
- Department for Functional Genomics, Center for Translational and Clinical Research, University of Zagreb School of Medicine, Zagreb University Hospital Center, Zagreb, Croatia
- Department of Neurology, University Hospital Center Zagreb, University of Zagreb School of Medicine, 10000, Zagreb, Croatia
| | - Silvana Markovic
- Department of Pediatrics, General Hospital dr. Tomislav Bardek Koprivnica, Koprivnica, Croatia
| | - Tamara Zigman
- Department of Paediatrics, University Hospital Center Zagreb and University of Zagreb School of Medicine, Zagreb, Croatia
| | - Krnjak Goran
- Department of Pediatrics, Varazdin General Hospital, Varazdin, Croatia
| | - Nina Barišić
- Department of Paediatrics, University Hospital Center Zagreb and University of Zagreb School of Medicine, Zagreb, Croatia
| | - Vlasta Duranovic
- Division of Neuropediatrics, Department of Pediatrics, Children's Hospital Zagre, Zagreb, Croatia
| | - Ana Ban
- Division of Neuropediatrics, Department of Pediatrics, Children's Hospital Zagre, Zagreb, Croatia
| | - Fran Borovecki
- Department for Functional Genomics, Center for Translational and Clinical Research, University of Zagreb School of Medicine, Zagreb University Hospital Center, Zagreb, Croatia
- Department of Neurology, University Hospital Center Zagreb, University of Zagreb School of Medicine, 10000, Zagreb, Croatia
| | - Danijela Petković Ramadža
- Department of Paediatrics, University Hospital Center Zagreb and University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ivo Barić
- Department of Paediatrics, University Hospital Center Zagreb and University of Zagreb School of Medicine, Zagreb, Croatia
| | - Walid Fazeli
- Department of Pediatric Neurology, University Hospital Bonn, Bonn, Germany
| | - Peter Herkenrath
- Department of Pediatric Neurology, University Hospital Bonn, Bonn, Germany
| | - Carla Marini
- Child Neurology and Psychiatry Unit Children's Hospital "G. Salesi" Azienda Ospedaliero-Universitaria delle Marche Ancona, Ancona, Italy
| | - Roberta Vittorini
- Department of Pediatrics, Regina Margherita Children's Hospital, Turin, Italy
| | - Vykuntaraju Gowda
- Department of Pediatric Neurology, Indira Gandhi Institute of Child Health, Bangalore, India
| | - Arjan Bouman
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Clarissa Rocca
- Department of Neuromuscular Disorders, UCL Institute of Neurology, Queen Square, London, WC1N 3BG, UK
| | - Issam Azmi Alkhawaja
- Pediatric Neurology Unit, Pediatric Department, Albashir Hospital, Amman, Jordan
| | - Bibi Nazia Murtaza
- Department of Zoology, Abbottabad University of Science and Technology, Abbottabad, Pakistan
| | - Malik Mujaddad Ur Rehman
- Department of Microbiology, Abbottabad University of Science and Technology KP, Abbottabad, Pakistan
| | - Chadi Al Alam
- Division of Pediatric Neurology, Department of Pediatrics, American University of Beirut Medical Center, Beirut, Lebanon
| | - Gisele Nader
- Division of Pediatric Neurology, Department of Pediatrics, American University of Beirut Medical Center, Beirut, Lebanon
| | - Maria Margherita Mancardi
- Unit of Child Neuropsychiatry, Department of Clinical and Surgical Neuroscience and Rehabilitation, Epilepsy Center, EPICARE Reference Network, IRCCS Giannina Gaslini, Genoa, Italy
| | - Thea Giacomini
- Unit of Child Neuropsychiatry, Department of Clinical and Surgical Neuroscience and Rehabilitation, Epilepsy Center, EPICARE Reference Network, IRCCS Giannina Gaslini, Genoa, Italy
| | - Siddharth Srivastava
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Javeria Raza Alvi
- Department of Pediatric Neurology, Institute of Child Health, Children's Hospital Lahore, Lahore, Pakistan
| | - Hoda Tomoum
- Department of Pediatrics, Ain Shams University, Cairo, Egypt
| | - Sara Matricardi
- Department of Pediatrics, University of Chieti, Chieti, Italy
| | - Michele Iacomino
- Unit of Medical Genetics-IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Antonella Riva
- Department of Neurosciences Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Via Gerolamo Gaslini 5, 16147, Genoa, Italy
| | - Marcello Scala
- Department of Neurosciences Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Via Gerolamo Gaslini 5, 16147, Genoa, Italy
| | - Francesca Madia
- Unit of Medical Genetics-IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Angela Pistorio
- Epidemiology and Biostatistics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Vincenzo Salpietro
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Carlo Minetti
- Department of Neurosciences Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Via Gerolamo Gaslini 5, 16147, Genoa, Italy
| | - Jean-Baptiste Rivière
- Department of Human Genetics, Faculty of Medicine, McGill University, Montreal, QC, H3A 1B1, Canada
- Bioinformatics Platform, Research Institute of the McGill University Health Centre, Montréal, QC, H4A 3J1, Canada
| | - Myriam Srour
- Research Institute, McGill University Health Centre, Montreal, QC, Canada
- Division of Pediatric Neurology, Department of Pediatrics, McGill University, Montreal, QC, Canada
| | - Stephanie Efthymiou
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Reza Maroofian
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Henry Houlden
- Department of Clinical Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Sonja Catherine Vernes
- School of Biology, The University of St Andrews, Fife, UK
- Neurogenetics of Vocal Communication, Max Planck Institute for Psycholinguistics, Nijmegen, The Netherlands
| | - Federico Zara
- Department of Neurosciences Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Via Gerolamo Gaslini 5, 16147, Genoa, Italy
- Department of Pediatrics, University of Chieti, Chieti, Italy
| | - Pasquale Striano
- Department of Neurosciences Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), University of Genoa, Via Gerolamo Gaslini 5, 16147, Genoa, Italy.
- Pediatric Neurology and Muscular Diseases Unit, IRCCS Istituto "Giannina Gaslini", Via Gerolamo Gaslini 5, 16147, Genoa, Italy.
| | - Vanja Nagy
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
23
|
Guise AJ, Misal SA, Carson R, Boekweg H, Watt DVD, Truong T, Liang Y, Chu JH, Welsh NC, Gagnon J, Payne SH, Plowey ED, Kelly RT. TDP-43-stratified single-cell proteomic profiling of postmortem human spinal motor neurons reveals protein dynamics in amyotrophic lateral sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.08.544233. [PMID: 37333094 PMCID: PMC10274884 DOI: 10.1101/2023.06.08.544233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Unbiased proteomics has been employed to interrogate central nervous system (CNS) tissues (brain, spinal cord) and fluid matrices (CSF, plasma) from amyotrophic lateral sclerosis (ALS) patients; yet, a limitation of conventional bulk tissue studies is that motor neuron (MN) proteome signals may be confounded by admixed non-MN proteins. Recent advances in trace sample proteomics have enabled quantitative protein abundance datasets from single human MNs (Cong et al., 2020b). In this study, we leveraged laser capture microdissection (LCM) and nanoPOTS (Zhu et al., 2018c) single-cell mass spectrometry (MS)-based proteomics to query changes in protein expression in single MNs from postmortem ALS and control donor spinal cord tissues, leading to the identification of 2515 proteins across MNs samples (>900 per single MN) and quantitative comparison of 1870 proteins between disease groups. Furthermore, we studied the impact of enriching/stratifying MN proteome samples based on the presence and extent of immunoreactive, cytoplasmic TDP-43 inclusions, allowing identification of 3368 proteins across MNs samples and profiling of 2238 proteins across TDP-43 strata. We found extensive overlap in differential protein abundance profiles between MNs with or without obvious TDP-43 cytoplasmic inclusions that together point to early and sustained dysregulation of oxidative phosphorylation, mRNA splicing and translation, and retromer-mediated vesicular transport in ALS. Our data are the first unbiased quantification of single MN protein abundance changes associated with TDP-43 proteinopathy and begin to demonstrate the utility of pathology-stratified trace sample proteomics for understanding single-cell protein abundance changes in human neurologic diseases.
Collapse
Affiliation(s)
| | - Santosh A. Misal
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602
| | - Richard Carson
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602
| | - Hannah Boekweg
- Biology Department, Brigham Young University, Provo, UT 84602, USA
| | | | - Thy Truong
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602
| | - Yiran Liang
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602
| | | | | | | | - Samuel H. Payne
- Biology Department, Brigham Young University, Provo, UT 84602, USA
| | | | - Ryan T. Kelly
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602
| |
Collapse
|
24
|
Zhang J, Cai F, Lu R, Xing X, Xu L, Wu K, Gong Z, Zhang Q, Zhang Y, Xing M, Song W, Li JD. CNTNAP2 intracellular domain (CICD) generated by γ-secretase cleavage improves autism-related behaviors. Signal Transduct Target Ther 2023; 8:219. [PMID: 37271769 DOI: 10.1038/s41392-023-01431-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/03/2023] [Accepted: 03/24/2023] [Indexed: 06/06/2023] Open
Abstract
As the most prevalent neurodevelopmental disorders in children, autism spectrum disorders (ASD) are characterized by deficits in language development, social interaction, and repetitive behaviors or inflexible interests. Contactin associated protein like 2 (CNTNAP2), encoding a single transmembrane protein (CNTNAP2) with 1331 amino acid residues, is a widely validated ASD-susceptible gene. Cntnap2-deficient mice also show core autism-relevant behaviors, including the social deficits and repetitive behavior. However, the cellular mechanisms underlying dysfunction CNTNAP2 and ASD remain elusive. In this study, we found a motif within the transmembrane domain of CNTNAP2 was highly homologous to the γ-secretase cleavage site of amyloid-β precursor protein (APP), suggesting that CNTNAP2 may undergo proteolytic cleavage. Further biochemical analysis indicated that CNTNAP2 is cleaved by γ-secretase to produce the CNTNAP2 intracellular domain (CICD). Virally delivery of CICD to the medial prefrontal cortex (mPFC) in Cntnap2-deficient (Cntnap2-/-) mice normalized the deficit in the ASD-related behaviors, including social deficit and repetitive behaviors. Furthermore, CICD promoted the nuclear translocation of calcium/calmodulin-dependent serine protein kinase (CASK) to regulate the transcription of genes, such as Prader Willi syndrome gene Necdin. Whereas Necdin deficiency led to reduced social interaction in mice, virally expression of Necdin in the mPFC normalized the deficit in social preference of Cntnap2-/- mice. Our results thus reveal a critical function of CICD and highlight a role of the CNTNAP2-CASK-Necdin signaling pathway in ASD.
Collapse
Affiliation(s)
- Jing Zhang
- Furong Laboratory, Center for Medical Genetics, Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Fang Cai
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Renbin Lu
- Furong Laboratory, Center for Medical Genetics, Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Xiaoliang Xing
- Furong Laboratory, Center for Medical Genetics, Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Lu Xu
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Kunyang Wu
- Furong Laboratory, Center for Medical Genetics, Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Zishan Gong
- Furong Laboratory, Center for Medical Genetics, Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China
| | - Qing Zhang
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Yun Zhang
- Advanced Innovation Center for Human Brain Protection, The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Mengen Xing
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Weihong Song
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China.
- Townsend Family Laboratories, Department of Psychiatry, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Jia-Da Li
- Furong Laboratory, Center for Medical Genetics, Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics, Hunan International Scientific and Technological Cooperation Base of Animal Models for Human Diseases, School of Life Sciences, Central South University, Changsha, 410078, Hunan, China.
| |
Collapse
|
25
|
Zhang Q, Sterling K, Xu L, Xing M, Cai F, Yu S, Bestard-Lorigados I, Song W. CNTNAP2 Protein Is Degraded by the Ubiquitin-Proteasome System and the Macroautophagy-Lysosome Pathway. Mol Neurobiol 2023; 60:2455-2469. [PMID: 36658382 DOI: 10.1007/s12035-023-03227-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 01/06/2023] [Indexed: 01/21/2023]
Abstract
Contactin-associated protein-like 2 (CNTNAP2) gene, located on chromosome 7q35, is one of the largest genes in the human genome. CNTNAP2 protein is a type-I transmembrane protein specifically expressed in the nervous system, with versatile roles in the axonal organization, synaptic functions, neuronal migration, and functional connectivity. CNTNAP2 has been widely investigated as a risk gene for autism spectrum disorder (ASD), and recent studies also implicated CNTNAP2 in Alzheimer's disease (AD). Knowledge of the regulations on CNTNAP2's life cycle is necessary for understanding the related physiological functions and pathological conditions. However, the mechanisms underlying CNTNAP2 protein degradation remain elusive. Therefore, we systematically investigated the half-life and degradation pathway of the human CNTNAP2 protein. We discovered that CNTNAP2 has C-terminal fragments (CTF), which may have essential physiological functions. Our results demonstrated that CNTNAP2 full-length protein and CTF have a short half-life of about 3-4 h. CNTNAP2 proteins are degraded by the ubiquitin-proteasome system and the macroautophagy-lysosome pathway, while the lysosome pathway is more common for CNTNAP2 degradation. This study will provide novel insights and valuable tools for CNTNAP2 functional research in physiological and pathological scenarios.
Collapse
Affiliation(s)
- Qing Zhang
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Lu Xu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Mengen Xing
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Fang Cai
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Sheng Yu
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Isabel Bestard-Lorigados
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Weihong Song
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and Kangning Hospital, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
26
|
Zhang Q, Xu L, Bai Y, Chen P, Xing M, Cai F, Wu Y, Song W. Intermittent hypoxia-induced enhancement of sociability and working memory associates with CNTNAP2 upregulation. Front Mol Neurosci 2023; 16:1155047. [PMID: 37089693 PMCID: PMC10118049 DOI: 10.3389/fnmol.2023.1155047] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/22/2023] [Indexed: 04/08/2023] Open
Abstract
IntroductionHypoxia is an environmental risk factor for many disorders throughout life. Perinatal hypoxia contributes to autism spectrum disorder (ASD), while hypoxic conditions in the elderly facilitate memory deficits. However, the effects of hypoxia on adolescence remains elusive. CNTNAP2 is a critical molecule in ASD pathogenesis with undefined mechanisms. We investigate hypoxia’s impact on adolescence and the underlying mechanism related to CNTNAP2.MethodsThree-chamber social approach test, Y maze, Morris Water Maze and Open Field Test were applied to evaluate behavioral alterations. Immunoblotting, 5′- RACE and dual-luciferase reporter assay were performed to examine CNTNAP2 protein expression, transcription start site (TSS) of human CNTNAP2 gene and CNTNAP2 promoter activity, respectively.ResultsIntermittent hypoxia treatment improved social behaviors and working memory in adolescent mice. CNTNAP2 was increased in the brains of hypoxia-treated mice. The sequencing results identified the TSS at 518 bp upstream of the translation start site ATG. Hypoxia upregulated CNTNAP2 by interacting with functional hypoxia response elements in CNTNAP2 promoter.ConclusionIntermittent hypoxia enhanced sociability and working memory associated with CNTNAP2 upregulation. Our study provides novel insights into intermittent hypoxia’s impact on development and the interaction between genetic and environmental risk factors in ASD pathogenesis.
Collapse
Affiliation(s)
- Qing Zhang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, Vancouver, BC, Canada
| | - Lu Xu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yang Bai
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Peiye Chen
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Mengen Xing
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fang Cai
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, Vancouver, BC, Canada
| | - Yili Wu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- *Correspondence: Yili Wu,
| | - Weihong Song
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Institute of Aging, Key Laboratory of Alzheimer’s Disease of Zhejiang Province, Zhejiang Provincial Clinical Research Center for Mental Disorders, School of Mental Health and Kangning Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, Vancouver, BC, Canada
- Weihong Song, ; orcid.org/0000-0001-9928-889X
| |
Collapse
|
27
|
Treatment-Resistant Schizophrenia, Clozapine Resistance, Genetic Associations, and Implications for Precision Psychiatry: A Scoping Review. Genes (Basel) 2023; 14:genes14030689. [PMID: 36980961 PMCID: PMC10048540 DOI: 10.3390/genes14030689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Treatment-resistant schizophrenia (TRS) is often associated with severe burden of disease, poor quality of life and functional impairment. Clozapine is the gold standard for the treatment of TRS, although it is also known to cause significant side effects in some patients. In view of the burgeoning interest in the role of genetic factors in precision psychiatry, we conducted a scoping review to narratively summarize the current genetic factors associated with TRS, clozapine resistance and side effects to clozapine treatment. We searched PubMed from inception to December 2022 and included 104 relevant studies in this review. Extant evidence comprised associations between TRS and clozapine resistance with genetic factors related to mainly dopaminergic and serotoninergic neurotransmitter systems, specifically, TRS and rs4680, rs4818 within COMT, and rs1799978 within DRD2; clozapine resistance and DRD3 polymorphisms, CYP1A2 polymorphisms; weight gain with LEP and SNAP-25 genes; and agranulocytosis risk with HLA-related polymorphisms. Future studies, including replication in larger multi-site samples, are still needed to elucidate putative risk genes and the interactions between different genes and their correlations with relevant clinical factors such as psychopathology, psychosocial functioning, cognition and progressive changes with treatment over time in TRS and clozapine resistance.
Collapse
|
28
|
Zheng A, Scott KE, Schormans AL, Mann R, Allman BL, Schmid S. Differences in Startle and Prepulse Inhibition in Contactin-associated Protein-like 2 Knock-out Rats are Associated with Sex-specific Alterations in Brainstem Neural Activity. Neuroscience 2023; 513:96-110. [PMID: 36708798 DOI: 10.1016/j.neuroscience.2023.01.020] [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: 08/05/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023]
Abstract
The contactin-associated protein-like 2 (CNTNAP2) gene encodes for the CASPR2 protein, which plays an essential role in neurodevelopment. Mutations in CNTNAP2 are associated with neurodevelopmental disorders, including autism spectrum disorder and schizophrenia. Rats with a loss of function mutation in the Cntnap2 gene show increased acoustic startle response (ASR) and decreased prepulse inhibition (PPI). The neural basis of this altered auditory processing in Cntnap2 knock-out rats is currently unknown. Auditory brainstem recordings previously revealed no differences between the genotypes. The next step is to investigate brainstem structures outside of the primary auditory pathway that mediate ASR and PPI, which are the pontine reticular nucleus (PnC) and pedunculopontine tegmentum (PPTg), respectively. Multi-unit responses from the PnC and PPTg in vivo of the same rats revealed sex-specific effects of loss of CASPR2 expression on PnC activity, but no effects on PPTg activity. Female Cntnap2-/- rats showed considerably increased PnC firing rates compared with female wildtypes, whereas the difference between the genotypes was modest in male rats. In contrast, for both females and males we found meager differences between the genotypes for PPTg firing rates and inhibition of PnC firing rates, indicating that altered firing rates of these brainstem structures are not responsible for decreased PPI in Cntnap2-/- rats. We conclude that the auditory processing changes seen in Cntnap2-/- rats are associated with, but cannot be fully explained by, differences in PnC firing rates, and that a loss of function mutation in the Cntnap2 gene has differential effects depending on sex.
Collapse
Affiliation(s)
- Alice Zheng
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, Canada
| | - Kaela E Scott
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, Canada
| | - Ashley L Schormans
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, Canada
| | - Rajkamalpreet Mann
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, Canada
| | - Brian L Allman
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, Canada
| | - Susanne Schmid
- Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, University of Western Ontario, Canada.
| |
Collapse
|
29
|
Ahmed NY, Knowles R, Liu L, Yan Y, Li X, Schumann U, Wang Y, Sontani Y, Reynolds N, Natoli R, Wen J, Del Pino I, Mi D, Dehorter N. Developmental deficits of MGE-derived interneurons in the Cntnap2 knockout mouse model of autism spectrum disorder. Front Cell Dev Biol 2023; 11:1112062. [PMID: 36819097 PMCID: PMC9930104 DOI: 10.3389/fcell.2023.1112062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
Interneurons are fundamental cells for maintaining the excitation-inhibition balance in the brain in health and disease. While interneurons have been shown to play a key role in the pathophysiology of autism spectrum disorder (ASD) in adult mice, little is known about how their maturation is altered in the developing striatum in ASD. Here, we aimed to track striatal developing interneurons and elucidate the molecular and physiological alterations in the Cntnap2 knockout mouse model. Using Stereo-seq and single-cell RNA sequencing data, we first characterized the pattern of expression of Cntnap2 in the adult brain and at embryonic stages in the medial ganglionic eminence (MGE), a transitory structure producing most cortical and striatal interneurons. We found that Cntnap2 is enriched in the striatum, compared to the cortex, particularly in the developing striatal cholinergic interneurons. We then revealed enhanced MGE-derived cell proliferation, followed by increased cell loss during the canonical window of developmental cell death in the Cntnap2 knockout mice. We uncovered specific cellular and molecular alterations in the developing Lhx6-expressing cholinergic interneurons of the striatum, which impacts interneuron firing properties during the first postnatal week. Overall, our work unveils some of the mechanisms underlying the shift in the developmental trajectory of striatal interneurons which greatly contribute to the ASD pathogenesis.
Collapse
Affiliation(s)
- Noorya Yasmin Ahmed
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Rhys Knowles
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Lixinyu Liu
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Yiming Yan
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xiaohan Li
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Ulrike Schumann
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Yumeng Wang
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Yovina Sontani
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Nathan Reynolds
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Riccardo Natoli
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Jiayu Wen
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia
| | - Isabel Del Pino
- Institute of Neurosciences, Spanish National Research Council (CSIC), Sant Joan d’Alacant, Spain
| | - Da Mi
- Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, School of Life Sciences, Tsinghua University, Beijing, China
| | - Nathalie Dehorter
- The Australian National University, The John Curtin School of Medical Research, Canberra, ACT, Australia,*Correspondence: Nathalie Dehorter,
| |
Collapse
|
30
|
Cifuentes-Diaz C, Canali G, Garcia M, Druart M, Manett T, Savariradjane M, Guillaume C, Le Magueresse C, Goutebroze L. Differential impacts of Cntnap2 heterozygosity and Cntnap2 null homozygosity on axon and myelinated fiber development in mouse. Front Neurosci 2023; 17:1100121. [PMID: 36793543 PMCID: PMC9922869 DOI: 10.3389/fnins.2023.1100121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/09/2023] [Indexed: 01/31/2023] Open
Abstract
Over the last decade, a large variety of alterations of the Contactin Associated Protein 2 (CNTNAP2) gene, encoding Caspr2, have been identified in several neuronal disorders, including neurodevelopmental disorders and peripheral neuropathies. Some of these alterations are homozygous but most are heterozygous, and one of the current challenges is to estimate to what extent they could affect the functions of Caspr2 and contribute to the development of these pathologies. Notably, it is not known whether the disruption of a single CNTNAP2 allele could be sufficient to perturb the functions of Caspr2. To get insights into this issue, we questioned whether Cntnap2 heterozygosity and Cntnap2 null homozygosity in mice could both impact, either similarly or differentially, some specific functions of Caspr2 during development and in adulthood. We focused on yet poorly explored functions of Caspr2 in axon development and myelination, and performed a morphological study from embryonic day E17.5 to adulthood of two major brain interhemispheric myelinated tracts, the anterior commissure (AC) and the corpus callosum (CC), comparing wild-type (WT), Cntnap2 -/- and Cntnap2 +/- mice. We also looked for myelinated fiber abnormalities in the sciatic nerves of mutant mice. Our work revealed that Caspr2 controls the morphology of the CC and AC throughout development, axon diameter at early developmental stages, cortical neuron intrinsic excitability at the onset of myelination, and axon diameter and myelin thickness at later developmental stages. Changes in axon diameter, myelin thickness and node of Ranvier morphology were also detected in the sciatic nerves of the mutant mice. Importantly, most of the parameters analyzed were affected in Cntnap2 +/- mice, either specifically, more severely, or oppositely as compared to Cntnap2 -/- mice. In addition, Cntnap2 +/- mice, but not Cntnap2 -/- mice, showed motor/coordination deficits in the grid-walking test. Thus, our observations show that both Cntnap2 heterozygosity and Cntnap2 null homozygosity impact axon and central and peripheral myelinated fiber development, but in a differential manner. This is a first step indicating that CNTNAP2 alterations could lead to a multiplicity of phenotypes in humans, and raising the need to evaluate the impact of Cntnap2 heterozygosity on the other neurodevelopmental functions of Caspr2.
Collapse
Affiliation(s)
- Carmen Cifuentes-Diaz
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Giorgia Canali
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Marta Garcia
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Mélanie Druart
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Taylor Manett
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Mythili Savariradjane
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Camille Guillaume
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Corentin Le Magueresse
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France
| | - Laurence Goutebroze
- Inserm, Unité Mixte de Recherche (UMR)-S 1270, Paris, France,Faculté des Sciences et Ingénierie, Sorbonne University, Paris, France,Institut du Fer à Moulin, Paris, France,*Correspondence: Laurence Goutebroze,
| |
Collapse
|
31
|
Abad-Rodríguez J, Brocca ME, Higuero AM. Glycans and Carbohydrate-Binding/Transforming Proteins in Axon Physiology. ADVANCES IN NEUROBIOLOGY 2023; 29:185-217. [PMID: 36255676 DOI: 10.1007/978-3-031-12390-0_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The mature nervous system relies on the polarized morphology of neurons for a directed flow of information. These highly polarized cells use their somatodendritic domain to receive and integrate input signals while the axon is responsible for the propagation and transmission of the output signal. However, the axon must perform different functions throughout development before being fully functional for the transmission of information in the form of electrical signals. During the development of the nervous system, axons perform environmental sensing functions, which allow them to navigate through other regions until a final target is reached. Some axons must also establish a regulated contact with other cells before reaching maturity, such as with myelinating glial cells in the case of myelinated axons. Mature axons must then acquire the structural and functional characteristics that allow them to perform their role as part of the information processing and transmitting unit that is the neuron. Finally, in the event of an injury to the nervous system, damaged axons must try to reacquire some of their immature characteristics in a regeneration attempt, which is mostly successful in the PNS but fails in the CNS. Throughout all these steps, glycans perform functions of the outermost importance. Glycans expressed by the axon, as well as by their surrounding environment and contacting cells, encode key information, which is fine-tuned by glycan modifying enzymes and decoded by glycan binding proteins so that the development, guidance, myelination, and electrical transmission functions can be reliably performed. In this chapter, we will provide illustrative examples of how glycans and their binding/transforming proteins code and decode instructive information necessary for fundamental processes in axon physiology.
Collapse
Affiliation(s)
- José Abad-Rodríguez
- Membrane Biology and Axonal Repair Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain.
| | - María Elvira Brocca
- Membrane Biology and Axonal Repair Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain
| | - Alonso Miguel Higuero
- Membrane Biology and Axonal Repair Laboratory, Hospital Nacional de Parapléjicos (SESCAM), Toledo, Spain
| |
Collapse
|
32
|
Lee K, Mills Z, Cheung P, Cheyne JE, Montgomery JM. The Role of Zinc and NMDA Receptors in Autism Spectrum Disorders. Pharmaceuticals (Basel) 2022; 16:ph16010001. [PMID: 36678498 PMCID: PMC9866730 DOI: 10.3390/ph16010001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
NMDA-type glutamate receptors are critical for synaptic plasticity in the central nervous system. Their unique properties and age-dependent arrangement of subunit types underpin their role as a coincidence detector of pre- and postsynaptic activity during brain development and maturation. NMDAR function is highly modulated by zinc, which is co-released with glutamate and concentrates in postsynaptic spines. Both NMDARs and zinc have been strongly linked to autism spectrum disorders (ASDs), suggesting that NMDARs are an important player in the beneficial effects observed with zinc in both animal models and children with ASDs. Significant evidence is emerging that these beneficial effects occur via zinc-dependent regulation of SHANK proteins, which form the backbone of the postsynaptic density. For example, dietary zinc supplementation enhances SHANK2 or SHANK3 synaptic recruitment and rescues NMDAR deficits and hypofunction in Shank3ex13-16-/- and Tbr1+/- ASD mice. Across multiple studies, synaptic changes occur in parallel with a reversal of ASD-associated behaviours, highlighting the zinc-dependent regulation of NMDARs and glutamatergic synapses as therapeutic targets for severe forms of ASDs, either pre- or postnatally. The data from rodent models set a strong foundation for future translational studies in human cells and people affected by ASDs.
Collapse
|
33
|
Binks S, Lamquet S, Crawford AH, Meurs A, Irani SR, Pakozdy A. Parallel roles of neuroinflammation in feline and human epilepsies. Vet J 2022; 290:105912. [PMID: 36209994 PMCID: PMC10912827 DOI: 10.1016/j.tvjl.2022.105912] [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: 01/22/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/06/2022]
Abstract
Autoimmune encephalitis refers to a group of disorders characterised by a non-infectious encephalitis, often with prominent seizures and surface neuronal autoantibodies. AE is an important cause of new-onset refractory status epilepticus in humans and is frequently responsive to immunotherapies including corticosteroids, plasma exchange, intravenous immunoglobulin G and rituximab. Recent research suggests that parallel autoantibodies can be detected in non-human mammalian species. The best documented example is leucine-rich glioma-inactivated 1 (LGI1)-antibodies in domestic cats with limbic encephalitis (LE). In this review, we discuss the role of neuroinflammation and autoantibodies in human and feline epilepsy and LE.
Collapse
Affiliation(s)
- Sophie Binks
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, OX3 9DU, UK; Department of Neurology, John Radcliffe Hospital, Oxford University Hospitals Foundation Trust, Oxford OX3 9DU, UK.
| | - Simon Lamquet
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Abbe H Crawford
- Clinical Science and Services, The Royal Veterinary College, Hertfordshire AL9 7TA, UK
| | - Alfred Meurs
- Department of Neurology, Ghent University Hospital, Ghent, Belgium
| | - Sarosh R Irani
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, OX3 9DU, UK; Department of Neurology, John Radcliffe Hospital, Oxford University Hospitals Foundation Trust, Oxford OX3 9DU, UK
| | - Akos Pakozdy
- University Clinic for Small Animals, University of Veterinary Medicine Vienna, Austria
| |
Collapse
|
34
|
Dziadkowiak E, Nowakowska-Kotas M, Budrewicz S, Koszewicz M. Pathology of Initial Axon Segments in Chronic Inflammatory Demyelinating Polyradiculoneuropathy and Related Disorders. Int J Mol Sci 2022; 23:13621. [PMID: 36362407 PMCID: PMC9658771 DOI: 10.3390/ijms232113621] [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: 09/30/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 07/30/2023] Open
Abstract
The diagnosis of chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) is based on a combination of clinical, electrodiagnostic and laboratory features. The different entities of the disease include chronic immune sensory polyradiculopathy (CISP) and autoimmune nodopathies. It is debatable whether CIDP occurring in the course of other conditions, i.e., monoclonal IgG or IgA gammopathy, should be treated as a separate disease entity from idiopathic CIDP. This study aims to evaluate the molecular differences of the nodes of Ranvier and the initial axon segment (AIS) and juxtaparanode region (JXP) as the potential cause of phenotypic variation of CIDP while also seeking new pathomechanisms since JXP is sequestered behind the paranode and autoantibodies may not access the site easily. The authors initially present the structure of the different parts of the neuron and its functional significance, then discuss the problem of whether damage to the juxtaparanodal region, Schwann cells and axons could cause CIDP or if these damages should be separated as separate disease entities. In particular, AIS's importance for modulating neural excitability and carrying out transport along the axon is highlighted. The disclosure of specific pathomechanisms, including novel target antigens, in the heterogeneous CIDP syndrome is important for diagnosing and treating these patients.
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Autoimmune neuromyotonia encompasses a group of rare immune-mediated neurological disorders frequently associated with anti-contactin-associated protein-like 2 (CASPR2) antibodies and featuring clinical and electrical signs of peripheral nerve hyperexcitability (PNH). We aim to summarize the current knowledge on immune-mediated neuromyotonia, focusing on clinical presentations, pathophysiology, and management. RECENT FINDINGS Neuromyotonia is a major feature of several autoimmune neurological syndromes characterized by PNH with or without central neurological system involvement. Experimental and clinical evidence suggest that anti-CASPR2 antibodies are directly pathogenic in autoimmune neuromyotonia patients. SUMMARY Neuromyotonia, a form of PNH, is a major feature in several syndromes associated with anti-CASPR2 antibodies, including cramp-fasciculation syndrome, Isaacs syndrome, Morvan syndrome, and autoimmune limbic encephalitis. Diagnosis relies on the identification of motor, sensory, and autonomic signs of PNH along with other neurological symptoms, anti-CASPR2 antibody-positivity, and of characteristic electroneuromyographic abnormalities. Paraneoplastic associations with thymoma are possible, especially in Morvan syndrome. Patients usually respond to immune-active treatments, including steroids, intravenous immunoglobulins, plasma exchanges, and rituximab.
Collapse
Affiliation(s)
- Louis Comperat
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon
| | - Antoine Pegat
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon
- Electroneuromyography and Neuromuscular Diseases Unit, Pierre Wertheimer Hospital, Hospices Civils de Lyon
| | - Jérôme Honnorat
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon
- Synaptopathies and Autoantibodies (SynatAc) Team, Institut NeuroMyoGène, INSERM U1217/CNRS UMR 5310, Université Claude Bernard Lyon 1
| | - Bastien Joubert
- French Reference Center on Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon
- Synaptopathies and Autoantibodies (SynatAc) Team, Institut NeuroMyoGène, INSERM U1217/CNRS UMR 5310, Université Claude Bernard Lyon 1
- Department of Neurology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
36
|
Chen TS, Lai MC, Huang HYI, Wu SN, Huang CW. Immunity, Ion Channels and Epilepsy. Int J Mol Sci 2022; 23:ijms23126446. [PMID: 35742889 PMCID: PMC9224225 DOI: 10.3390/ijms23126446] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 12/10/2022] Open
Abstract
Epilepsy is a common chronic neurological disorder in modern society. One of the major unmet challenges is that current antiseizure medications are basically not disease-modifying. Among the multifaceted etiologies of epilepsy, the role of the immune system has attracted considerable attention in recent years. It is known that both innate and adaptive immunity can be activated in response to insults to the central nervous system, leading to seizures. Moreover, the interaction between ion channels, which have a well-established role in epileptogenesis and epilepsy, and the immune system is complex and is being actively investigated. Some examples, including the interaction between ion channels and mTOR pathways, will be discussed in this paper. Furthermore, there has been substantial progress in our understanding of the pathophysiology of epilepsy associated with autoimmune encephalitis, and numerous neural-specific autoantibodies have been found and documented. Early recognition of immune-mediated epilepsy is important, especially in cases of pharmacoresistant epilepsy and in the presence of signs of autoimmune encephalitis, as early intervention with immunotherapy shows promise.
Collapse
Affiliation(s)
- Tsang-Shan Chen
- Department of Neurology, Tainan Sin-Lau Hospital, Tainan 701002, Taiwan;
| | - Ming-Chi Lai
- Department of Pediatrics, Chi-Mei Medical Center, Tainan 71004, Taiwan;
| | | | - Sheng-Nan Wu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan;
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan 70101, Taiwan
| | - Chin-Wei Huang
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70101, Taiwan
- Correspondence: ; Tel.: +886-6-2353535 (ext. 5485)
| |
Collapse
|
37
|
Lim HK, Yoon JH, Song M. Autism Spectrum Disorder Genes: Disease-Related Networks and Compensatory Strategies. Front Mol Neurosci 2022; 15:922840. [PMID: 35726297 PMCID: PMC9206533 DOI: 10.3389/fnmol.2022.922840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/17/2022] [Indexed: 11/16/2022] Open
Abstract
The mammalian brain comprises structurally and functionally distinct regions. Each of these regions has characteristic molecular mechanisms that mediate higher-order tasks, such as memory, learning, emotion, impulse, and motor control. Many genes are involved in neuronal signaling and contribute to normal brain development. Dysfunction of essential components of neural signals leads to various types of brain disorders. Autism spectrum disorder is a neurodevelopmental disorder characterized by social deficits, communication challenges, and compulsive repetitive behaviors. Long-term genetic studies have uncovered key genes associated with autism spectrum disorder, such as SH3 and multiple ankyrin repeat domains 3, methyl-CpG binding protein 2, neurexin 1, and chromodomain helicase DNA binding protein 8. In addition, disease-associated networks have been identified using animal models, and the understanding of the impact of these genes on disease susceptibility and compensation is deepening. In this review, we examine rescue strategies using key models of autism spectrum disorder.
Collapse
Affiliation(s)
- Hye Kyung Lim
- Department of Life Sciences, Yeungnam University, Gyeongsan, South Korea
| | - Jong Hyuk Yoon
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan, South Korea
| |
Collapse
|
38
|
Nisar S, Bhat AA, Masoodi T, Hashem S, Akhtar S, Ali TA, Amjad S, Chawla S, Bagga P, Frenneaux MP, Reddy R, Fakhro K, Haris M. Genetics of glutamate and its receptors in autism spectrum disorder. Mol Psychiatry 2022; 27:2380-2392. [PMID: 35296811 PMCID: PMC9135628 DOI: 10.1038/s41380-022-01506-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 02/11/2022] [Accepted: 02/22/2022] [Indexed: 12/11/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental impairment characterized by deficits in social interaction skills, impaired communication, and repetitive and restricted behaviors that are thought to be due to altered neurotransmission processes. The amino acid glutamate is an essential excitatory neurotransmitter in the human brain that regulates cognitive functions such as learning and memory, which are usually impaired in ASD. Over the last several years, increasing evidence from genetics, neuroimaging, protein expression, and animal model studies supporting the notion of altered glutamate metabolism has heightened the interest in evaluating glutamatergic dysfunction in ASD. Numerous pharmacological, behavioral, and imaging studies have demonstrated the imbalance in excitatory and inhibitory neurotransmitters, thus revealing the involvement of the glutamatergic system in ASD pathology. Here, we review the effects of genetic alterations on glutamate and its receptors in ASD and the role of non-invasive imaging modalities in detecting these changes. We also highlight the potential therapeutic targets associated with impaired glutamatergic pathways.
Collapse
Affiliation(s)
- Sabah Nisar
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Ajaz A Bhat
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Tariq Masoodi
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Sheema Hashem
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Sabah Akhtar
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Tayyiba Akbar Ali
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Sara Amjad
- Shibli National College, Azamgarh, Uttar Pradesh, 276001, India
| | - Sanjeev Chawla
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Puneet Bagga
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Michael P Frenneaux
- Academic Health System, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Ravinder Reddy
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Khalid Fakhro
- Department of Human Genetics, Sidra Medicine, P.O. Box 26999, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medical College, P.O. Box 24144, Doha, Qatar
| | - Mohammad Haris
- Laboratory of Molecular and Metabolic Imaging, Sidra Medicine, P.O. Box 26999, Doha, Qatar.
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Laboratory of Animal Research, Qatar University, P.O. Box 2713, Doha, Qatar.
| |
Collapse
|
39
|
Balasco L, Pagani M, Pangrazzi L, Chelini G, Viscido F, Chama AGC, Galbusera A, Provenzano G, Gozzi A, Bozzi Y. Somatosensory cortex hyperconnectivity and impaired whisker-dependent responses in Cntnap2 -/- mice. Neurobiol Dis 2022; 169:105742. [PMID: 35483565 DOI: 10.1016/j.nbd.2022.105742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/16/2022] [Accepted: 04/21/2022] [Indexed: 11/16/2022] Open
Abstract
Sensory abnormalities are a common feature in autism spectrum disorders (ASDs). Tactile responsiveness is altered in autistic individuals, with hypo-responsiveness being associated with the severity of ASD core symptoms. Similarly, sensory abnormalities have been described in mice lacking ASD-associated genes. Loss-of-function mutations in CNTNAP2 result in cortical dysplasia-focal epilepsy syndrome (CDFE) and autism. Likewise, Cntnap2-/- mice show epilepsy and deficits relevant with core symptoms of human ASDs, and are considered a reliable model to study ASDs. Altered synaptic transmission and synchronicity found in the cerebral cortex of Cntnap2-/- mice would suggest a network dysfunction. Here, we investigated the neural substrates of whisker-dependent responses in Cntnap2+/+ and Cntnap2-/- adult mice. When compared to controls, Cntnap2-/- mice showed focal hyper-connectivity within the primary somatosensory cortex (S1), in the absence of altered connectivity between S1 and other somatosensory areas. This data suggests the presence of impaired somatosensory processing in these mutants. Accordingly, Cntnap2-/- mice displayed impaired whisker-dependent discrimination in the textured novel object recognition test (tNORT) and increased c-fos mRNA induction within S1 following whisker stimulation. S1 functional hyperconnectivity might underlie the aberrant whisker-dependent responses observed in Cntnap2-/- mice, indicating that Cntnap2 mice are a reliable model to investigate sensory abnormalities that characterize ASDs.
Collapse
Affiliation(s)
- Luigi Balasco
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy
| | - Marco Pagani
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy
| | - Luca Pangrazzi
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy
| | - Gabriele Chelini
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy
| | - Francesca Viscido
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy
| | | | - Alberto Galbusera
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy
| | - Giovanni Provenzano
- Department of Cellular, Computational, and Integrative Biology (CIBIO), University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Alessandro Gozzi
- Functional Neuroimaging Laboratory, Center for Neuroscience and Cognitive Systems, Istituto Italiano di Tecnologia, Corso Bettini 31, 38068 Rovereto, Italy
| | - Yuri Bozzi
- Center for Mind/Brain Sciences - CIMeC, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, TN, Italy; CNR Neuroscience Institute, via Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
40
|
A Pilot Study of Whole-Blood Transcriptomic Analysis to Identify Genes Associated with Repetitive Low-Level Blast Exposure in Career Breachers. Biomedicines 2022; 10:biomedicines10030690. [PMID: 35327492 PMCID: PMC8945695 DOI: 10.3390/biomedicines10030690] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023] Open
Abstract
Repetitive low-level blast exposure is one of the major occupational health concerns among US military service members and law enforcement. This study seeks to identify gene expression using microRNA and RNA sequencing in whole-blood samples from experienced breachers and unexposed controls. We performed experimental RNA sequencing using Illumina’s HiSeq 2500 Sequencing System, and microRNA analysis using NanoString Technology nCounter miRNA expression panel in whole-blood total RNA samples from 15 experienced breachers and 14 age-, sex-, and race-matched unexposed controls. We identified 10 significantly dysregulated genes between experienced breachers and unexposed controls, with FDR corrected <0.05: One upregulated gene, LINC00996 (long intergenic non-protein coding RNA 996); and nine downregulated genes, IGLV3-16 (immunoglobulin lambda variable 3-16), CD200 (CD200 molecule), LILRB5 (leukocyte immunoglobulin-like receptor B5), ZNF667-AS1 (ZNF667 antisense RNA 1), LMOD1 (leiomodin 1), CNTNAP2 (contactin-associated protein 2), EVPL (envoplakin), DPF3 (double PHD fingers 3), and IGHV4-34 (immunoglobulin heavy variable 4-34). The dysregulated gene expressions reported here have been associated with chronic inflammation and immune response, suggesting that these pathways may relate to the risk of lasting neurological symptoms following high exposures to blast over a career.
Collapse
|
41
|
Hassan WM, Al-Dbass A, Al-Ayadhi L, Bhat RS, El-Ansary A. Discriminant analysis and binary logistic regression enable more accurate prediction of autism spectrum disorder than principal component analysis. Sci Rep 2022; 12:3764. [PMID: 35260688 PMCID: PMC8904630 DOI: 10.1038/s41598-022-07829-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 01/31/2022] [Indexed: 12/04/2022] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impaired social interaction and restricted, repetitive behavior. Multiple studies have suggested mitochondrial dysfunction, glutamate excitotoxicity, and impaired detoxification mechanism as accepted etiological mechanisms of ASD that can be targeted for therapeutic intervention. In the current study, blood samples were collected from 40 people with autism and 40 control participants after informed consent and full approval from the Institutional Review Board of King Saud University. Sodium (Na+), Potassium (K+), lactate dehydrogenase (LDH), glutathione-s-transferase (GST), and mitochondrial respiratory chain complex I (MRC1) were measured in plasma of both groups. Predictive models were established to discriminate individuals with ASD from controls. The predictive power of these five variables, individually and in combination, was compared using the area under a ROC curve (AUC). We compared the performance of principal component analysis (PCA), discriminant analysis (DA), and binary logistic regression (BLR) as ways to combine single variables and create the predictive models. K+ had the highest AUC (0.801) of any single variable, followed by GST, LDH, Na+, and MRC1, respectively. Combining the five variables resulted in higher AUCs than those obtained using single variables across all models. Both DA and BLR were superior to PCA and comparable to each other. In our study, the combination of Na+, K+, LDH, GST, and MRC1 showed the highest promise in discriminating individuals with autism from controls. These results provide a platform that can potentially be used to verify the efficacy of our models with a larger sample size or evaluate other biomarkers.
Collapse
Affiliation(s)
- Wail M Hassan
- Department of Biomedical Sciences, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Abeer Al-Dbass
- Biochemistry Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Laila Al-Ayadhi
- Department of Physiology, Faculty of Medicine, King Saud University, Riyadh, Saudi Arabia.,Autism Research and Treatment Center, Riyadh, Saudi Arabia
| | - Ramesa Shafi Bhat
- Biochemistry Department, College of Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Autism Research and Treatment Center, Riyadh, Saudi Arabia. .,Central Research Laboratory, Female Centre for Scientific and Medical Studies, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
42
|
Seery N, Butzkueven H, O'Brien TJ, Monif M. Contemporary advances in antibody-mediated encephalitis: anti-LGI1 and anti-Caspr2 antibody (Ab)-mediated encephalitides. Autoimmun Rev 2022; 21:103074. [PMID: 35247644 DOI: 10.1016/j.autrev.2022.103074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 02/27/2022] [Indexed: 01/17/2023]
Abstract
Encephalitides with antibodies directed against leucine-rich glioma-inactivated 1 (LGI1) and contactin-associated protein-like 2 (Caspr2) represent two increasingly well characterised forms of autoimmune encephalitis. Both share overlapping and distinct clinical features, are mediated by autoantibodies directed against differing proteins complexed with voltage-gated potassium channels, with unique genetic predisposition identified to date. Herein we summarise disease mechanisms, clinical features, treatment considerations, prognostic factors and clinical outcomes regarding these disorders.
Collapse
Affiliation(s)
- Nabil Seery
- Department of Neuroscience, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Helmut Butzkueven
- Department of Neuroscience, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Mastura Monif
- Department of Neuroscience, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria, Australia; Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia; Department of Neurology, Royal Melbourne Hospital, Melbourne, Victoria, Australia.
| |
Collapse
|
43
|
Choe KY, Bethlehem RAI, Safrin M, Dong H, Salman E, Li Y, Grinevich V, Golshani P, DeNardo LA, Peñagarikano O, Harris NG, Geschwind DH. Oxytocin normalizes altered circuit connectivity for social rescue of the Cntnap2 knockout mouse. Neuron 2022; 110:795-808.e6. [PMID: 34932941 PMCID: PMC8944915 DOI: 10.1016/j.neuron.2021.11.031] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 09/03/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022]
Abstract
The neural basis of abnormal social behavior in autism spectrum disorders (ASDs) remains incompletely understood. Here we used two complementary but independent brain-wide mapping approaches, mouse resting-state fMRI and c-Fos-iDISCO+ imaging, to construct brain-wide activity and connectivity maps of the Cntnap2 knockout (KO) mouse model of ASD. At the macroscale level, we detected reduced functional coupling across social brain regions despite general patterns of hyperconnectivity across major brain structures. Oxytocin administration, which rescues social deficits in KO mice, strongly stimulated many brain areas and normalized connectivity patterns. Notably, chemogenetically triggered release of endogenous oxytocin strongly stimulated the nucleus accumbens (NAc), a forebrain nucleus implicated in social reward. Furthermore, NAc-targeted approaches to activate local oxytocin receptors sufficiently rescued their social deficits. Our findings establish circuit- and systems-level mechanisms of social deficits in Cntnap2 KO mice and reveal the NAc as a region that can be modulated by oxytocin to promote social interactions.
Collapse
Affiliation(s)
- Katrina Y Choe
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Psychology, Neuroscience & Behaviour, McMaster University, Hamilton, ON L8S 4K1, Canada.
| | - Richard A I Bethlehem
- Autism Research Centre, Department of Psychiatry, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Martin Safrin
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Hongmei Dong
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Elena Salman
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Ying Li
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Valery Grinevich
- Department of Neuropeptide Research for Psychiatry, Central Institute of Mental Health, University of Heidelberg, Mannheim 68159, Germany
| | - Peyman Golshani
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Laura A DeNardo
- Department of Physiology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Olga Peñagarikano
- Department of Pharmacology, School of Medicine, University of the Basque Country (UPV/EHU), Vizcaya 48940, Spain
| | - Neil G Harris
- Department of Neurosurgery, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Daniel H Geschwind
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
44
|
Martín-de-Saavedra MD, Dos Santos M, Culotta L, Varea O, Spielman BP, Parnell E, Forrest MP, Gao R, Yoon S, McCoig E, Jalloul HA, Myczek K, Khalatyan N, Hall EA, Turk LS, Sanz-Clemente A, Comoletti D, Lichtenthaler SF, Burgdorf JS, Barbolina MV, Savas JN, Penzes P. Shed CNTNAP2 ectodomain is detectable in CSF and regulates Ca 2+ homeostasis and network synchrony via PMCA2/ATP2B2. Neuron 2022; 110:627-643.e9. [PMID: 34921780 PMCID: PMC8857041 DOI: 10.1016/j.neuron.2021.11.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 10/11/2021] [Accepted: 11/19/2021] [Indexed: 11/29/2022]
Abstract
Although many neuronal membrane proteins undergo proteolytic cleavage, little is known about the biological significance of neuronal ectodomain shedding (ES). Here, we show that the neuronal sheddome is detectable in human cerebrospinal fluid (hCSF) and is enriched in neurodevelopmental disorder (NDD) risk factors. Among shed synaptic proteins is the ectodomain of CNTNAP2 (CNTNAP2-ecto), a prominent NDD risk factor. CNTNAP2 undergoes activity-dependent ES via MMP9 (matrix metalloprotease 9), and CNTNAP2-ecto levels are reduced in the hCSF of individuals with autism spectrum disorder. Using mass spectrometry, we identified the plasma membrane Ca2+ ATPase (PMCA) extrusion pumps as novel CNTNAP2-ecto binding partners. CNTNAP2-ecto enhances the activity of PMCA2 and regulates neuronal network dynamics in a PMCA2-dependent manner. Our data underscore the promise of sheddome analysis in discovering neurobiological mechanisms, provide insight into the biology of ES and its relationship with the CSF, and reveal a mechanism of regulation of Ca2+ homeostasis and neuronal network synchrony by a shed ectodomain.
Collapse
Affiliation(s)
| | - Marc Dos Santos
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Lorenza Culotta
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Olga Varea
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Benjamin P Spielman
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Euan Parnell
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Marc P Forrest
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ruoqi Gao
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Sehyoun Yoon
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Emmarose McCoig
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Hiba A Jalloul
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kristoffer Myczek
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Natalia Khalatyan
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Elizabeth A Hall
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Liam S Turk
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA; Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Antonio Sanz-Clemente
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Davide Comoletti
- Child Health Institute of New Jersey, New Brunswick, NJ 08901, USA; Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, NJ 08901, USA; Department of Pediatrics, Robert Wood Johnson Medical School, Rutgers, the State University of New Jersey, New Brunswick, NJ 08901, USA; School of Biological Sciences, Victoria University of Wellington, Wellington 6140, New Zealand
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Department of Neuroproteomics, School of Medicine, Klinikum rechts der Isar, and Institute for Advanced Study, Technical University of Munich, 81675 Munich, Germany; Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Jeffrey S Burgdorf
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL 60611, USA; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Maria V Barbolina
- Department of Biopharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Jeffrey N Savas
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Peter Penzes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Autism and Neurodevelopment, Northwestern University, Chicago, IL 60611, USA; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
45
|
Papale LA, Madrid A, Zhang Q, Chen K, Sak L, Keleş S, Alisch RS. Gene by environment interaction mouse model reveals a functional role for 5-hydroxymethylcytosine in neurodevelopmental disorders. Genome Res 2022; 32:266-279. [PMID: 34949667 PMCID: PMC8805724 DOI: 10.1101/gr.276137.121] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 12/22/2021] [Indexed: 11/25/2022]
Abstract
Mouse knockouts of Cntnap2 show altered neurodevelopmental behavior, deficits in striatal GABAergic signaling, and a genome-wide disruption of an environmentally sensitive DNA methylation modification (5-hydroxymethylcytosine [5hmC]) in the orthologs of a significant number of genes implicated in human neurodevelopmental disorders. We tested adult Cntnap2 heterozygous mice (Cntnap2 +/-; lacking behavioral or neuropathological abnormalities) subjected to a prenatal stress and found that prenatally stressed Cntnap2 +/- female mice show repetitive behaviors and altered sociability, similar to the homozygote phenotype. Genomic profiling revealed disruptions in hippocampal and striatal 5hmC levels that are correlated to altered transcript levels of genes linked to these phenotypes (e.g., Reln, Dst, Trio, and Epha5). Chromatin immunoprecipitation coupled with high-throughput sequencing and hippocampal nuclear lysate pull-down data indicated that 5hmC abundance alters the binding of the transcription factor CLOCK near the promoters of these genes (e.g., Palld, Gigyf1, and Fry), providing a mechanistic role for 5hmC in gene regulation. Together, these data support gene-by-environment hypotheses for the origins of mental illness and provide a means to identify the elusive factors contributing to complex human diseases.
Collapse
Affiliation(s)
- Ligia A Papale
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin 53719, USA
| | - Andy Madrid
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin 53719, USA
- Neuroscience Training Program, University of Wisconsin, Madison, Wisconsin 53719, USA
| | - Qi Zhang
- Department Mathematics and Statistics, University of New Hampshire, Durham, New Hampshire 03824, USA
| | - Kailei Chen
- Department of Statistics, Biostatistics, and Medical Informatics, University of Wisconsin, Madison, Wisconsin 53719, USA
| | - Lara Sak
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin 53719, USA
| | - Sündüz Keleş
- Department of Statistics, Biostatistics, and Medical Informatics, University of Wisconsin, Madison, Wisconsin 53719, USA
| | - Reid S Alisch
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin 53719, USA
| |
Collapse
|
46
|
Sheppard K, Gardin J, Sabnis GS, Peer A, Darrell M, Deats S, Geuther B, Lutz CM, Kumar V. Stride-level analysis of mouse open field behavior using deep-learning-based pose estimation. Cell Rep 2022; 38:110231. [PMID: 35021077 PMCID: PMC8796662 DOI: 10.1016/j.celrep.2021.110231] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 04/29/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022] Open
Abstract
Gait and posture are often perturbed in many neurological, neuromuscular, and neuropsychiatric conditions. Rodents provide a tractable model for elucidating disease mechanisms and interventions. Here, we develop a neural-network-based assay that adopts the commonly used open field apparatus for mouse gait and posture analysis. We quantitate both with high precision across 62 strains of mice. We characterize four mutants with known gait deficits and demonstrate that multiple autism spectrum disorder (ASD) models show gait and posture deficits, implying this is a general feature of ASD. Mouse gait and posture measures are highly heritable and fall into three distinct classes. We conduct a genome-wide association study to define the genetic architecture of stride-level mouse movement in the open field. We provide a method for gait and posture extraction from the open field and one of the largest laboratory mouse gait and posture data resources for the research community. Sheppard et al. present a method for gait and posture analysis in the common open field apparatus using neural-network-based pose estimation. They apply this high-throughput method to dissect the genetic architecture of mouse movement.
Collapse
Affiliation(s)
- Keith Sheppard
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Justin Gardin
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Gautam S Sabnis
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Asaf Peer
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Megan Darrell
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Sean Deats
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Brian Geuther
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Cathleen M Lutz
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Vivek Kumar
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA.
| |
Collapse
|
47
|
St George-Hyslop F, Kivisild T, Livesey FJ. The role of contactin-associated protein-like 2 in neurodevelopmental disease and human cerebral cortex evolution. Front Mol Neurosci 2022; 15:1017144. [PMID: 36340692 PMCID: PMC9630569 DOI: 10.3389/fnmol.2022.1017144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/20/2022] [Indexed: 12/04/2022] Open
Abstract
The contactin-associated protein-like 2 (CNTNAP2) gene is associated with multiple neurodevelopmental disorders, including autism spectrum disorder (ASD), intellectual disability (ID), and specific language impairment (SLI). Experimental work has shown that CNTNAP2 is important for neuronal development and synapse formation. There is also accumulating evidence for the differential use of CNTNAP2 in the human cerebral cortex compared with other primates. Here, we review the current literature on CNTNAP2, including what is known about its expression, disease associations, and molecular/cellular functions. We also review the evidence for its role in human brain evolution, such as the presence of eight human accelerated regions (HARs) within the introns of the gene. While progress has been made in understanding the function(s) of CNTNAP2, more work is needed to clarify the precise mechanisms through which CNTNAP2 acts. Such information will be crucial for developing effective treatments for CNTNAP2 patients. It may also shed light on the longstanding question of what makes us human.
Collapse
Affiliation(s)
- Frances St George-Hyslop
- Zayed Centre for Research Into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Toomas Kivisild
- Estonian Biocentre, Institute of Genomics, University of Tartu, Tartu, Estonia.,Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Frederick J Livesey
- Zayed Centre for Research Into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
48
|
Panganiban CH, Barth JL, Tan J, Noble KV, McClaskey CM, Howard BA, Jafri SH, Dias JW, Harris KC, Lang H. Two distinct types of nodes of Ranvier support auditory nerve function in the mouse cochlea. Glia 2021; 70:768-791. [DOI: 10.1002/glia.24138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 11/12/2021] [Accepted: 12/17/2021] [Indexed: 11/09/2022]
Affiliation(s)
- Clarisse H. Panganiban
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
- Wolfson Centre for Age‐Related Diseases King's College London London UK
| | - Jeremy L. Barth
- Department of Regenerative Medicine and Cell Biology Medical University of South Carolina Charleston South Carolina USA
| | - Junying Tan
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| | - Kenyaria V. Noble
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| | - Carolyn M. McClaskey
- Department of Otolaryngology & Head and Neck Surgery Medical University of South Carolina Charleston South Carolina USA
| | - Blake A. Howard
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| | - Shabih H. Jafri
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| | - James W. Dias
- Department of Otolaryngology & Head and Neck Surgery Medical University of South Carolina Charleston South Carolina USA
| | - Kelly C. Harris
- Department of Otolaryngology & Head and Neck Surgery Medical University of South Carolina Charleston South Carolina USA
| | - Hainan Lang
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| |
Collapse
|
49
|
Riemersma IW, Havekes R, Kas MJH. Spatial and Temporal Gene Function Studies in Rodents: Towards Gene-Based Therapies for Autism Spectrum Disorder. Genes (Basel) 2021; 13:28. [PMID: 35052369 PMCID: PMC8774890 DOI: 10.3390/genes13010028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/06/2021] [Accepted: 12/20/2021] [Indexed: 12/26/2022] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition that is characterized by differences in social interaction, repetitive behaviors, restricted interests, and sensory differences beginning early in life. Especially sensory symptoms are highly correlated with the severity of other behavioral differences. ASD is a highly heterogeneous condition on multiple levels, including clinical presentation, genetics, and developmental trajectories. Over a thousand genes have been implicated in ASD. This has facilitated the generation of more than two hundred genetic mouse models that are contributing to understanding the biological underpinnings of ASD. Since the first symptoms already arise during early life, it is especially important to identify both spatial and temporal gene functions in relation to the ASD phenotype. To further decompose the heterogeneity, ASD-related genes can be divided into different subgroups based on common functions, such as genes involved in synaptic function. Furthermore, finding common biological processes that are modulated by this subgroup of genes is essential for possible patient stratification and the development of personalized early treatments. Here, we review the current knowledge on behavioral rodent models of synaptic dysfunction by focusing on behavioral phenotypes, spatial and temporal gene function, and molecular targets that could lead to new targeted gene-based therapy.
Collapse
Affiliation(s)
| | | | - Martien J. H. Kas
- Groningen Institute for Evolutionary Life Sciences, Neurobiology, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands; (I.W.R.); (R.H.)
| |
Collapse
|
50
|
Otazu GH, Li Y, Lodato Z, Elnasher A, Keever KM, Li Y, Ramos RL. Neurodevelopmental malformations of the cerebellum and neocortex in the Shank3 and Cntnap2 mouse models of autism. Neurosci Lett 2021; 765:136257. [PMID: 34555490 DOI: 10.1016/j.neulet.2021.136257] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/28/2022]
Abstract
There are many mouse models of autism with broad use in neuroscience research. Genetic background can be a major contributor to the phenotype observed in any mouse model of disease, including genetic models of autism. C57BL/6 mice display spontaneous glio-neuronal heterotopia in the cerebellar vermis and neocortex which may also exist in mouse models of autism created on this background. In the present report, we document the presence of cerebellar and neocortical heterotopia in heterozygous and KO Shank3 and Cntnap2 mice which are due to the C57BL/6 genotype and discuss the role these malformations may play in research using these genetic models of autism.
Collapse
Affiliation(s)
- Gonzalo H Otazu
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, United States
| | - Yan Li
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, United States
| | - Zachary Lodato
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, United States
| | - Adel Elnasher
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, United States
| | - Katherine M Keever
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, United States
| | - Ying Li
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, United States
| | - Raddy L Ramos
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY 11568, United States.
| |
Collapse
|