1
|
Szukiewicz D. Histaminergic System Activity in the Central Nervous System: The Role in Neurodevelopmental and Neurodegenerative Disorders. Int J Mol Sci 2024; 25:9859. [PMID: 39337347 PMCID: PMC11432521 DOI: 10.3390/ijms25189859] [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: 08/15/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Histamine (HA), a biogenic monoamine, exerts its pleiotropic effects through four H1R-H4R histamine receptors, which are also expressed in brain tissue. Together with the projections of HA-producing neurons located within the tuberomammillary nucleus (TMN), which innervate most areas of the brain, they constitute the histaminergic system. Thus, while remaining a mediator of the inflammatory reaction and immune system function, HA also acts as a neurotransmitter and a modulator of other neurotransmitter systems in the central nervous system (CNS). Although the detailed causes are still not fully understood, neuroinflammation seems to play a crucial role in the etiopathogenesis of both neurodevelopmental and neurodegenerative (neuropsychiatric) diseases, such as autism spectrum disorders (ASDs), attention-deficit/hyperactivity disorder (ADHD), Alzheimer's disease (AD) and Parkinson's disease (PD). Given the increasing prevalence/diagnosis of these disorders and their socioeconomic impact, the need to develop effective forms of therapy has focused researchers' attention on the brain's histaminergic activity and other related signaling pathways. This review presents the current state of knowledge concerning the involvement of HA and the histaminergic system within the CNS in the development of neurodevelopmental and neurodegenerative disorders. To this end, the roles of HA in neurotransmission, neuroinflammation, and neurodevelopment are also discussed.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
2
|
Wasnik K, Gupta PS, Mukherjee S, Oviya A, Prakash R, Pareek D, Patra S, Maity S, Rai V, Singh M, Singh G, Yadav DD, Das S, Maiti P, Paik P. Poly( N-acryloylglycine-acrylamide) Hydrogel Mimics the Cellular Microenvironment and Promotes Neurite Growth with Protection from Oxidative Stress. ACS APPLIED BIO MATERIALS 2023; 6:5644-5661. [PMID: 37993284 DOI: 10.1021/acsabm.3c00807] [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] [Indexed: 11/24/2023]
Abstract
In this work, the glycine-based acryloyl monomer is polymerized to obtain a neurogenic polymeric hydrogel for regenerative applications. The synthesized poly(N-acryloylglycine-acrylamide) [poly(NAG-b-A)] nanohydrogel exhibits high swelling (∼1500%) and is mechanically very stable, biocompatible, and proliferative in nature. The poly(NAG-b-A) nanohydrogel provides a stable 3D extracellular mimetic environment and promotes healthy neurite growth for primary cortical neurons by facilitating cellular adhesion, proliferation, actin filament stabilization, and neuronal differentiation. Furthermore, the protective role of the poly(NAG-b-A) hydrogel for the neurons in oxidative stress conditions is revealed and it is found that it is a clinically relevant material for neuronal regenerative applications, such as for promoting nerve regeneration via GSK3β inhibition. This hydrogel additionally plays an important role in modulating the biological microenvironment, either as an agonist and antagonist or as an antioxidant. Furthermore, it favors the physiological responses and eases the neurite growth efficiency. Additionally, we found out that the conversion of glycine-based acryloyl monomers into their corresponding polymer modulates the mechanical performance, mimics the cellular microenvironment, and accelerates the self-healing capability due to the responsive behavior towards reactive oxygen species (ROS). Thus, the p(NAG-b-A) hydrogel could be a potential candidate to induce neuronal regeneration since it provides a physical cue and significantly boosts neurite outgrowth and also maintains the microtubule integrity in neuronal cells.
Collapse
Affiliation(s)
- Kirti Wasnik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Prem Shankar Gupta
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Sudip Mukherjee
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Alagu Oviya
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Ravi Prakash
- School of Material Science, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Divya Pareek
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Sukanya Patra
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Somedutta Maity
- School of Engineering Sciences and Technology, University of Hyderabad, Hyderabad, Telangana State 500 046, India
| | - Vipin Rai
- Department of Biochemistry, Institute of Sciences, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Monika Singh
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Gurmeet Singh
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Desh Deepak Yadav
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Santanu Das
- Department of Ceramic Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Pralay Maiti
- School of Material Science, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Pradip Paik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| |
Collapse
|
3
|
Salceda R. Glycine neurotransmission: Its role in development. Front Neurosci 2022; 16:947563. [PMID: 36188468 PMCID: PMC9525178 DOI: 10.3389/fnins.2022.947563] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
The accurate function of the central nervous system (CNS) depends of the consonance of multiple genetic programs and external signals during the ontogenesis. A variety of molecules including neurotransmitters, have been implied in the regulation of proliferation, survival, and cell-fate of neurons and glial cells. Among these, neurotransmitters may play a central role since functional ligand-gated ionic channel receptors have been described before the establishment of synapses. This review argues on the function of glycine during development, and show evidence indicating it regulates morphogenetic events by means of their transporters and receptors, emphasizing the role of glycinergic activity in the balance of excitatory and inhibitory signals during development. Understanding the mechanisms involved in these processes would help us to know the etiology of cognitive dysfunctions and lead to improve brain repair strategies.
Collapse
|
4
|
Benevento M, Hökfelt T, Harkany T. Ontogenetic rules for the molecular diversification of hypothalamic neurons. Nat Rev Neurosci 2022; 23:611-627. [PMID: 35906427 DOI: 10.1038/s41583-022-00615-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2022] [Indexed: 11/09/2022]
Abstract
The hypothalamus is an evolutionarily conserved endocrine interface that, among other roles, links central homeostatic control to adaptive bodily responses by releasing hormones and neuropeptides from its many neuronal subtypes. In its preoptic, anterior, tuberal and mammillary subdivisions, a kaleidoscope of magnocellular and parvocellular neuroendocrine command neurons, local-circuit neurons, and neurons that project to extrahypothalamic areas are intermingled in partially overlapping patches of nuclei. Molecular fingerprinting has produced data of unprecedented mass and depth to distinguish and even to predict the synaptic and endocrine competences, connectivity and stimulus selectivity of many neuronal modalities. These new insights support eminent studies from the past century but challenge others on the molecular rules that shape the developmental segregation of hypothalamic neuronal subtypes and their use of morphogenic cues for terminal differentiation. Here, we integrate single-cell RNA sequencing studies with those of mouse genetics and endocrinology to describe key stages of hypothalamus development, including local neurogenesis, the direct terminal differentiation of glutamatergic neurons, transition cascades for GABAergic and GABAergic cell-derived dopamine cells, waves of local neuronal migration, and sequential enrichment in neuropeptides and hormones. We particularly emphasize how transcription factors determine neuronal identity and, consequently, circuit architecture, and whether their deviations triggered by environmental factors and hormones provoke neuroendocrine illnesses.
Collapse
Affiliation(s)
- Marco Benevento
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Tomas Hökfelt
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria. .,Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
5
|
Abstract
During evolution, the cerebral cortex advances by increasing in surface and the introduction of new cytoarchitectonic areas among which the prefrontal cortex (PFC) is considered to be the substrate of highest cognitive functions. Although neurons of the PFC are generated before birth, the differentiation of its neurons and development of synaptic connections in humans extend to the 3rd decade of life. During this period, synapses as well as neurotransmitter systems including their receptors and transporters, are initially overproduced followed by selective elimination. Advanced methods applied to human and animal models, enable investigation of the cellular mechanisms and role of specific genes, non-coding regulatory elements and signaling molecules in control of prefrontal neuronal production and phenotypic fate, as well as neuronal migration to establish layering of the PFC. Likewise, various genetic approaches in combination with functional assays and immunohistochemical and imaging methods reveal roles of neurotransmitter systems during maturation of the PFC. Disruption, or even a slight slowing of the rate of neuronal production, migration and synaptogenesis by genetic or environmental factors, can induce gross as well as subtle changes that eventually can lead to cognitive impairment. An understanding of the development and evolution of the PFC provide insight into the pathogenesis and treatment of congenital neuropsychiatric diseases as well as idiopathic developmental disorders that cause intellectual disabilities.
Collapse
Affiliation(s)
- Sharon M Kolk
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, The Netherlands.
| | - Pasko Rakic
- Department of Neuroscience and Kavli Institute for Neuroscience, Yale University, New Haven, Connecticut, USA.
| |
Collapse
|
6
|
Keller M, Brennenstuhl H, Kuseyri Hübschmann O, Manti F, Julia Palacios NA, Friedman J, Yıldız Y, Koht JA, Wong SN, Zafeiriou DI, López-Laso E, Pons R, Kulhánek J, Jeltsch K, Serrano-Lomelin J, Garbade SF, Opladen T, Goez H, Burlina A, Cortès-Saladelafont E, Fernández Ramos JA, García-Cazorla A, Hoffmann GF, Kiat Hong ST, Honzík T, Kavecan I, Kurian MA, Leuzzi V, Lücke T, Manzoni F, Mastrangelo M, Mercimek-Andrews S, Mir P, Oppebøen M, Pearson TS, Sivri HS, Steel D, Stevanović G, Fung CW. Assessment of intellectual impairment, health-related quality of life, and behavioral phenotype in patients with neurotransmitter related disorders: Data from the iNTD registry. J Inherit Metab Dis 2021; 44:1489-1502. [PMID: 34245036 DOI: 10.1002/jimd.12416] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/30/2021] [Accepted: 07/07/2021] [Indexed: 12/30/2022]
Abstract
Inherited disorders of neurotransmitter metabolism are a group of rare diseases, which are caused by impaired synthesis, transport, or degradation of neurotransmitters or cofactors and result in various degrees of delayed or impaired psychomotor development. To assess the effect of neurotransmitter deficiencies on intelligence, quality of life, and behavior, the data of 148 patients in the registry of the International Working Group on Neurotransmitter Related Disorders (iNTD) was evaluated using results from standardized age-adjusted tests and questionnaires. Patients with a primary disorder of monoamine metabolism had lower IQ scores (mean IQ 58, range 40-100) within the range of cognitive impairment (<70) compared to patients with a BH4 deficiency (mean IQ 84, range 40-129). Short attention span and distractibility were most frequently mentioned by parents, while patients reported most frequently anxiety and distractibility when asked for behavioral traits. In individuals with succinic semialdehyde dehydrogenase deficiency, self-stimulatory behaviors were commonly reported by parents, whereas in patients with dopamine transporter deficiency, DNAJC12 deficiency, and monoamine oxidase A deficiency, self-injurious or mutilating behaviors have commonly been observed. Phobic fears were increased in patients with 6-pyruvoyltetrahydropterin synthase deficiency, while individuals with sepiapterin reductase deficiency frequently experienced communication and sleep difficulties. Patients with BH4 deficiencies achieved significantly higher quality of life as compared to other groups. This analysis of the iNTD registry data highlights: (a) difference in IQ and subdomains of quality of life between BH4 deficiencies and primary neurotransmitter-related disorders and (b) previously underreported behavioral traits.
Collapse
Affiliation(s)
- Mareike Keller
- Division of Child Neurology and Metabolic Medicine, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Heiko Brennenstuhl
- Division of Child Neurology and Metabolic Medicine, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Oya Kuseyri Hübschmann
- Division of Child Neurology and Metabolic Medicine, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Filippo Manti
- Department of Human Neuroscience, Unit of Child Neurology and Psychiatry, Università degli Studi di Roma La Sapienza, Rome, Italy
| | - Natalia Alexandra Julia Palacios
- Inborn errors of metabolism Unit, Department of Neurology, Institut de Recerca Sant Joan de Déu and CIBERER-ISCIII, Barcelona, Spain
| | - Jennifer Friedman
- UCSD Departments of Neuroscience and Pediatrics; Rady Children's Hospital Division of Neurology, Rady Children's Institute for Genomic Medicine, San Diego, California, USA
| | - Yılmaz Yıldız
- Hacettepe University, Faculty of Medicine, Department of Pediatrics, Section of Pediatric Metabolism, Ankara, Turkey
| | | | - Suet-Na Wong
- Department of Pediatrics and Adolescent Medicine, The Hong Kong Children's Hospital, Hong Kong, Hong Kong
| | - Dimitrios I Zafeiriou
- First Department of Pediatrics Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eduardo López-Laso
- Pediatric Neurology Unit, Department of Pediatrics, University Hospital Reina Sofía, IMIBIC and CIBERER, Córdoba, Spain
| | - Roser Pons
- First Department of Pediatrics of the University of Athens, Aghia Sofia Hospital, Athens, Greece
| | - Jan Kulhánek
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Kathrin Jeltsch
- Division of Child Neurology and Metabolic Medicine, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Jesus Serrano-Lomelin
- Women and Children's Health Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Sven F Garbade
- Division of Child Neurology and Metabolic Medicine, University Children's Hospital Heidelberg, Heidelberg, Germany
- Dietmar-Hopp Metabolic Center, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Opladen
- Division of Child Neurology and Metabolic Medicine, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Helly Goez
- Department of Pediatrics, University of Alberta, Glenrose Rehabilitation Hospital, Edmonton, Alberta, Canada
| | - Alberto Burlina
- U.O.C. Malattie Metaboliche Ereditarie, Dipartimento della Salute della Donna e del Bambino, Azienda Ospedaliera Universitaria di Padova - Campus Biomedico Pietro d'Abano, Padova, Italy
| | - Elisenda Cortès-Saladelafont
- Inborn errors of metabolism Unit, Department of Neurology, Institut de Recerca Sant Joan de Déu and CIBERER-ISCIII, Barcelona, Spain
- Inborn Errors of Metabolism and Child Neurology Unit, Department of Pediatrics, Hospital Germans Trias i Pujol, Badalona and Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | - Angeles García-Cazorla
- Inborn errors of metabolism Unit, Department of Neurology, Institut de Recerca Sant Joan de Déu and CIBERER-ISCIII, Barcelona, Spain
| | - Georg F Hoffmann
- Division of Child Neurology and Metabolic Medicine, University Children's Hospital Heidelberg, Heidelberg, Germany
| | - Stacey Tay Kiat Hong
- KTP-National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Tomáš Honzík
- Department of Pediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czech Republic
| | - Ivana Kavecan
- Faculty of Medicine, University of Novi Sad, Institute for Children and Youth Health Care of Vojvodina, Novi Sad, Serbia
| | - Manju A Kurian
- Developmental Neurosciences, UCL Great Ormond Street-Institute of Child Health and Department of Neurology, Great Ormond Street Hospital, London, UK
| | - Vincenzo Leuzzi
- Department of Human Neuroscience, Unit of Child Neurology and Psychiatry, Università degli Studi di Roma La Sapienza, Rome, Italy
| | - Thomas Lücke
- University Children's Hospital, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Francesca Manzoni
- U.O.C. Malattie Metaboliche Ereditarie, Dipartimento della Salute della Donna e del Bambino, Azienda Ospedaliera Universitaria di Padova - Campus Biomedico Pietro d'Abano, Padova, Italy
| | - Mario Mastrangelo
- Department of Human Neuroscience, Unit of Child Neurology and Psychiatry, Università degli Studi di Roma La Sapienza, Rome, Italy
| | - Saadet Mercimek-Andrews
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, University of Toronto, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Medical Genetics, University of Alberta, Women and Children's Health Research Institute, Stollery Children's Hospital, Edmonton, Alberta, Canada
| | - Pablo Mir
- Unidad de Trastornos del Movimiento Servicio de Neurología y Neurofisiología Clínica Unidad de Gestión Clínica de Neurociencias Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Mari Oppebøen
- Children's Department Division of Child Neurology Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Toni S Pearson
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - H Serap Sivri
- Hacettepe University, Faculty of Medicine, Department of Pediatrics, Section of Pediatric Metabolism, Ankara, Turkey
| | - Dora Steel
- Developmental Neurosciences, UCL Great Ormond Street-Institute of Child Health and Department of Neurology, Great Ormond Street Hospital, London, UK
| | - Galina Stevanović
- Clinic of Neurology and Psychiatry for Children and Youth, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Cheuk-Wing Fung
- Department of Pediatrics and Adolescent Medicine, The Hong Kong Children's Hospital, Hong Kong, Hong Kong
| |
Collapse
|
7
|
Reyes-Pinto R, Ferrán JL, Vega-Zuniga T, González-Cabrera C, Luksch H, Mpodozis J, Puelles L, Marín GJ. Change in the neurochemical signature and morphological development of the parvocellular isthmic projection to the avian tectum. J Comp Neurol 2021; 530:553-573. [PMID: 34363623 DOI: 10.1002/cne.25229] [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: 05/04/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 11/05/2022]
Abstract
Neurons can change their classical neurotransmitters during ontogeny, sometimes going through stages of dual release. Here, we explored the development of the neurotransmitter identity of neurons of the avian nucleus isthmi parvocellularis (Ipc), whose axon terminals are retinotopically arranged in the optic tectum (TeO) and exert a focal gating effect upon the ascending transmission of retinal inputs. Although cholinergic and glutamatergic markers are both found in Ipc neurons and terminals of adult pigeons and chicks, the mRNA expression of the vesicular acetylcholine transporter, VAChT, is weak or absent. To explore how the Ipc neurotransmitter identity is established during ontogeny, we analyzed the expression of mRNAs coding for cholinergic (ChAT, VAChT, and CHT) and glutamatergic (VGluT2 and VGluT3) markers in chick embryos at different developmental stages. We found that between E12 and E18, Ipc neurons expressed all cholinergic mRNAs and also VGluT2 mRNA; however, from E16 through posthatch stages, VAChT mRNA expression was specifically diminished. Our ex vivo deposits of tracer crystals and intracellular filling experiments revealed that Ipc axons exhibit a mature paintbrush morphology late in development, experiencing marked morphological transformations during the period of presumptive dual vesicular transmitter release. Additionally, although ChAT protein immunoassays increasingly label the growing Ipc axon, this labeling was consistently restricted to sparse portions of the terminal branches. Combined, these results suggest that the synthesis of glutamate and acetylcholine, and their vesicular release, is complexly linked to the developmental processes of branching, growing and remodeling of these unique axons.
Collapse
Affiliation(s)
- Rosana Reyes-Pinto
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - José L Ferrán
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, University of Murcia, Murcia, Spain
| | - Tomas Vega-Zuniga
- Lehrstuhl für Zoologie, Technical University of Munich, Freising, Germany.,Institute of Science and Technology Austria (IST Austria), Klosterneuburg, Austria
| | | | - Harald Luksch
- Lehrstuhl für Zoologie, Technical University of Munich, Freising, Germany
| | - Jorge Mpodozis
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, University of Murcia, Murcia, Spain
| | - Gonzalo J Marín
- Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.,Facultad de Medicina, Universidad Finis Terrae, Santiago, Chile
| |
Collapse
|
8
|
Wang L, Tang S, Wu S, Yao L, Su D, Wang Y. Maternal Exposure to Pesticides and Risk of Autism Spectrum Disorders in Offspring: A Meta-analysis. J Autism Dev Disord 2021; 52:1640-1651. [PMID: 33978908 DOI: 10.1007/s10803-021-05063-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2021] [Indexed: 12/16/2022]
Abstract
This meta-analysis was conducted to estimate the overall association between maternal exposure to pesticides and risk of ASD in offspring. PubMed, EMBASE, Web of Science, and the PsycINFO were searched until December 30, 2020 to include eligible studies. Eight studies with 50,426 participants, 5810 of whom had ASD, were involved in the study. Overall, the summary OR (95% confidence interval) of ASDs in offspring for maternal exposure to pesticide estimated by residential proximity measures and self-report was 1.88 (1.10-3.20). However, maternal exposure to pesticide measured by biomarkers was not associated with an increased risk of ASDs (pooled OR 1.13; 95% CI 0.83-1.54). Further well-designed studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Li Wang
- Mental Health Center, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuchang District, Wuhan, 430060, People's Republic of China
| | - Shiming Tang
- Mental Health Center, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuchang District, Wuhan, 430060, People's Republic of China
| | - Songjie Wu
- Healthcare-Associated Infection Management Office, Zhongnan Hospital of Wuhan University, Donghu Road 158#, Wuchang District, Wuhan, 430071, People's Republic of China
| | - Lihua Yao
- Mental Health Center, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuchang District, Wuhan, 430060, People's Republic of China
| | - Dezhen Su
- Mental Health Center, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuchang District, Wuhan, 430060, People's Republic of China
| | - Ying Wang
- Mental Health Center, Renmin Hospital of Wuhan University, Jiefang Road 238#, Wuchang District, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
9
|
Ribeiro J, Procyk CA, West EL, O'Hara-Wright M, Martins MF, Khorasani MM, Hare A, Basche M, Fernando M, Goh D, Jumbo N, Rizzi M, Powell K, Tariq M, Michaelides M, Bainbridge JWB, Smith AJ, Pearson RA, Gonzalez-Cordero A, Ali RR. Restoration of visual function in advanced disease after transplantation of purified human pluripotent stem cell-derived cone photoreceptors. Cell Rep 2021; 35:109022. [PMID: 33882303 PMCID: PMC8065177 DOI: 10.1016/j.celrep.2021.109022] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 01/08/2021] [Accepted: 03/31/2021] [Indexed: 12/19/2022] Open
Abstract
Age-related macular degeneration and other macular diseases result in the loss of light-sensing cone photoreceptors, causing irreversible sight impairment. Photoreceptor replacement may restore vision by transplanting healthy cells, which must form new synaptic connections with the recipient retina. Despite recent advances, convincing evidence of functional connectivity arising from transplanted human cone photoreceptors in advanced retinal degeneration is lacking. Here, we show restoration of visual function after transplantation of purified human pluripotent stem cell-derived cones into a mouse model of advanced degeneration. Transplanted human cones elaborate nascent outer segments and make putative synapses with recipient murine bipolar cells (BCs), which themselves undergo significant remodeling. Electrophysiological and behavioral assessments demonstrate restoration of surprisingly complex light-evoked retinal ganglion cell responses and improved light-evoked behaviors in treated animals. Stringent controls exclude alternative explanations, including material transfer and neuroprotection. These data provide crucial validation for photoreceptor replacement therapy and for the potential to rescue cone-mediated vision.
Collapse
Affiliation(s)
- Joana Ribeiro
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | | | - Emma L West
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | | | - Monica F Martins
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | | | - Aura Hare
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Mark Basche
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Milan Fernando
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Debbie Goh
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Neeraj Jumbo
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Matteo Rizzi
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Kate Powell
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Menahil Tariq
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | | | | | - Alexander J Smith
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Rachael A Pearson
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | | | - Robin R Ali
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK; Kellogg Eye Centre, University of Michigan, 1000 Wall St., Ann Arbor, MI 48105, USA.
| |
Collapse
|
10
|
Vicenzi S, Foa L, Gasperini RJ. Serotonin functions as a bidirectional guidance molecule regulating growth cone motility. Cell Mol Life Sci 2021; 78:2247-2262. [PMID: 32939562 PMCID: PMC11072016 DOI: 10.1007/s00018-020-03628-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/14/2020] [Accepted: 08/21/2020] [Indexed: 02/02/2023]
Abstract
The neurotransmitter serotonin has been implicated in a range of complex neurological disorders linked to alterations of neuronal circuitry. Serotonin is synthesized in the developing brain before most neuronal circuits become fully functional, suggesting that serotonin might play a distinct regulatory role in shaping circuits prior to its function as a classical neurotransmitter. In this study, we asked if serotonin acts as a guidance cue by examining how serotonin alters growth cone motility of rodent sensory neurons in vitro. Using a growth cone motility assay, we found that serotonin acted as both an attractive and repulsive guidance cue through a narrow concentration range. Extracellular gradients of 50 µM serotonin elicited attraction, mediated by the serotonin 5-HT2a receptor while 100 µM serotonin elicited repulsion mediated by the 5-HT1b receptor. Importantly, high resolution imaging of growth cones indicated that these receptors signalled through their canonical pathways of endoplasmic reticulum-mediated calcium release and cAMP depletion, respectively. This novel characterisation of growth cone motility in response to serotonin gradients provides compelling evidence that secreted serotonin acts at the molecular level as an axon guidance cue to shape neuronal circuit formation during development.
Collapse
Affiliation(s)
- Silvia Vicenzi
- School of Medicine, University of Tasmania, Hobart, Australia
| | - Lisa Foa
- School of Psychological Sciences, University of Tasmania, Hobart, Australia
| | | |
Collapse
|
11
|
García-Guillén IM, Alonso A, Puelles L, Marín F, Aroca P. Multiple Regionalized Genes and Their Putative Networks in the Interpeduncular Nucleus Suggest Complex Mechanisms of Neuron Development and Axon Guidance. Front Neuroanat 2021; 15:643320. [PMID: 33664652 PMCID: PMC7921722 DOI: 10.3389/fnana.2021.643320] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/26/2021] [Indexed: 12/27/2022] Open
Abstract
The interpeduncular nucleus (IPN) is a highly conserved limbic structure in the vertebrate brain, located in the isthmus and rhombomere 1. It is formed by various populations that migrate from different sites to the distinct domains within the IPN: the prodromal, rostral interpeduncular, and caudal interpeduncular nuclei. The aim here was to identify genes that are differentially expressed across these domains, characterizing their putative functional roles and interactions. To this end, we screened the 2,038 genes in the Allen Developing Mouse Brain Atlas database expressed at E18.5 and we identified 135 genes expressed within the IPN. The functional analysis of these genes highlighted an overrepresentation of gene families related to neuron development, cell morphogenesis and axon guidance. The interactome analysis within each IPN domain yielded specific networks that mainly involve members of the ephrin/Eph and Cadherin families, transcription factors and molecules related to synaptic neurotransmission. These results bring to light specific mechanisms that might participate in the formation, molecular regionalization, axon guidance and connectivity of the different IPN domains. This genoarchitectonic model of the IPN enables data on gene expression and interactions to be integrated and interpreted, providing a basis for the further study of the connectivity and function of this poorly understood nuclear complex under both normal and pathological conditions.
Collapse
Affiliation(s)
- Isabel M García-Guillén
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia and IMIB-Arrixaca, Murcia, Spain
| | - Antonia Alonso
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia and IMIB-Arrixaca, Murcia, Spain
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia and IMIB-Arrixaca, Murcia, Spain
| | - Faustino Marín
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia and IMIB-Arrixaca, Murcia, Spain
| | - Pilar Aroca
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, Regional Campus of International Excellence "Campus Mare Nostrum", University of Murcia and IMIB-Arrixaca, Murcia, Spain
| |
Collapse
|
12
|
Sailem HZ, Al Haj Zen A. Morphological landscape of endothelial cell networks reveals a functional role of glutamate receptors in angiogenesis. Sci Rep 2020; 10:13829. [PMID: 32796870 PMCID: PMC7428010 DOI: 10.1038/s41598-020-70440-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/20/2020] [Indexed: 12/16/2022] Open
Abstract
Angiogenesis plays a key role in several diseases including cancer, ischemic vascular disease, and Alzheimer's disease. Chemical genetic screening of endothelial tube formation provides a robust approach for identifying signalling components that impact microvascular network morphology as well as endothelial cell biology. However, the analysis of the resulting imaging datasets has been limited to a few phenotypic features such as the total tube length or the number of branching points. Here we developed a high content analysis framework for detailed quantification of various aspects of network morphology including network complexity, symmetry and topology. By applying our approach to a high content screen of 1,280 characterised drugs, we found that drugs that result in a similar phenotype share the same mechanism of action or common downstream signalling pathways. Our multiparametric analysis revealed that a group of glutamate receptor antagonists enhances branching and network connectivity. Using an integrative meta-analysis approach, we validated the link between these receptors and angiogenesis. We further found that the expression of these genes is associated with the prognosis of Alzheimer's patients. In conclusion, our work shows that detailed image analysis of complex endothelial phenotypes can reveal new insights into biological mechanisms modulating the morphogenesis of endothelial networks and identify potential therapeutics for angiogenesis-related diseases.
Collapse
Affiliation(s)
- Heba Z Sailem
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7LF, UK.
| | - Ayman Al Haj Zen
- College of Health and Life Sciences, Hamad Bin Khalifa University, Education City, Doha, Qatar.
- Radcliffe Department of Medicine, British Heart Foundation Centre of Research Excellence, University of Oxford, Oxford, UK.
| |
Collapse
|
13
|
Singh L, Kaur A, Garg S, Singh AP, Bhatti R. Protective Effect of Esculetin, Natural Coumarin in Mice Model of Fibromyalgia: Targeting Pro-Inflammatory Cytokines and MAO-A. Neurochem Res 2020; 45:2364-2374. [DOI: 10.1007/s11064-020-03095-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/04/2020] [Accepted: 07/09/2020] [Indexed: 10/23/2022]
|
14
|
Léger C, Dupré N, Aligny C, Bénard M, Lebon A, Henry V, Hauchecorne M, Galas L, Frebourg T, Leroux P, Vivien D, Lecointre M, Marret S, Gonzalez BJ. Glutamate controls vessel-associated migration of GABA interneurons from the pial migratory route via NMDA receptors and endothelial protease activation. Cell Mol Life Sci 2020; 77:1959-1986. [PMID: 31392351 PMCID: PMC7229000 DOI: 10.1007/s00018-019-03248-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 07/08/2019] [Accepted: 07/23/2019] [Indexed: 12/20/2022]
Abstract
During cortex development, fine interactions between pyramidal cells and migrating GABA neurons are required to orchestrate correct positioning of interneurons, but cellular and molecular mechanisms are not yet clearly understood. Functional and age-specific expression of NMDA receptors by neonate endothelial cells suggests a vascular contribution to the trophic role of glutamate during cortical development. Associating functional and loss-of-function approaches, we found that glutamate stimulates activity of the endothelial proteases MMP-9 and t-PA along the pial migratory route (PMR) and radial cortical microvessels. Activation of MMP-9 was NMDAR-dependent and abrogated in t-PA-/- mice. Time-lapse recordings revealed that glutamate stimulated migration of GABA interneurons along vessels through an NMDAR-dependent mechanism. In Gad67-GFP mice, t-PA invalidation and in vivo administration of an MMP inhibitor impaired positioning of GABA interneurons in superficial cortical layers, whereas Grin1 endothelial invalidation resulted in a strong reduction of the thickness of the pial migratory route, a marked decrease of the glutamate-induced MMP-9-like activity along the PMR and a depopulation of interneurons in superficial cortical layers. This study supports that glutamate controls the vessel-associated migration of GABA interneurons by regulating the activity of endothelial proteases. This effect requires endothelial NMDAR and is t-PA-dependent. These neurodevelopmental data reinforce the debate regarding safety of molecules with NMDA-antagonist properties administered to preterm and term neonates.
Collapse
Affiliation(s)
- Cécile Léger
- Normandie University, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Nicolas Dupré
- Normandie University, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Caroline Aligny
- Normandie University, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Magalie Bénard
- Normandie University, UNIROUEN, INSERM, PRIMACEN, Rouen, France
| | - Alexis Lebon
- Normandie University, UNIROUEN, INSERM, PRIMACEN, Rouen, France
| | - Vincent Henry
- Normandie University, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Michelle Hauchecorne
- Normandie University, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Ludovic Galas
- Normandie University, UNIROUEN, INSERM, PRIMACEN, Rouen, France
| | - Thierry Frebourg
- Normandie University, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Philippe Leroux
- Normandie University, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Denis Vivien
- Inserm, Université Caen-Normandie, Inserm, UMR-S U1237 "Physiopathology and Imaging of Neurological Disorders" (PhIND), GIP Cyceron, Caen, France
- Department of Clinical Research, Caen University Hospital, CHU Caen, Caen, France
| | - Maryline Lecointre
- Normandie University, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Stéphane Marret
- Normandie University, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Bruno J Gonzalez
- Normandie University, UNIROUEN, INSERM U1245 and Rouen University Hospital, Department of Neonatal Paediatrics and Intensive Care, F 76000, Normandy Centre for Genomic and Personalized Medicine, Rouen, France.
| |
Collapse
|
15
|
Liu C, Li Z, Huang Z, Zhang K, Hu C, Zuo Z, Li Y. Ligustrazine Enhances the Hypnotic and Analgesic Effect of Ketamine in Mice. Biol Pharm Bull 2018; 41:690-696. [PMID: 29467345 DOI: 10.1248/bpb.b17-00869] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Chuiliang Liu
- Department of Anesthesiology, ChanCheng Center Hospital
| | - Zhipeng Li
- Department of Anesthesiology, ChanCheng Center Hospital
| | - Zeqi Huang
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
- Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Kun Zhang
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
- Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Chuwen Hu
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
- Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| | - Zhiyi Zuo
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
- Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
- Department of Anesthesiology, University of Virginia Health System
| | - Yujuan Li
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
- Laboratory of RNA and Major Diseases of Brain and Hearts, Sun Yat-sen Memorial Hospital, Sun Yat-sen University
| |
Collapse
|
16
|
Jarero-Basulto JJ, Gasca-Martínez Y, Rivera-Cervantes MC, Ureña-Guerrero ME, Feria-Velasco AI, Beas-Zarate C. Interactions Between Epilepsy and Plasticity. Pharmaceuticals (Basel) 2018; 11:ph11010017. [PMID: 29414852 PMCID: PMC5874713 DOI: 10.3390/ph11010017] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/01/2018] [Accepted: 02/06/2018] [Indexed: 02/06/2023] Open
Abstract
Undoubtedly, one of the most interesting topics in the field of neuroscience is the ability of the central nervous system to respond to different stimuli (normal or pathological) by modifying its structure and function, either transiently or permanently, by generating neural cells and new connections in a process known as neuroplasticity. According to the large amount of evidence reported in the literature, many stimuli, such as environmental pressures, changes in the internal dynamic steady state of the organism and even injuries or illnesses (e.g., epilepsy) may induce neuroplasticity. Epilepsy and neuroplasticity seem to be closely related, as the two processes could positively affect one another. Thus, in this review, we analysed some neuroplastic changes triggered in the hippocampus in response to seizure-induced neuronal damage and how these changes could lead to the establishment of temporal lobe epilepsy, the most common type of focal human epilepsy.
Collapse
Affiliation(s)
- José J Jarero-Basulto
- Cellular Neurobiology Laboratory, Cell and Molecular Biology Department, CUCBA, University of Guadalajara, 45220 Zapopan, Jalisco, Mexico.
| | - Yadira Gasca-Martínez
- Cellular Neurobiology Laboratory, Cell and Molecular Biology Department, CUCBA, University of Guadalajara, 45220 Zapopan, Jalisco, Mexico.
| | - Martha C Rivera-Cervantes
- Cellular Neurobiology Laboratory, Cell and Molecular Biology Department, CUCBA, University of Guadalajara, 45220 Zapopan, Jalisco, Mexico.
| | - Mónica E Ureña-Guerrero
- Neurotransmission Biology Laboratory, Cell and Molecular Biology Department, CUCBA, University of Guadalajara, 45220 Zapopan, Jalisco, Mexico.
| | - Alfredo I Feria-Velasco
- Cellular Neurobiology Laboratory, Cell and Molecular Biology Department, CUCBA, University of Guadalajara, 45220 Zapopan, Jalisco, Mexico.
| | - Carlos Beas-Zarate
- Development and Neural Regeneration Laboratory, Cell and Molecular Biology Department, CUCBA, University of Guadalajara, 45220 Zapopan, Jalisco, Mexico.
| |
Collapse
|
17
|
Morelli G, Avila A, Ravanidis S, Aourz N, Neve RL, Smolders I, Harvey RJ, Rigo JM, Nguyen L, Brône B. Cerebral Cortical Circuitry Formation Requires Functional Glycine Receptors. Cereb Cortex 2017; 27:1863-1877. [PMID: 26891984 DOI: 10.1093/cercor/bhw025] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The development of the cerebral cortex is a complex process that requires the generation, migration, and differentiation of neurons. Interfering with any of these steps can impair the establishment of connectivity and, hence, function of the adult brain. Neurotransmitter receptors have emerged as critical players to regulate these biological steps during brain maturation. Among them, α2 subunit-containing glycine receptors (GlyRs) regulate cortical neurogenesis and the present work demonstrates the long-term consequences of their genetic disruption on neuronal connectivity in the postnatal cerebral cortex. Our data indicate that somatosensory cortical neurons of Glra2 knockout mice (Glra2KO) have more dendritic branches with an overall increase in total spine number. These morphological defects correlate with a disruption of the excitation/inhibition balance, thereby increasing network excitability and enhancing susceptibility to epileptic seizures after pentylenetetrazol tail infusion. Taken together, our findings show that the loss of embryonic GlyRα2 ultimately impairs the formation of cortical circuits in the mature brain.
Collapse
Affiliation(s)
- Giovanni Morelli
- BIOMED Research Institute, Hasselt University, Hasselt 3500, Belgium.,GIGA-Neurosciences.,Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R)
| | - Ariel Avila
- Program in Neurosciences and Mental Health, SickKids Research Institute, The Hospital for Sick Children (SickKids), Toronto, ON, CanadaM5G 1X8
| | | | - Najat Aourz
- Department of Pharmaceutical Chemistry and Drug Analysis, C4N, Center for Neuroscience, Vrije Universiteit Brussel, 1090 Brussel, Belgium
| | - Rachael L Neve
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ilse Smolders
- Department of Pharmaceutical Chemistry and Drug Analysis, C4N, Center for Neuroscience, Vrije Universiteit Brussel, 1090 Brussel, Belgium
| | - Robert J Harvey
- Department of Pharmacology, UCL School of Pharmacy, London WC1N 1AX, UK
| | - Jean-Michel Rigo
- BIOMED Research Institute, Hasselt University, Hasselt 3500, Belgium
| | - Laurent Nguyen
- GIGA-Neurosciences.,Interdisciplinary Cluster for Applied Genoproteomics (GIGA-R).,Walloon Excellence in Lifesciences and Biotechnology (WELBIO), University of Liège, C.H.U. Sart Tilman, Liège 4000, Belgium
| | - Bert Brône
- BIOMED Research Institute, Hasselt University, Hasselt 3500, Belgium
| |
Collapse
|
18
|
Vizlin-Hodzic D, Zhai Q, Illes S, Södersten K, Truvé K, Parris TZ, Sobhan PK, Salmela S, Kosalai ST, Kanduri C, Strandberg J, Seth H, Bontell TO, Hanse E, Ågren H, Funa K. Early onset of inflammation during ontogeny of bipolar disorder: the NLRP2 inflammasome gene distinctly differentiates between patients and healthy controls in the transition between iPS cell and neural stem cell stages. Transl Psychiatry 2017; 7:e1010. [PMID: 28117838 PMCID: PMC5545741 DOI: 10.1038/tp.2016.284] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 11/27/2016] [Indexed: 01/09/2023] Open
Abstract
Neuro-inflammation and neuronal communication are considered as mis-regulated processes in the aetiology and pathology of bipolar disorder (BD). Which and when specific signal pathways become abnormal during the ontogeny of bipolar disorder patients is unknown. To address this question, we applied induced pluripotent stem cell (iPSC) technology followed by cortical neural differentiation on adipocyte-derived cells from BD type I patients (with psychotic episodes in psychiatric history) and healthy volunteers (controls). RNA sequencing in iPSC and cortical neural stem cell (NSC) lines were used to examine alterations between the transcriptomes from BD I and control samples during transition from the pluripotent stage towards the neural developmental stage. At the iPSC stage, the most highly significant differentially expressed gene (DEG) was the NLRP2 inflammasome (P=2.66 × 10-10). Also among 42 DEGs at the NSC stage, NLRP2 showed the strongest statistical significance (P=3.07 × 10-19). In addition, we have also identified several cytoskeleton-associated genes as DEGs from the NSC stage, such as TMP2, TAGLN and ACTA2; the former two genes are recognised for the first time to be associated with BD. Our results also suggest that iPSC-derived BD-cortical NSCs carry several abnormalities in dopamine and GABA receptor canonical pathways, underlining that our in vitro BD model reflects pathology in the central nervous system. This would indicate that mis-regulated gene expression of inflammatory, neurotransmitter and cytoskeletal signalling occurs during early fetal brain development of BD I patients.
Collapse
Affiliation(s)
- D Vizlin-Hodzic
- Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Q Zhai
- Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - S Illes
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Oncology Laboratory, Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - K Södersten
- Institute of Neuroscience and Physiology, Section of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - K Truvé
- Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - T Z Parris
- Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - P K Sobhan
- Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - S Salmela
- Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - S T Kosalai
- Institute of Biomedicine, Department of Medical Genetics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - C Kanduri
- Institute of Biomedicine, Department of Medical Genetics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - J Strandberg
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - H Seth
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - T O Bontell
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Department of Clinical Pathology and Cytology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - E Hanse
- Institute of Neuroscience and Physiology, Department of Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - H Ågren
- Institute of Neuroscience and Physiology, Section of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Institute of Neuroscience and Physiology, Section of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, SE 405 30 Gothenburg, Sweden E-mail:
| | - K Funa
- Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden,Oncology Laboratory, Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden,Oncology Laboratory, Department of Pathology, Sahlgrenska University Hospital, SE 413 45 Gothenburg, Sweden. E-mail:
| |
Collapse
|
19
|
Schaefers ATU, Teuchert-Noodt G. Developmental neuroplasticity and the origin of neurodegenerative diseases. World J Biol Psychiatry 2016; 17:587-599. [PMID: 23705632 DOI: 10.3109/15622975.2013.797104] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVES Neurodegenerative diseases like Alzheimer's and Parkinson's Disease, marked by characteristic protein aggregations, are more and more accepted to be synaptic disorders and to arise from a combination of genetic and environmental factors. In this review we propose our concept that neuroplasticity might constitute a link between early life challenges and neurodegeneration. METHODS After introducing the general principles of neuroplasticity, we show how adverse environmental stimuli during development impact adult neuroplasticity and might lead to neurodegenerative processes. RESULTS There are significant overlaps between neurodevelopmental and neurodegenerative processes. Proteins that represent hallmarks of neurodegeneration are involved in plastic processes under physiological conditions. Brain regions - particularly the hippocampus - that retain life-long plastic capacities are the key targets of neurodegeneration. Neuroplasticity is highest in young age making the brain more susceptible to external influences than later in life. Impacts during critical periods have life-long consequences on neuroplasticity and structural self-organization and are known to be common risk factors for neurodegenerative diseases. CONCLUSIONS Several lines of evidence support a link between developmental neuroplasticity and neurodegenerative processes later in life. A deeper insight into these processes is necessary to design strategies to mitigate or even prevent neurodegenerative pathologies.
Collapse
|
20
|
Lamin A/C Is Required for ChAT-Dependent Neuroblastoma Differentiation. Mol Neurobiol 2016; 54:3729-3744. [PMID: 27221609 DOI: 10.1007/s12035-016-9902-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 05/03/2016] [Indexed: 01/21/2023]
Abstract
The mouse neuroblastoma N18TG2 clone is unable to differentiate and is defective for the enzymes of the biosynthesis of neurotransmitters. The forced expression of choline acetyltransferase (ChAT) in these cells results in the synthesis and release of acetylcholine (Ach) and hence in the expression of neurospecific features and markers. To understand how the expression of ChAT triggered neuronal differentiation, we studied the differences in genome-wide transcription profiles between the N18TG2 parental cells and its ChAT-expressing 2/4 derived clone. The engagement of the 2/4 cells in the neuronal developmental program was confirmed by the increase of the expression level of several differentiation-related genes and by the reduction of the amount of transcripts of cell cycle genes. At the same time, we observed a massive reorganization of cytoskeletal proteins in terms of gene expression, with the accumulation of the nucleoskeletal lamina component Lamin A/C in differentiating cells. The increase of the Lmna transcripts induced by ChAT expression in 2/4 cells was mimicked treating the parental N18TG2 cells with the acetylcholine receptor agonist carbachol, thus demonstrating the direct role played by this receptor in neuron nuclei maturation. Conversely, a treatment of 2/4 cells with the muscarinic receptor antagonist atropine resulted in the reduction of the amount of Lmna RNA. Finally, the hypothesis that Lmna gene product might play a crucial role in the ChAT-dependent molecular differentiation cascade was strongly supported by Lmna knockdown in 2/4 cells leading to the downregulation of genes involved in differentiation and cytoskeleton formation and to the upregulation of genes known to regulate self-renewal and stemness.
Collapse
|
21
|
Gill I, Droubi S, Giovedi S, Fedder KN, Bury LAD, Bosco F, Sceniak MP, Benfenati F, Sabo SL. Presynaptic NMDA receptors - dynamics and distribution in developing axons in vitro and in vivo. J Cell Sci 2014; 128:768-80. [PMID: 25526735 DOI: 10.1242/jcs.162362] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
During cortical development, N-methyl-D-aspartate (NMDA) receptors (NMDARs) facilitate presynaptic terminal formation, enhance neurotransmitter release and are required in presynaptic neurons for spike-timing-dependent long-term depression (tLTD). However, the extent to which NMDARs are found within cortical presynaptic terminals has remained controversial, and the sub-synaptic localization and dynamics of axonal NMDARs are unknown. Here, using live confocal imaging and biochemical purification of presynaptic membranes, we provide strong evidence that NMDARs localize to presynaptic terminals in vitro and in vivo in a developmentally regulated manner. The NR1 and NR2B subunits (also known as GRIN1 and GRIN2B, respectively) were found within the active zone membrane, where they could respond to synaptic glutamate release. Surprisingly, NR1 also appeared in glutamatergic and GABAergic synaptic vesicles. During synaptogenesis, NR1 was mobile throughout axons - including growth cones and filopodia, structures that are involved in synaptogenesis. Upon synaptogenic contact, NMDA receptors were quickly recruited to terminals by neuroligin-1 signaling. Unlike dendrites, the trafficking and distribution of axonal NR1 were insensitive to activity changes, including NMDA exposure, local glutamate uncaging or action potential blockade. These results support the idea that presynaptic NMDARs play an early role in presynaptic development.
Collapse
Affiliation(s)
- Ishwar Gill
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Sammy Droubi
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Silvia Giovedi
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy
| | - Karlie N Fedder
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Luke A D Bury
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Federica Bosco
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy
| | - Michael P Sceniak
- Department of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Fabio Benfenati
- Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Shasta L Sabo
- Department of Pharmacology, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA Department of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| |
Collapse
|
22
|
Miller DJ, Lackey EP, Hackett TA, Kaas JH. Development of myelination and cholinergic innervation in the central auditory system of a prosimian primate (Otolemur garnetti). J Comp Neurol 2014; 521:3804-16. [PMID: 23749337 DOI: 10.1002/cne.23379] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/22/2013] [Accepted: 05/23/2013] [Indexed: 11/07/2022]
Abstract
Change in the timeline of neurobiological growth is an important source of biological variation, and thus phenotypic evolution. However, no study has to date investigated sensory system development in any of the prosimian primates that are thought to most closely resemble our earliest primate ancestors. Acetylcholine (ACh) is a neurotransmitter critical to normal brain function by regulating synaptic plasticity associated with attention and learning. Myelination is an important structural component of the brain because it facilitates rapid neuronal communication. In this work we investigated the expression of acetylcholinesterase (AChE) and the density of myelinated axons throughout postnatal development in the inferior colliculus (IC), medial geniculate complex (MGC), and auditory cortex (auditory core, belt, and parabelt) in Garnett's greater galago (Otolemur garnetti). We found that the IC and MGC exhibit relatively high myelinated fiber length density (MFLD) values at birth and attain adult-like values by the species-typical age at weaning. In contrast, neocortical auditory fields are relatively unmyelinated at birth and only attain adult-like MFLD values by the species-typical age at puberty. Analysis of AChE expression indicated that, in contrast to evidence from rodent samples, the adult-like distribution of AChE in the core area of auditory cortex, dense bands in layers I, IIIb/IV, and Vb/VI, is present at birth. These data indicate the differential developmental trajectory of central auditory system structures and demonstrate the early onset of adult-like AChE expression in primary auditory cortex in O. garnetti, suggesting the auditory system is more developed at birth in primates compared to rodents.
Collapse
Affiliation(s)
- Daniel J Miller
- Department of Psychology, Vanderbilt University, Nashville, Tennessee, 37205
| | | | | | | |
Collapse
|
23
|
Feltes BC, de Faria Poloni J, Nunes IJG, Bonatto D. Fetal alcohol syndrome, chemo-biology and OMICS: ethanol effects on vitamin metabolism during neurodevelopment as measured by systems biology analysis. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2014; 18:344-63. [PMID: 24816220 DOI: 10.1089/omi.2013.0144] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Fetal alcohol syndrome (FAS) is a prenatal disease characterized by fetal morphological and neurological abnormalities originating from exposure to alcohol. Although FAS is a well-described pathology, the molecular mechanisms underlying its progression are virtually unknown. Moreover, alcohol abuse can affect vitamin metabolism and absorption, although how alcohol impairs such biochemical pathways remains to be elucidated. We employed a variety of systems chemo-biology tools to understand the interplay between ethanol metabolism and vitamins during mouse neurodevelopment. For this purpose, we designed interactomes and employed transcriptomic data analysis approaches to study the neural tissue of Mus musculus exposed to ethanol prenatally and postnatally, simulating conditions that could lead to FAS development at different life stages. Our results showed that FAS can promote early changes in neurotransmitter release and glutamate equilibrium, as well as an abnormal calcium influx that can lead to neuroinflammation and impaired neurodifferentiation, both extensively connected with vitamin action and metabolism. Genes related to retinoic acid, niacin, vitamin D, and folate metabolism were underexpressed during neurodevelopment and appear to contribute to neuroinflammation progression and impaired synapsis. Our results also indicate that genes coding for tubulin, tubulin-associated proteins, synapse plasticity proteins, and proteins related to neurodifferentiation are extensively affected by ethanol exposure. Finally, we developed a molecular model of how ethanol can affect vitamin metabolism and impair neurodevelopment.
Collapse
Affiliation(s)
- Bruno César Feltes
- Centro de Biotecnologia da Universidade Federal do Rio Grande do Sul , Departamento de Biologia Molecular e Biotecnologia, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | | | | | | |
Collapse
|
24
|
Money KM, Stanwood GD. Developmental origins of brain disorders: roles for dopamine. Front Cell Neurosci 2013; 7:260. [PMID: 24391541 PMCID: PMC3867667 DOI: 10.3389/fncel.2013.00260] [Citation(s) in RCA: 147] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 12/01/2013] [Indexed: 01/11/2023] Open
Abstract
Neurotransmitters and neuromodulators, such as dopamine, participate in a wide range of behavioral and cognitive functions in the adult brain, including movement, cognition, and reward. Dopamine-mediated signaling plays a fundamental neurodevelopmental role in forebrain differentiation and circuit formation. These developmental effects, such as modulation of neuronal migration and dendritic growth, occur before synaptogenesis and demonstrate novel roles for dopaminergic signaling beyond neuromodulation at the synapse. Pharmacologic and genetic disruptions demonstrate that these effects are brain region- and receptor subtype-specific. For example, the striatum and frontal cortex exhibit abnormal neuronal structure and function following prenatal disruption of dopamine receptor signaling. Alterations in these processes are implicated in the pathophysiology of neuropsychiatric disorders, and emerging studies of neurodevelopmental disruptions may shed light on the pathophysiology of abnormal neuronal circuitry in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Kelli M Money
- Neuroscience Graduate Program, Vanderbilt University Nashville, TN, USA ; Vanderbilt Medical Scientist Training Program, Vanderbilt University Nashville, TN, USA
| | - Gregg D Stanwood
- Department of Pharmacology, Vanderbilt University Nashville, TN, USA ; Vanderbilt Kennedy Center for Research on Human Development, Vanderbilt University Nashville, TN, USA
| |
Collapse
|
25
|
Abstract
Throughout development, the nervous system produces patterned spontaneous activity. Research over the past two decades has revealed a core group of mechanisms that mediate spontaneous activity in diverse circuits. Many circuits engage several of these mechanisms sequentially to accommodate developmental changes in connectivity. In addition to shared mechanisms, activity propagates through developing circuits and neuronal pathways (i.e., linked circuits in different brain areas) in stereotypic patterns. Increasing evidence suggests that spontaneous network activity shapes synaptic development in vivo Variations in activity-dependent plasticity may explain how similar mechanisms and patterns of activity can be employed to establish diverse circuits. Here, I will review common mechanisms and patterns of spontaneous activity in emerging neural networks and discuss recent insights into their contribution to synaptic development.
Collapse
Affiliation(s)
- Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, MO, USA Department of Anatomy and Neurobiology, Washington University School of Medicine, Saint Louis, MO, USA Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
26
|
Karlsson O, Bergquist J, Andersson M. Quality measures of imaging mass spectrometry aids in revealing long-term striatal protein changes induced by neonatal exposure to the cyanobacterial toxin β-N-methylamino-L-alanine (BMAA). Mol Cell Proteomics 2013; 13:93-104. [PMID: 24126143 PMCID: PMC3879633 DOI: 10.1074/mcp.m113.031435] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Many pathological processes are not directly correlated to dramatic alterations in protein levels. The changes in local concentrations of important proteins in a subset of cells or at specific loci are likely to play a significant role in disease etiologies, but the precise location might be unknown, or the concentration might be too small to be adequately sampled for traditional proteomic techniques. Matrix-assisted laser desorption ionization (MALDI) imaging mass spectrometry (IMS) is a unique analytical method that combines analysis of multiple molecular species and of their distribution in a single platform. As reproducibility is essential for successful biomarker discovery, it is important to systematically assess data quality in biologically relevant MALDI IMS experiments. In the present study, we applied four simple tools to study the reproducibility for individual sections, within-group variation, and between-group variation of data acquired from brain sections of 21 animals divided into three treatment groups. We also characterized protein changes in distinct regions of the striatum from six-month-old rats treated neonatally (postnatal days 9–10) with the cyanobacterial toxin β-N-methylamino-l-alanine (BMAA), which has been implicated in neurodegenerative diseases. The results showed that optimized experimental settings can yield high-quality MALDI IMS data with relatively low variation (14% to 15% coefficient of variance) that allow the characterization of subtle changes in protein expression in various subregions of the brain. This was further exemplified by the dose-dependent reduction of myelin basic protein in the caudate putamen and the nucleus accumbens of adult rats neonatally treated with BMAA (150 and 460 mg/kg). The reduction in myelin basic protein was confirmed through immunohistochemistry and indicates that developmental exposure to BMAA may induce structural effects on axonal growth and/or directly on the proliferation of oligodendrocytes and myelination, which might be important for the previously shown BMAA-induced long-term cognitive impairments.
Collapse
Affiliation(s)
- Oskar Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, 751 24 Uppsala, Sweden
| | | | | |
Collapse
|
27
|
Engskog MKR, Karlsson O, Haglöf J, Elmsjö A, Brittebo E, Arvidsson T, Pettersson C. The cyanobacterial amino acid β-N-methylamino-l-alanine perturbs the intermediary metabolism in neonatal rats. Toxicology 2013; 312:6-11. [PMID: 23886855 DOI: 10.1016/j.tox.2013.07.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 06/27/2013] [Accepted: 07/12/2013] [Indexed: 01/21/2023]
Abstract
The neurotoxic amino acid β-N-methylamino-l-alanine (BMAA) is produced by most cyanobacteria. BMAA is considered as a potential health threat because of its putative role in neurodegenerative diseases. We have previously observed cognitive disturbances and morphological brain changes in adult rodents exposed to BMAA during the development. The aim of this study was to characterize changes of major intermediary metabolites in serum following neonatal exposure to BMAA using a non-targeted metabolomic approach. NMR spectroscopy was used to obtain serum metabolic profiles from neonatal rats exposed to BMAA (40, 150, 460mg/kg) or vehicle on postnatal days 9-10. Multivariate data analysis of binned NMR data indicated metabolic pattern differences between the different treatment groups. In particular five metabolites, d-glucose, lactate, 3-hydroxybutyrate, creatine and acetate, were changed in serum of BMAA-treated neonatal rats. These metabolites are associated with changes in energy metabolism and amino acid metabolism. Further statistical analysis disclosed that all the identified serum metabolites in the lowest dose group were significantly (p<0.05) decreased. The neonatal rat model used in this study is so far the only animal model that displays significant biochemical and behavioral effects after a low short-term dose of BMAA. The demonstrated perturbation of intermediary metabolism may contribute to BMAA-induced developmental changes that result in long-term effects on adult brain function.
Collapse
Affiliation(s)
- Mikael K R Engskog
- Division of Analytical Pharmaceutical Chemistry, Uppsala University, Box 574, SE-751 23 Uppsala, Sweden.
| | | | | | | | | | | | | |
Collapse
|
28
|
Karlsson O, Kultima K, Wadensten H, Nilsson A, Roman E, Andrén PE, Brittebo EB. Neurotoxin-induced neuropeptide perturbations in striatum of neonatal rats. J Proteome Res 2013; 12:1678-90. [PMID: 23410195 DOI: 10.1021/pr3010265] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The cyanobacterial toxin β-N-methylamino-l-alanine (BMAA) is suggested to play a role in neurodegenerative disease. We have previously shown that although the selective uptake of BMAA in the rodent neonatal striatum does not cause neuronal cell death, exposure during the neonatal development leads to cognitive impairments in adult rats. The aim of the present study was to characterize the changes in the striatal neuropeptide systems of male and female rat pups treated neonatally (postnatal days 9-10) with BMAA (40-460 mg/kg). The label-free quantification of the relative levels of endogenous neuropeptides using mass spectrometry revealed that 25 peptides from 13 neuropeptide precursors were significantly changed in the rat neonatal striatum. The exposure to noncytotoxic doses of BMAA induced a dose-dependent increase of neurosecretory protein VGF-derived peptides, and changes in the relative levels of cholecystokinin, chromogranin, secretogranin, MCH, somatostatin and cortistatin-derived peptides were observed at the highest dose. In addition, the results revealed a sex-dependent increase in the relative level of peptides derived from the proenkephalin-A and protachykinin-1 precursors, including substance P and neurokinin A, in female pups. Because several of these peptides play a critical role in the development and survival of neurons, the observed neuropeptide changes might be possible mediators of BMAA-induced behavioral changes. Moreover, some neuropeptide changes suggest potential sex-related differences in susceptibility toward this neurotoxin. The present study also suggests that neuropeptide profiling might provide a sensitive characterization of the BMAA-induced noncytotoxic effects on the developing brain.
Collapse
Affiliation(s)
- Oskar Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University , SE-751 24 Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
29
|
Layer PG, Klaczinski J, Salfelder A, Sperling LE, Thangaraj G, Tuschl C, Vogel-Höpker A. Cholinesterases in development: AChE as a firewall to inhibit cell proliferation and support differentiation. Chem Biol Interact 2012; 203:269-76. [PMID: 23047026 DOI: 10.1016/j.cbi.2012.09.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 08/31/2012] [Accepted: 09/06/2012] [Indexed: 10/27/2022]
Abstract
Acetylcholinesterase (AChE) is a most remarkable protein, not only because it is one of the fastest enzymes in nature, but also since it appears in many molecular forms and is regulated by elaborate genetic networks. AChE is expressed in many tissues during development and in mature organisms, as well as in healthy and diseased states. In search for alternative, "non-classical" functions of cholinesterases (ChEs), AChE could either work within the frame of classic cholinergic systems, but in non-neural tissues ("non-synaptic function"), or act non-enzymatically. Here, we review briefly some of the major ideas and advances of this field, and report on some recent progress from our own experimental work, e.g. that (i) non-neural ChEs have pronounced, predominantly enzymatic effects on early embryonic (limb) development in chick and mouse, that (ii) retinal R28 cells of the rat overexpressing synaptic AChE present a significantly decreased cell proliferation, and that (iii) in developing chick retina ACh-synthesizing and ACh-degrading cells originate from the same postmitotic precursor cells, which later form two locally opposing cell populations. We suggest that such distinct distributions of ChAT(+) vs. AChE(+) cells in the inner half retina provide graded distributions of ACh, which can direct cell differentiation and network formation. Thus, as corroborated by works from many labs, AChE can be considered a highly co-opting protein, which can combine enzymatic and non-enzymatic functions within one molecule.
Collapse
Affiliation(s)
- Paul G Layer
- Technische Universität Darmstadt, Entwicklungsbiologie & Neurogenetik, Darmstadt, Germany.
| | | | | | | | | | | | | |
Collapse
|
30
|
Gupta R, Palchaudhuri S, Chattopadhyay D. Glutamate induces neutrophil cell migration by activating class I metabotropic glutamate receptors. Amino Acids 2012; 44:757-67. [DOI: 10.1007/s00726-012-1400-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Accepted: 09/05/2012] [Indexed: 12/11/2022]
|
31
|
Transient knockdown of tyrosine hydroxylase during development has persistent effects on behaviour in adult zebrafish (Danio rerio). PLoS One 2012; 7:e42482. [PMID: 22879998 PMCID: PMC3411795 DOI: 10.1371/journal.pone.0042482] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Accepted: 07/06/2012] [Indexed: 01/11/2023] Open
Abstract
Abnormal dopamine (DA) signaling is often suggested as causative in schizophrenia. The other prominent hypothesis for this disorder, largely driven by epidemiological data, is that certain adverse events during the early stages of brain development increase an individual's risk of developing schizophrenia later in life. However, the clinical and preclinical literature consistently implicates behavioural, cognitive, and pharmacological abnormalities, implying that DA signaling is abnormal in the adult brain. How can we reconcile these two major hypotheses underlying much of the clinical and basic research into schizophrenia? In this study we have transiently knocked down tyrosine hydroxylase (TH, the rate limiting enzyme in DA synthesis) gene expression in the early stages of brain development in zebrafish using morpholinos. We show that by adulthood, TH and DA levels have returned to normal and basic DA-mediated behaviours, such as locomotion, are also normal. However, when they were exposed to a novel environment the levels of freezing and immediate positioning in deeper zones were significantly reduced in these adult fish. The neurochemistry underlying these behaviours is complex, and the exact mechanisms for these abnormal behaviours remains unknown. This study demonstrates that early transient alterations in DA ontogeny can produce persistent alterations in adult brain function and suggests that the zebrafish may be a promising model animal for future studies directed at clarifying the basic neurodevelopmental mechanisms behind complex psychiatric disease.
Collapse
|
32
|
From abnormal hippocampal synaptic plasticity in down syndrome mouse models to cognitive disability in down syndrome. Neural Plast 2012; 2012:101542. [PMID: 22848844 PMCID: PMC3403629 DOI: 10.1155/2012/101542] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 05/02/2012] [Accepted: 05/07/2012] [Indexed: 12/17/2022] Open
Abstract
Down syndrome (DS) is caused by the overexpression of genes on triplicated regions of human chromosome 21 (Hsa21). While the resulting physiological and behavioral phenotypes vary in their penetrance and severity, all individuals with DS have variable but significant levels of cognitive disability. At the core of cognitive processes is the phenomenon of synaptic plasticity, a functional change in the strength at points of communication between neurons. A wide variety of evidence from studies on DS individuals and mouse models of DS indicates that synaptic plasticity is adversely affected in human trisomy 21 and mouse segmental trisomy 16, respectively, an outcome that almost certainly extensively contributes to the cognitive impairments associated with DS. In this review, we will highlight some of the neurophysiological changes that we believe reduce the ability of trisomic neurons to undergo neuroplasticity-related adaptations. We will focus primarily on hippocampal networks which appear to be particularly impacted in DS and where consequently the majority of cellular and neuronal network research has been performed using DS animal models, in particular the Ts65Dn mouse. Finally, we will postulate on how altered plasticity may contribute to the DS cognitive disability.
Collapse
|
33
|
Krüger HS, Hanganu-Opatz IL. Neonatal cholinergic lesion alters the acoustic structure of infant rat vocalization but not the early cognitive development. Dev Psychobiol 2012; 55:294-308. [DOI: 10.1002/dev.21029] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 03/02/2012] [Indexed: 11/09/2022]
|
34
|
Palanisamy A. Maternal anesthesia and fetal neurodevelopment. Int J Obstet Anesth 2012; 21:152-62. [DOI: 10.1016/j.ijoa.2012.01.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 01/21/2012] [Accepted: 01/28/2012] [Indexed: 12/01/2022]
|
35
|
Morrow BA, Roth RH, Redmond DE, Diano S, Elsworth JD. Susceptibility to a parkinsonian toxin varies during primate development. Exp Neurol 2012; 235:273-81. [PMID: 22366325 DOI: 10.1016/j.expneurol.2012.02.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 01/18/2012] [Accepted: 02/06/2012] [Indexed: 12/21/2022]
Abstract
Symptoms of Parkinson's disease typically emerge later in life when loss of nigrostriatal dopamine neuron function exceeds the threshold of compensatory mechanisms in the basal ganglia. Although nigrostriatal dopamine neurons are lost during aging, in Parkinson's disease other detrimental factors must play a role to produce greater than normal loss of these neurons. Early development has been hypothesized to be a potentially vulnerable period when environmental or genetic abnormalities may compromise central dopamine neurons. This study uses a specific parkinsonian neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), to probe the relative vulnerability of nigrostriatal dopamine neurons at different stages of primate development. Measures of dopamine, homovanillic acid, 1-methyl-pyridinium concentrations and tyrosine hydroxylase immunoreactive neurons indicated that at mid-gestation dopamine neurons are relatively vulnerable to MPTP, whereas later in development or in the young primate these neurons are resistant to the neurotoxin. These studies highlight a potentially greater risk to the fetus of exposure during mid-gestation to environmental agents that cause oxidative stress. In addition, the data suggest that uncoupling protein-2 may be a target for retarding the progressive loss of nigrostriatal dopamine neurons that occurs in Parkinson's disease and aging.
Collapse
Affiliation(s)
- B A Morrow
- Neuropsychopharmacology Research Laboratory, Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA
| | | | | | | | | |
Collapse
|
36
|
del Puerto A, Díaz-Hernández JI, Tapia M, Gomez-Villafuertes R, Benitez MJ, Zhang J, Miras-Portugal MT, Wandosell F, Díaz-Hernández M, Garrido JJ. Adenylate cyclase 5 coordinates the action of ADP, P2Y1, P2Y13 and ATP-gated P2X7 receptors on axonal elongation. J Cell Sci 2012; 125:176-88. [PMID: 22250198 DOI: 10.1242/jcs.091736] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
In adult brains, ionotropic or metabotropic purinergic receptors are widely expressed in neurons and glial cells. They play an essential role in inflammation and neurotransmission in response to purines secreted to the extracellular medium. Recent studies have demonstrated a role for purinergic receptors in proliferation and differentiation of neural stem cells although little is known about their role in regulating the initial neuronal development and axon elongation. The objective of our study was to investigate the role of some different types of purinergic receptors, P2Y1, P2Y13 and P2X7, which are activated by ADP or ATP. To study the role and crosstalk of P2Y1, P2Y13 and P2X7 purinergic receptors in axonal elongation, we treated neurons with specific agonists and antagonists, and we nucleofected neurons with expression or shRNA plasmids. ADP and P2Y1-GFP expression improved axonal elongation; conversely, P2Y13 and ATP-gated P2X7 receptors halted axonal elongation. Signaling through each of these receptor types was coordinated by adenylate cyclase 5. In neurons nucleofected with a cAMP FRET biosensor (ICUE3), addition of ADP or Blue Brilliant G, a P2X7 antagonist, increased cAMP levels in the distal region of the axon. Adenylate cyclase 5 inhibition or suppression impaired these cAMP increments. In conclusion, our results demonstrate a crosstalk between two metabotropic and one ionotropic purinergic receptor that regulates cAMP levels through adenylate cyclase 5 and modulates axonal elongation triggered by neurotropic factors and the PI3K-Akt-GSK3 pathway.
Collapse
Affiliation(s)
- Ana del Puerto
- Department of Molecular, Cellular and Developmental Neurobiology, Instituto Cajal, CSIC, Madrid, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Xu Y, Ren XC, Quinn CC, Wadsworth WG. Axon response to guidance cues is stimulated by acetylcholine in Caenorhabditis elegans. Genetics 2011; 189:899-906. [PMID: 21868605 PMCID: PMC3213382 DOI: 10.1534/genetics.111.133546] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2011] [Accepted: 08/07/2011] [Indexed: 11/18/2022] Open
Abstract
Gradients of acetylcholine can stimulate growth cone turning when applied to neurons grown in culture, and it has been suggested that acetylcholine could act as a guidance cue. However, the role acetylcholine plays in directing axon migrations in vivo is not clear. Here, we show that acetylcholine positively regulates signaling pathways that mediate axon responses to guidance cues in Caenorhabditis elegans. Mutations that disrupt acetylcholine synthesis, transportation, and secretion affect circumferential axon guidance of the AVM neuron and in these mutants exogenously supplied acetylcholine improves AVM circumferential axon guidance. These effects are not observed for the circumferential guidance of the DD and VD motor neuron axons, which are neighbors of the AVM axon. Circumferential guidance is directed by the UNC-6 (netrin) and SLT-1 (slit) extracellular cues, and exogenously supplied acetylcholine can improve AVM axon guidance in mutants when either UNC-6- or SLT-1-induced signaling is disrupted, but not when both signaling pathways are perturbed. Not in any of the mutants does exogenously supplied acetylcholine improve DD and VD axon guidance. The ability of acetylcholine to enhance AVM axon guidance only in the presence of either UNC-6 or SLT-1 indicates that acetylcholine potentiates UNC-6 and SLT-1 guidance activity, rather than acting itself as a guidance cue. Together, our results show that for specific neurons acetylcholine plays an important role in vivo as a modulator of axon responses to guidance cues.
Collapse
Affiliation(s)
- Yan Xu
- Department of Pathology, University of Medicine and Dentistry of New Jersey–Robert Wood Johnson Medical School, Piscataway, New Jersey 08854-5635
- Department of Biological Sciences, University of Wisconsin, Milwaukee, Wisconsin 53201
| | - Xing-Cong Ren
- Department of Pathology, University of Medicine and Dentistry of New Jersey–Robert Wood Johnson Medical School, Piscataway, New Jersey 08854-5635
| | - Christopher C. Quinn
- Department of Pathology, University of Medicine and Dentistry of New Jersey–Robert Wood Johnson Medical School, Piscataway, New Jersey 08854-5635
- Department of Biological Sciences, University of Wisconsin, Milwaukee, Wisconsin 53201
| | - William G. Wadsworth
- Department of Pathology, University of Medicine and Dentistry of New Jersey–Robert Wood Johnson Medical School, Piscataway, New Jersey 08854-5635
| |
Collapse
|
38
|
Cholinergic systems mediate action from movement to higher consciousness. Behav Brain Res 2011; 221:488-98. [DOI: 10.1016/j.bbr.2009.12.046] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 12/26/2009] [Indexed: 02/06/2023]
|
39
|
Sinner B, Friedrich O, Zink W, Zausig Y, Graf BM. The toxic effects of s(+)-ketamine on differentiating neurons in vitro as a consequence of suppressed neuronal Ca2+ oscillations. Anesth Analg 2011; 113:1161-9. [PMID: 21788311 DOI: 10.1213/ane.0b013e31822747df] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND In the immature brain, neuronal Ca2+ oscillations are present during a time period of high plasticity and regulate neuronal differentiation and synaptogenesis. In this study we examined the long-term blockade of hippocampal Ca2+ oscillations, the role of the N-methyl-D-aspartate (NMDA) receptors and the effects of S(+)-ketamine on neuronal synapsin expression. METHODS Hippocampal neurons were incubated at day 15 in culture with the specific NMDA receptor antagonists dizocilpine (MK 801, 100 μM) or S(+)-ketamine (3 μM to 25 μM) for 24 hours. Terminal-deoxynucleotidyl-transferase (TUNEL) and activated caspase3 were used to detect apoptotic neurons. Ca2+ oscillations were detected after loading the neurons with the Ca2+-sensitive dye fura-2AM, and dual wavelength excitation fluorescence microscopy was performed. Ca2+/calmodulin kinase II (CaMKII) was measured using Western blots. Synapsin was identified with confocal antisynapsin immunofluorescence. RESULTS Blocking the NMDA receptor with MK 801 or 25 μM S(+)-ketamine resulted in a significant increase in apoptotic neurons. MK 801 led to a significant increase in cytosolic Ca2+ concentration and reduction of the amplitude and frequency of the Ca2+ oscillations. Similar to MK 801, the long-term application of S(+)-ketamine resulted in a significant increase in cytosolic Ca2+ concentration 24 hours after washout. This was associated with a down-regulation of the CaMKII and a reduction of the synapsin 24 hours after washout. CONCLUSION Neuronal Ca2+ oscillations mediate neuronal differentiation and synaptogenesis via activating CaMKII. By acting via the NMDA receptor, S(+)-ketamine exerts its toxic effect through the suppression of neuronal Ca2+ oscillations, down-regulation of the CaMKII, and consecutively reduced synaptic integrity.
Collapse
Affiliation(s)
- Barbara Sinner
- Department of Anesthesiology, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany.
| | | | | | | | | |
Collapse
|
40
|
Early hippocampal cell death, and late learning and memory deficits in rats exposed to the environmental toxin BMAA (β-N-methylamino-L-alanine) during the neonatal period. Behav Brain Res 2011; 219:310-20. [PMID: 21315110 DOI: 10.1016/j.bbr.2011.01.056] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Accepted: 01/29/2011] [Indexed: 11/22/2022]
Abstract
We have reported previously that exposure to the cyanobacterial neurotoxin β-N-methylamino-L-alanine (BMAA) during the neonatal period causes cognitive impairments in adult rats. The aim of this study was to investigate the long-term effects of neonatal BMAA exposure on learning and memory mechanisms and to identify early morphological changes in the neonatal brain. BMAA was injected subcutaneously in rat pups on postnatal days 9-10. BMAA (50 and 200 mg/kg) caused distinct deficits in spatial learning and memory in adult animals but no morphological changes. No impairment of recognition memory was detected, suggesting that neonatal exposure to BMAA preferentially affects neuronal systems that are important for spatial tasks. Histopathological examination revealed early neuronal cell death as determined by TUNEL staining in the hippocampus 24 h after a high dose (600 mg/kg) of BMAA whereas no changes were observed at lower doses (50 and 200 mg/kg). In addition, there was a low degree of neuronal cell death in the retrosplenial and cingulate cortices, areas that are also important for cognitive function. Taken together, these results indicate that BMAA is a developmental neurotoxin inducing long-term changes in cognitive function. The risk posed by BMAA as a potential human neurotoxin merits further consideration, particularly if the proposed biomagnifications in the food chain are confirmed.
Collapse
|
41
|
The effects of embryonic knockdown of the candidate dyslexia susceptibility gene homologue Dyx1c1 on the distribution of GABAergic neurons in the cerebral cortex. Neuroscience 2010; 172:535-46. [PMID: 21070838 DOI: 10.1016/j.neuroscience.2010.11.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 11/01/2010] [Accepted: 11/02/2010] [Indexed: 01/30/2023]
Abstract
Developmental dyslexia is a language-based learning disability, and a number of candidate dyslexia susceptibility genes have been identified, including DYX1C1, KIAA0319, and DCDC2. Knockdown of function by embryonic transfection of small hairpin RNA (shRNA) of rat homologues of these genes dramatically disrupts neuronal migration to the cerebral cortex by both cell autonomous and non-cell autonomous effects. Here we sought to investigate the extent of non-cell autonomous effects following in utero disruption of the candidate dyslexia susceptibility gene homolog Dyx1c1 by assessing the effects of this disruption on GABAergic neurons. We transfected the ventricular zone of embryonic day (E) 15.5 rat pups with either Dyx1c1 shRNA, DYX1C1 expression construct, both Dyx1c1 shRNA and DYX1C1 expression construct, or a scrambled version of Dyx1c1 shRNA, and sacrificed them at postnatal day 21. The mothers of these rats were injected with BrdU at either E13.5, E15.5, or E17.5. Neurons transfected with Dyx1c1 shRNA were bi-modally distributed in the cerebral cortex with one population in heterotopic locations at the white matter border and another migrating beyond their expected location in the cerebral cortex. In contrast, there was no disruption of migration following transfection with the DYX1C1 expression construct. We found untransfected GABAergic neurons (parvalbumin, calretinin, and neuropeptide Y) in the heterotopic collections of neurons in Dyx1c1 shRNA treated animals, supporting the hypothesis of non-cell autonomous effects. In contrast, we found no evidence that the position of the GABAergic neurons that made it to the cerebral cortex was disrupted by the embryonic transfection with any of the constructs. Taken together, these results support the notion that neurons within heterotopias caused by transfection with Dyx1c1 shRNA result from both cell autonomous and non-cell autonomous effects, but there is no evidence to support non-cell autonomous disruption of neuronal position in the cerebral cortex itself.
Collapse
|
42
|
Bonner JF, Blesch A, Neuhuber B, Fischer I. Promoting directional axon growth from neural progenitors grafted into the injured spinal cord. J Neurosci Res 2010; 88:1182-92. [PMID: 19908250 DOI: 10.1002/jnr.22288] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Spinal cord injury (SCI) is a devastating condition characterized by disruption of axonal connections, failure of axonal regeneration, and loss of motor and sensory function. The therapeutic promise of neural stem cells has been focused on cell replacement, but many obstacles remain in obtaining neuronal integration following transplantation into the injured CNS. This study investigated the neurotransmitter identity and axonal growth potential of neural progenitors following grafting into adult rats with a dorsal column lesion. We found that using a combination of neuronal and glial restricted progenitors (NRP and GRP) produced graft-derived glutamatergic and GABAergic neurons within the injury site, with minimal axonal extension. Administration of brain-derived neurotrophic factor (BDNF) with the graft promoted modest axonal growth from grafted cells. In contrast, injecting a lentiviral vector expressing BDNF rostral into the injured area generated a neurotrophin gradient and promoted directional growth of axons for up to 9 mm. Animals injected with BDNF lentivirus (at 2.5 and 5.0 mm) showed significantly more axons and significantly longer axons than control animals injected with GFP lentivirus. However, only the 5.0-mm-BDNF group showed a preference for extension in the rostral direction. We concluded that NRP/GRP grafts can be used to produce excitatory and inhibitory neurons, and neurotrophin gradients can guide axonal growth from graft-derived neurons toward putative targets. Together they can serve as a building block for neuronal cell replacement of local circuits and formation of neuronal relays.
Collapse
Affiliation(s)
- Joseph F Bonner
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, Pennsylvania
| | | | | | | |
Collapse
|
43
|
Archer T. Effects of exogenous agents on brain development: stress, abuse and therapeutic compounds. CNS Neurosci Ther 2010; 17:470-89. [PMID: 20553311 DOI: 10.1111/j.1755-5949.2010.00171.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The range of exogenous agents likely to affect, generally detrimentally, the normal development of the brain and central nervous system defies estimation although the amount of accumulated evidence is enormous. The present review is limited to certain types of chemotherapeutic and "use-and-abuse" compounds and environmental agents, exemplified by anesthetic, antiepileptic, sleep-inducing and anxiolytic compounds, nicotine and alcohol, and stress as well as agents of infection; each of these agents have been investigated quite extensively and have been shown to contribute to the etiopathogenesis of serious neuropsychiatric disorders. To greater or lesser extent, all of the exogenous agents discussed in the present treatise have been investigated for their influence upon neurodevelopmental processes during the period of the brain growth spurt and during other phases uptill adulthood, thereby maintaining the notion of critical phases for the outcome of treatment whether prenatal, postnatal, or adolescent. Several of these agents have contributed to the developmental disruptions underlying structural and functional brain abnormalities that are observed in the symptom and biomarker profiles of the schizophrenia spectrum disorders and the fetal alcohol spectrum disorders. In each case, the effects of the exogenous agents upon the status of the affected brain, within defined parameters and conditions, is generally permanent and irreversible.
Collapse
Affiliation(s)
- Trevor Archer
- Department of Psychology, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
44
|
Bérubé-Carrière N, Riad M, Dal Bo G, Lévesque D, Trudeau LE, Descarries L. The dual dopamine-glutamate phenotype of growing mesencephalic neurons regresses in mature rat brain. J Comp Neurol 2010; 517:873-91. [PMID: 19844994 DOI: 10.1002/cne.22194] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Coexpression of tyrosine hydroxylase (TH) and vesicular glutamate transporter 2 (VGLUT2) mRNAs in the ventral tegmental area (VTA) and colocalization of these proteins in axon terminals of the nucleus accumbens (nAcb) have recently been demonstrated in immature (15-day-old) rat. After neonatal 6-hydroxydopamine (6-OHDA) lesion, the proportion of VTA neurons expressing both mRNAs and of nAcb terminals displaying the two proteins was enhanced. To determine the fate of this dual phenotype in adults, double in situ hybridization and dual immunolabeling for TH and VGLUT2 were performed in 90-day-old rats subjected or not to the neonatal 6-OHDA lesion. Very few neurons expressed both mRNAs in the VTA and substantia nigra (SN) of P90 rats, even after neonatal 6-OHDA. Dually immunolabeled terminals were no longer found in the nAcb of normal P90 rats and were exceedingly rare in the nAcb of 6-OHDA-lesioned rats, although they had represented 28% and 37% of all TH terminals at P15. Similarly, 17% of all TH terminals in normal neostriatum and 46% in the dopamine neoinnervation of SN in 6-OHDA-lesioned rats were also immunoreactive for VGLUT2 at P15, but none at P90. In these three regions, all dually labeled terminals made synapse, in contradistinction to those immunolabeled for only TH or VGLUT2 at P15. These results suggest a regression of the VGLUT2 phenotype of dopamine neurons with age, following normal development, lesion, or sprouting after injury, and a role for glutamate in the establishment of synapses by these neurons.
Collapse
Affiliation(s)
- Noémie Bérubé-Carrière
- Department of Pathology and Cell Biology, Université de Montréal, Montréal, Québec, Canada H3C 3J7
| | | | | | | | | | | |
Collapse
|
45
|
Karlsson O, Roman E, Brittebo EB. Long-term Cognitive Impairments in Adult Rats Treated Neonatally with β-N-Methylamino-L-Alanine. Toxicol Sci 2009; 112:185-95. [DOI: 10.1093/toxsci/kfp196] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
46
|
Karlsson O, Lindquist NG, Brittebo EB, Roman E. Selective brain uptake and behavioral effects of the cyanobacterial toxin BMAA (beta-N-methylamino-L-alanine) following neonatal administration to rodents. Toxicol Sci 2009; 109:286-95. [PMID: 19321797 DOI: 10.1093/toxsci/kfp062] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cyanobacteria are extensively distributed in terrestrial and aquatic environments all over the world. Most cyanobacteria can produce the neurotoxin beta-N-methylamino-L-alanine (BMAA), which has been detected in several water systems and could accumulate in food chains. The aim of the study was to investigate the transfer of BMAA to fetal and neonatal brains and the effects of BMAA on the development of behavioral characteristics during the brain growth spurt (BGS) in rodents. Pregnant and neonatal mice were given an injection of (3)H-BMAA on gestational day 14 and postnatal day (PND) 10, respectively, and processed for tape-section autoradiography. The study revealed transplacental transfer of (3)H-BMAA and a significant uptake in fetal mouse brain. The radioactivity was specifically located in the hippocampus, striatum, brainstem, spinal cord and cerebellum of 10-day-old mice. The effect of repeated BMAA treatment (200 or 600 mg/kg s.c.) during BGS on rat behavior was also studied. BMAA treatment on PND 9-10 induced acute alterations, such as impaired locomotor ability and hyperactivity, in the behavior of neonatal rats. Furthermore, rats given the high dose of BMAA failed to habituate to the test environment when tested at juvenile age. In conclusion, the results demonstrated that BMAA was transferred to the neonatal brain and induced significant changes in the behavior of neonatal rats following administration during BGS. The observed behavioral changes suggest possible cognitive impairment. Increased information on the long-term effects of BMAA on cognitive function following fetal and neonatal exposure is required for assessment of the risk to children's health.
Collapse
Affiliation(s)
- Oskar Karlsson
- Department of Pharmaceutical Biosciences, Uppsala University, 751 24 Uppsala, Sweden
| | | | | | | |
Collapse
|