1
|
Symeonides C, Vacy K, Thomson S, Tanner S, Chua HK, Dixit S, Mansell T, O'Hely M, Novakovic B, Herbstman JB, Wang S, Guo J, Chia J, Tran NT, Hwang SE, Britt K, Chen F, Kim TH, Reid CA, El-Bitar A, Bernasochi GB, Delbridge LMD, Harley VR, Yap YW, Dewey D, Love CJ, Burgner D, Tang MLK, Sly PD, Saffery R, Mueller JF, Rinehart N, Tonge B, Vuillermin P, Ponsonby AL, Boon WC. Male autism spectrum disorder is linked to brain aromatase disruption by prenatal BPA in multimodal investigations and 10HDA ameliorates the related mouse phenotype. Nat Commun 2024; 15:6367. [PMID: 39112449 PMCID: PMC11306638 DOI: 10.1038/s41467-024-48897-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 05/16/2024] [Indexed: 08/10/2024] Open
Abstract
Male sex, early life chemical exposure and the brain aromatase enzyme have been implicated in autism spectrum disorder (ASD). In the Barwon Infant Study birth cohort (n = 1074), higher prenatal maternal bisphenol A (BPA) levels are associated with higher ASD symptoms at age 2 and diagnosis at age 9 only in males with low aromatase genetic pathway activity scores. Higher prenatal BPA levels are predictive of higher cord blood methylation across the CYP19A1 brain promoter I.f region (P = 0.009) and aromatase gene methylation mediates (P = 0.01) the link between higher prenatal BPA and brain-derived neurotrophic factor methylation, with independent cohort replication. BPA suppressed aromatase expression in vitro and in vivo. Male mice exposed to mid-gestation BPA or with aromatase knockout have ASD-like behaviors with structural and functional brain changes. 10-hydroxy-2-decenoic acid (10HDA), an estrogenic fatty acid alleviated these features and reversed detrimental neurodevelopmental gene expression. Here we demonstrate that prenatal BPA exposure is associated with impaired brain aromatase function and ASD-related behaviors and brain abnormalities in males that may be reversible through postnatal 10HDA intervention.
Collapse
Grants
- This multimodal project was supported by funding from the Minderoo Foundation. Funding was also provided by the National Health and Medical Research Council of Australia (NHMRC), the NHMRC-EU partnership grant for the ENDpoiNT consortium, the Australian Research Council, the Jack Brockhoff Foundation, the Shane O’Brien Memorial Asthma Foundation, the Our Women’s Our Children’s Fund Raising Committee Barwon Health, The Shepherd Foundation, the Rotary Club of Geelong, the Ilhan Food Allergy Foundation, GMHBA Limited, Vanguard Investments Australia Ltd, and the Percy Baxter Charitable Trust, Perpetual Trustees, Fred P Archer Fellowship; the Scobie Trust; Philip Bushell Foundation; Pierce Armstrong Foundation; The Canadian Institutes of Health Research; BioAutism, William and Vera Ellen Houston Memorial Trust Fund, Homer Hack Research Small Grants Scheme and the Medical Research Commercialisation Fund. This work was also supported by Ms. Loh Kia Hui. This project received funding from a NHMRC-EU partner grant with the European Union’s Horizon 2020 Research and Innovation Programme, under Grant Agreement number: 825759 (ENDpoiNTs project). This work was also supported by NHMRC Investigator Fellowships (GTN1175744 to D.B, APP1197234 to A-L.P, and GRT1193840 to P.S). The study sponsors were not involved in the collection, analysis, and interpretation of data; writing of the report; or the decision to submit the report for publication.
Collapse
Affiliation(s)
- Christos Symeonides
- Minderoo Foundation, Perth, Australia
- Murdoch Children's Research Institute, Parkville, Australia
- Centre for Community Child Health, Royal Children's Hospital, Parkville, Australia
| | - Kristina Vacy
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
- School of Population and Global Health, The University of Melbourne, Parkville, Australia
| | - Sarah Thomson
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Sam Tanner
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Hui Kheng Chua
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
- The Hudson Institute of Medical Research, Clayton, Australia
| | - Shilpi Dixit
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Toby Mansell
- Murdoch Children's Research Institute, Parkville, Australia
- Department of Pediatrics, The University of Melbourne, Parkville, Australia
| | - Martin O'Hely
- Murdoch Children's Research Institute, Parkville, Australia
- School of Medicine, Deakin University, Geelong, Australia
| | - Boris Novakovic
- Murdoch Children's Research Institute, Parkville, Australia
- School of Medicine, Deakin University, Geelong, Australia
| | - Julie B Herbstman
- Columbia Center for Children's Environmental Health, Columbia University, New York, NY, USA
- Department of Environmental Health Sciences, Columbia University, New York, NY, USA
| | - Shuang Wang
- Columbia Center for Children's Environmental Health, Columbia University, New York, NY, USA
- Department of Biostatistics, Columbia University, New York, NY, USA
| | - Jia Guo
- Columbia Center for Children's Environmental Health, Columbia University, New York, NY, USA
- Department of Biostatistics, Columbia University, New York, NY, USA
| | - Jessalynn Chia
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Nhi Thao Tran
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
- The Ritchie Centre, Department of Obstetrics and Gynaecology, School of Clinical Sciences, Monash University, Clayton, Australia
| | - Sang Eun Hwang
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Kara Britt
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
- Breast Cancer Risk and Prevention Laboratory, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia
| | - Feng Chen
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Tae Hwan Kim
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Christopher A Reid
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Anthony El-Bitar
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Gabriel B Bernasochi
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
- Faculty Medicine, Dentistry & Health Sciences, University of Melbourne, Parkville, Australia
| | - Lea M Durham Delbridge
- Faculty Medicine, Dentistry & Health Sciences, University of Melbourne, Parkville, Australia
| | - Vincent R Harley
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
- Sex Development Laboratory, Hudson Institute of Medical Research, Clayton, Australia
| | - Yann W Yap
- The Hudson Institute of Medical Research, Clayton, Australia
- Sex Development Laboratory, Hudson Institute of Medical Research, Clayton, Australia
| | - Deborah Dewey
- Departments of Paediatrics and Community Health Sciences, The University of Calgary, Calgary, Canada
| | - Chloe J Love
- School of Medicine, Deakin University, Geelong, Australia
- Barwon Health, Geelong, Australia
| | - David Burgner
- Murdoch Children's Research Institute, Parkville, Australia
- Department of Pediatrics, The University of Melbourne, Parkville, Australia
- Department of General Medicine, Royal Children's Hospital, Parkville, Australia
- Department of Pediatrics, Monash University, Clayton, Australia
| | - Mimi L K Tang
- Murdoch Children's Research Institute, Parkville, Australia
- Faculty Medicine, Dentistry & Health Sciences, University of Melbourne, Parkville, Australia
| | - Peter D Sly
- School of Medicine, Deakin University, Geelong, Australia
- Child Health Research Centre, The University of Queensland, Brisbane, Australia
- WHO Collaborating Centre for Children's Health and Environment, Brisbane, Australia
| | | | - Jochen F Mueller
- Queensland Alliance for Environmental Health Sciences, The University of Queensland, Brisbane, Australia
| | - Nicole Rinehart
- Monash Krongold Clinic, Faculty of Education, Monash University, Clayton, Australia
| | - Bruce Tonge
- Centre for Developmental Psychiatry and Psychology, Monash University, Clayton, Australia
| | - Peter Vuillermin
- Murdoch Children's Research Institute, Parkville, Australia
- School of Medicine, Deakin University, Geelong, Australia
- Barwon Health, Geelong, Australia
| | - Anne-Louise Ponsonby
- Murdoch Children's Research Institute, Parkville, Australia
- Centre for Community Child Health, Royal Children's Hospital, Parkville, Australia
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia
| | - Wah Chin Boon
- The Florey Institute of Neuroscience and Mental Health, Parkville, Australia.
- School of BioSciences, Faculty of Science, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
2
|
Sugimoto C, Perna MK, Regan SL, Tepe EA, Liou R, Fritz AL, Williams MT, Vorhees CV, Skelton MR. A Gad2 specific Slc6a8 deletion recapitulates the contextual and cued freezing deficits seen in Slc6a8 -/y mice. Brain Res 2024; 1825:148690. [PMID: 38030104 PMCID: PMC10875619 DOI: 10.1016/j.brainres.2023.148690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/14/2023] [Accepted: 11/25/2023] [Indexed: 12/01/2023]
Abstract
The creatine (Cr)-phosphocreatine shuttle is essential for ATP homeostasis. In humans, the absence of brain Cr causes significant intellectual disability, epilepsy, and language delay. Mutations of the creatine transporter (SLC6A8) are the most common cause of Cr deficiency. In rodents, Slc6a8 deletion causes deficits in spatial learning, novel object recognition (NOR), as well as in contextual and cued freezing. The mechanisms that underlie these cognitive deficits are not known. Due to the heterogeneous nature of the brain, it is important to determine which systems are affected by a loss of Cr. In this study, we generated mice lacking Slc6a8 in GABAergic neurons by crossing Slc6a8FL mice with Gad2-Cre mice. These Gad2-specific Slc6a8 knockout (cKO) mice, along with the ubiquitous Slc6a8 KO (Slc6a8-/y), Gad2-Cre+, and wild-type (WT) mice were tested in the Morris water maze, NOR, conditioned freezing, and the radial water maze. Similar to the Slc6a8-/y mice, cKO mice had reduced contextual and cued freezing compared with WT mice. The cKO mice had a mild spatial learning deficit during the reversal phase of the MWM, however they were not as pronounced as in Slc6a8-/y mice. In NOR, the Gad2-Cre mice spent less time with the novel object, similar to the reduced novel time in the cKO mice. There were no changes in radial water maze performance. Slc6a8 deletion in GABAergic neurons is sufficient to recapitulate the conditioned freezing deficits seen in Slc6a8-/y mice.
Collapse
Affiliation(s)
- Chiho Sugimoto
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States
| | - Marla K Perna
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| | - Samantha L Regan
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| | - Erin A Tepe
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States
| | - Rosalyn Liou
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| | - Adam L Fritz
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States
| | - Michael T Williams
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| | - Charles V Vorhees
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| | - Matthew R Skelton
- Department of Pediatrics, University of Cincinnati College of Medicine and Division of Neurology, Cincinnati Children's Research Foundation, Cincinnati, OH 45229, United States.
| |
Collapse
|
3
|
Przybysz KR, Spodnick MB, Johnson JM, Varlinskaya EI, Diaz MR. Moderate prenatal alcohol exposure produces sex-specific social impairments and attenuates prelimbic excitability and amygdala-cortex modulation of adult social behaviour. Addict Biol 2023; 28:e13252. [PMID: 36577734 PMCID: PMC10509785 DOI: 10.1111/adb.13252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/23/2022] [Accepted: 10/21/2022] [Indexed: 11/19/2022]
Abstract
Lifelong social impairments are common in individuals with prenatal alcohol exposure (PAE), and preclinical studies have identified gestational day (G)12 as a vulnerable timepoint for producing social deficits following binge-level PAE. While moderate (m)PAE also produces social impairments, the long-term neuroadaptations underlying them are poorly understood. Activity of the projection from the basolateral amygdala to the prelimbic cortex (BLA → PL) leads to social avoidance, and the PL is implicated in negative social behaviours, making each of these potential candidates for the neuroadaptations underlying mPAE-induced social impairments. To examine this, we first established that G12 mPAE produced sex-specific social impairments lasting into adulthood in Sprague-Dawley rats. We then chemogenetically inhibited the BLA → PL using clozapine N-oxide (CNO) during adult social testing. This revealed that CNO reduced social investigation in control males but had no effect on mPAE males or females of either exposure, indicating that mPAE attenuated the role of this projection in regulating male social behaviour and highlighting one potential mechanism by which mPAE affects male social behaviour more severely. Using whole-cell electrophysiology, we also examined mPAE-induced changes to PL pyramidal cell physiology and determined that mPAE reduced cell excitability, likely due to increased suppression by inhibitory interneurons. Overall, this work identified two mPAE-induced neuroadaptations that last into adulthood and that may underlie the sex-specific vulnerability to mPAE-induced social impairments. Future research is necessary to expand upon how these circuits modulate both normal and pathological social behaviours and to identify sex-specific mechanisms, leading to differential vulnerability in males and females.
Collapse
Affiliation(s)
- Kathryn R. Przybysz
- Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY, 13902, USA
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY, 13902, USA
| | - Mary B. Spodnick
- Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY, 13902, USA
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY, 13902, USA
| | - Julia M. Johnson
- Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY, 13902, USA
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY, 13902, USA
| | - Elena I. Varlinskaya
- Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY, 13902, USA
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY, 13902, USA
| | - Marvin R. Diaz
- Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY, 13902, USA
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY, 13902, USA
| |
Collapse
|
4
|
Simmons SC, Grecco GG, Atwood BK, Nugent FS. Effects of prenatal opioid exposure on synaptic adaptations and behaviors across development. Neuropharmacology 2023; 222:109312. [PMID: 36334764 PMCID: PMC10314127 DOI: 10.1016/j.neuropharm.2022.109312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
In this review, we focus on prenatal opioid exposure (POE) given the significant concern for the mental health outcomes of children with parents affected by opioid use disorder (OUD) in the view of the current opioid crisis. We highlight some of the less explored interactions between developmental age and sex on synaptic plasticity and associated behavioral outcomes in preclinical POE research. We begin with an overview of the rich literature on hippocampal related behaviors and plasticity across POE exposure paradigms. We then discuss recent work on reward circuit dysregulation following POE. Additional risk factors such as early life stress (ELS) could further influence synaptic and behavioral outcomes of POE. Therefore, we include an overview on the use of preclinical ELS models where ELS exposure during key critical developmental periods confers considerable vulnerability to addiction and stress psychopathology. Here, we hope to highlight the similarity between POE and ELS on development and maintenance of opioid-induced plasticity and altered opioid-related behaviors where similar enduring plasticity in reward circuits may occur. We conclude the review with some of the limitations that should be considered in future investigations. This article is part of the Special Issue on 'Opioid-induced addiction'.
Collapse
Affiliation(s)
- Sarah C Simmons
- Department of Pharmacology and Molecular Therapeutics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Greg G Grecco
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA; Medical Scientist Training Program, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Brady K Atwood
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Fereshteh S Nugent
- Department of Pharmacology and Molecular Therapeutics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
5
|
Rouzer SK, Diaz MR. Moderate prenatal alcohol exposure modifies sex-specific CRFR1 activity in the central amygdala and anxiety-like behavior in adolescent offspring. Neuropsychopharmacology 2022; 47:2140-2149. [PMID: 35478009 PMCID: PMC9556708 DOI: 10.1038/s41386-022-01327-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 04/04/2022] [Accepted: 04/06/2022] [Indexed: 02/08/2023]
Abstract
Anxiety disorders are highly prevalent among individuals with a history of prenatal alcohol exposure (PAE), and adolescent rodents demonstrate anxiety-like behavior following moderate PAE on Gestational Day (G) 12. A likely systemic target of PAE is the stress peptide corticotropin-releasing factor (CRF), as activation of CRF receptor 1 (CRFR1) in the medial nucleus of the central amygdala (CeM) is known to increase anxiety-like behavior in adults. To determine if CRF-CRFR1 interactions underly PAE-induced anxiety, functional changes in CRF system activity were investigated in adolescent male and female Sprague Dawley rats following G12 PAE. Compared to air-exposed controls, PAE increased basal spontaneous (s) inhibitory postsynaptic current (IPSC) frequency in the CeM of males, but not females. Furthermore, PAE blunted CRFR1-regulated miniature (m) IPSCs in a sex- and concentration-specific manner, and only PAE males demonstrated tonic CRFR1 activity in the CeM. It was further determined that G12 PAE decreased CRFR1 mRNA in the CeM of males while increasing regional expression in females. Finally, infusion of a CRFR1 agonist into the CeM of adolescents produced a blunted expression of CRFR1-induced anxiety-like behavior exclusively in PAE males, mirroring the blunted physiology demonstrated by PAE males. Cumulatively, these data suggest that CRFR1 function within the CeM is age- and sex-specific, and PAE not only increases the expression of anxiety-like behavior, but may reduce the efficacy of treatment for PAE-induced anxiety through CRFR1-associated mechanisms. Therefore, future research will be necessary to develop targeted treatment of anxiety disorders in individuals with a history of PAE.
Collapse
Affiliation(s)
- Siara Kate Rouzer
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY, 13902, USA
- Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY, 13902, USA
| | - Marvin R Diaz
- Department of Psychology, Center for Development and Behavioral Neuroscience, Binghamton University, Binghamton, NY, 13902, USA.
- Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY, 13902, USA.
| |
Collapse
|
6
|
Ochi S, Manabe S, Kikkawa T, Osumi N. Thirty Years' History since the Discovery of Pax6: From Central Nervous System Development to Neurodevelopmental Disorders. Int J Mol Sci 2022; 23:6115. [PMID: 35682795 PMCID: PMC9181425 DOI: 10.3390/ijms23116115] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 05/19/2022] [Accepted: 05/27/2022] [Indexed: 12/23/2022] Open
Abstract
Pax6 is a sequence-specific DNA binding transcription factor that positively and negatively regulates transcription and is expressed in multiple cell types in the developing and adult central nervous system (CNS). As indicated by the morphological and functional abnormalities in spontaneous Pax6 mutant rodents, Pax6 plays pivotal roles in various biological processes in the CNS. At the initial stage of CNS development, Pax6 is responsible for brain patterning along the anteroposterior and dorsoventral axes of the telencephalon. Regarding the anteroposterior axis, Pax6 is expressed inversely to Emx2 and Coup-TF1, and Pax6 mutant mice exhibit a rostral shift, resulting in an alteration of the size of certain cortical areas. Pax6 and its downstream genes play important roles in balancing the proliferation and differentiation of neural stem cells. The Pax6 gene was originally identified in mice and humans 30 years ago via genetic analyses of the eye phenotypes. The human PAX6 gene was discovered in patients who suffer from WAGR syndrome (i.e., Wilms tumor, aniridia, genital ridge defects, mental retardation). Mutations of the human PAX6 gene have also been reported to be associated with autism spectrum disorder (ASD) and intellectual disability. Rodents that lack the Pax6 gene exhibit diverse neural phenotypes, which might lead to a better understanding of human pathology and neurodevelopmental disorders. This review describes the expression and function of Pax6 during brain development, and their implications for neuropathology.
Collapse
Affiliation(s)
| | | | | | - Noriko Osumi
- Department of Developmental Neuroscience, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan; (S.O.); (S.M.); (T.K.)
| |
Collapse
|
7
|
Aerts T, Seuntjens E. Novel Perspectives on the Development of the Amygdala in Rodents. Front Neuroanat 2021; 15:786679. [PMID: 34955766 PMCID: PMC8696165 DOI: 10.3389/fnana.2021.786679] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/10/2021] [Indexed: 12/14/2022] Open
Abstract
The amygdala is a hyperspecialized brain region composed of strongly inter- and intraconnected nuclei involved in emotional learning and behavior. The cellular heterogeneity of the amygdalar nuclei has complicated straightforward conclusions on their developmental origin, and even resulted in contradictory data. Recently, the concentric ring theory of the pallium and the radial histogenetic model of the pallial amygdala have cleared up several uncertainties that plagued previous models of amygdalar development. Here, we provide an extensive overview on the developmental origin of the nuclei of the amygdaloid complex. Starting from older gene expression data, transplantation and lineage tracing studies, we systematically summarize and reinterpret previous findings in light of the novel perspectives on amygdalar development. In addition, migratory routes that these cells take on their way to the amygdala are explored, and known transcription factors and guidance cues that seemingly drive these cells toward the amygdala are emphasized. We propose some future directions for research on amygdalar development and highlight that a better understanding of its development could prove critical for the treatment of several neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Tania Aerts
- Laboratory of Developmental Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| | - Eve Seuntjens
- Laboratory of Developmental Neurobiology, Department of Biology, KU Leuven, Leuven, Belgium
| |
Collapse
|
8
|
Deckmann I, Santos-Terra J, Fontes-Dutra M, Körbes-Rockenbach M, Bauer-Negrini G, Schwingel GB, Riesgo R, Bambini-Junior V, Gottfried C. Resveratrol prevents brain edema, blood-brain barrier permeability, and altered aquaporin profile in autism animal model. Int J Dev Neurosci 2021; 81:579-604. [PMID: 34196408 DOI: 10.1002/jdn.10137] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/08/2021] [Accepted: 06/21/2021] [Indexed: 12/17/2022] Open
Abstract
Autism spectrum disorder can present a plethora of clinical conditions associated with the disorder, such as greater brain volume in the first years of life in a significant percentage of patients. We aimed to evaluate the brain water content, the blood-brain barrier permeability, and the expression of aquaporin 1 and 4, and GFAP in a valproic acid-animal model, assessing the effect of resveratrol. On postnatal day 30, Wistar rats of the valproic acid group showed greater permeability of the blood-brain barrier to the Evans blue dye and a higher proportion of brain water volume, prevented both by resveratrol. Prenatal exposition to valproic acid diminished aquaporin 1 in the choroid plexus, in the primary somatosensory area, in the amygdala region, and in the medial prefrontal cortex, reduced aquaporin 4 in medial prefrontal cortex and increased aquaporin 4 levels in primary somatosensory area (with resveratrol prevention). Valproic acid exposition also increased the number of astrocytes and GFAP fluorescence in both primary somatosensory area and medial prefrontal cortex. In medial prefrontal cortex, resveratrol prevented the increased fluorescence. Finally, there was an effect of resveratrol per se on the number of astrocytes and GFAP fluorescence in the amygdala region and in the hippocampus. Thus, this work demonstrates significant changes in blood-brain barrier permeability, edema formation, distribution of aquaporin 1 and 4, in addition to astrocytes profile in the animal model of autism, as well as the use of resveratrol as a tool to investigate the mechanisms involved in the pathophysiology of autism spectrum disorder.
Collapse
Affiliation(s)
- Iohanna Deckmann
- Translational Research Group in Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Porto Alegre, Brazil.,Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, University of Central Lancashire, Preston, UK
| | - Júlio Santos-Terra
- Translational Research Group in Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Porto Alegre, Brazil.,Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, University of Central Lancashire, Preston, UK
| | - Mellanie Fontes-Dutra
- Translational Research Group in Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Porto Alegre, Brazil.,Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, University of Central Lancashire, Preston, UK
| | - Marília Körbes-Rockenbach
- Translational Research Group in Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Porto Alegre, Brazil
| | - Guilherme Bauer-Negrini
- Translational Research Group in Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Porto Alegre, Brazil.,Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, University of Central Lancashire, Preston, UK
| | - Gustavo Brum Schwingel
- Translational Research Group in Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Porto Alegre, Brazil.,Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, University of Central Lancashire, Preston, UK
| | - Rudimar Riesgo
- Translational Research Group in Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Porto Alegre, Brazil.,Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, University of Central Lancashire, Preston, UK.,Department of Pediatrics, Child Neurology Unit, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Victorio Bambini-Junior
- Translational Research Group in Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Porto Alegre, Brazil.,Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, University of Central Lancashire, Preston, UK.,School of Pharmacology and Biomedical Sciences, University of Central Lancashire, Preston, UK
| | - Carmem Gottfried
- Translational Research Group in Autism Spectrum Disorder - GETTEA, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,Department of Biochemistry, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil.,National Institute of Science and Technology in Neuroimmunomodulation - INCT-NIM, Porto Alegre, Brazil.,Autism Wellbeing and Research Development - AWARD - Initiative BR-UK-CA, University of Central Lancashire, Preston, UK
| |
Collapse
|
9
|
Raam T, Hong W. Organization of neural circuits underlying social behavior: A consideration of the medial amygdala. Curr Opin Neurobiol 2021; 68:124-136. [PMID: 33940499 DOI: 10.1016/j.conb.2021.02.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/18/2021] [Accepted: 02/19/2021] [Indexed: 12/14/2022]
Abstract
The medial amygdala (MeA) is critical for the expression of a broad range of social behaviors, and is also connected to many other brain regions that mediate those same behaviors. Here, we summarize recent advances toward elucidating mechanisms that enable the MeA to regulate a diversity of social behaviors, and also consider what role the MeA plays within the broader network of regions that orchestrate social sensorimotor transformations. We outline the molecular, anatomical, and electrophysiological features of the MeA that segregate distinct social behaviors, propose experimental strategies to disambiguate sensory representations from behavioral function in the context of a social interaction, and consider to what extent MeA function may overlap with other regions mediating similar behaviors.
Collapse
Affiliation(s)
- Tara Raam
- Department of Biological Chemistry and Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Weizhe Hong
- Department of Biological Chemistry and Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
10
|
Morales L, Castro-Robles B, Abellán A, Desfilis E, Medina L. A novel telencephalon-opto-hypothalamic morphogenetic domain coexpressing Foxg1 and Otp produces most of the glutamatergic neurons of the medial extended amygdala. J Comp Neurol 2021; 529:2418-2449. [PMID: 33386618 DOI: 10.1002/cne.25103] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 12/14/2022]
Abstract
Deficits in social cognition and behavior are a hallmark of many psychiatric disorders. The medial extended amygdala, including the medial amygdala and the medial bed nucleus of the stria terminalis, is a key component of functional networks involved in sociality. However, this nuclear complex is highly heterogeneous and contains numerous GABAergic and glutamatergic neuron subpopulations. Deciphering the connections of different neurons is essential in order to understand how this structure regulates different aspects of sociality, and it is necessary to evaluate their differential implication in distinct mental disorders. Developmental studies in different vertebrates are offering new venues to understand neuronal diversity of the medial extended amygdala and are helping to establish a relation between the embryonic origin and molecular signature of distinct neurons with the functional subcircuits in which they are engaged. These studies have provided many details on the distinct GABAergic neurons of the medial extended amygdala, but information on the glutamatergic neurons is still scarce. Using an Otp-eGFP transgenic mouse and multiple fluorescent labeling, we show that most glutamatergic neurons of the medial extended amygdala originate in a distinct telencephalon-opto-hypothalamic embryonic domain (TOH), located at the transition between telencephalon and hypothalamus, which produces Otp-lineage neurons expressing the telencephalic marker Foxg1 but not Nkx2.1 during development. These glutamatergic cells include a subpopulation of projection neurons of the medial amygdala, which activation has been previously shown to promote autistic-like behavior. Our data open new venues for studying the implication of this neuron subtype in neurodevelopmental disorders producing social deficits.
Collapse
Affiliation(s)
- Lorena Morales
- Laboratory of Evolutionary and Developmental Neurobiology, Lleida's Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Catalonia, Spain
| | - Beatriz Castro-Robles
- Laboratory of Cerebrovascular, Neurodegenerative and Neuro-oncology Diseases, Research Unit, Complejo Hospitalario Universitario de Albacete, Castilla-La Mancha, Spain
| | - Antonio Abellán
- Laboratory of Evolutionary and Developmental Neurobiology, Lleida's Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Catalonia, Spain
| | - Ester Desfilis
- Laboratory of Evolutionary and Developmental Neurobiology, Lleida's Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Catalonia, Spain
| | - Loreta Medina
- Laboratory of Evolutionary and Developmental Neurobiology, Lleida's Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Catalonia, Spain
| |
Collapse
|
11
|
Garcia-Calero E, Puelles L. Histogenetic Radial Models as Aids to Understanding Complex Brain Structures: The Amygdalar Radial Model as a Recent Example. Front Neuroanat 2020; 14:590011. [PMID: 33240050 PMCID: PMC7683391 DOI: 10.3389/fnana.2020.590011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/05/2020] [Indexed: 01/17/2023] Open
Abstract
The radial dimension expands during central nervous system development after the proliferative neuroepithelium is molecularly patterned. The process is associated with neurogenesis, radial glia scaffolding, and migration of immature neurons into the developing mantle stratum. Radial histogenetic units, defined as a delimited neural polyclone whose cells share the same molecular profile, are molded during these processes, and usually become roughly stratified into periventricular, intermediate, and superficial (subpial) strata wherein neuronal cell types may differ and be distributed in various patterns. Cell-cell adhesion or repulsion phenomena together with interaction with local intercellular matrix cues regulate the acquisition of nuclear, reticular, or layer histogenetic forms in such strata. Finally, the progressive addition of inputs and outputs soon follows the purely neurogenetic and radial migratory phase. Frequently there is heterochrony in the radial development of adjacent histogenetic units, apart of peculiarities in differentiation due to non-shared aspects of the respective molecular profiles. Tangential migrations may add complexity to radial unit cytoarchitecture and function. The study of the contributions of such genetically controlled radial histogenetic units to the emerging complex neural structure is a key instrument to understand central nervous system morphology and function. One recent example in this scenario is the recently proposed radial model of the mouse pallial amygdala. This is theoretically valid generally in mammals (Garcia-Calero et al., 2020), and subdivides the nuclear complex of the pallial amygdala into five main radial units. The approach applies a novel ad hoc amygdalar section plane, given the observed obliquity of the amygdalar radial glial framework. The general relevance of radial unit studies for clarifying structural analysis of all complex brain regions such as the pallial amygdala is discussed, with additional examples.
Collapse
Affiliation(s)
- Elena Garcia-Calero
- Department of Human Anatomy, School of Medicine and IMIB-Arrixaca Institute, University of Murcia, Murcia, Spain
| | | |
Collapse
|
12
|
Diab A, Qi J, Shahin I, Milligan C, Fawcett JP. NCK1 Regulates Amygdala Activity to Control Context-dependent Stress Responses and Anxiety in Male Mice. Neuroscience 2020; 448:107-125. [PMID: 32946951 DOI: 10.1016/j.neuroscience.2020.09.026] [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: 04/16/2020] [Revised: 08/20/2020] [Accepted: 09/08/2020] [Indexed: 10/23/2022]
Abstract
Anxiety disorder (AD) is characterized by the development of maladaptive neuronal circuits and changes to the excitatory/inhibitory (E/I) balance of the central nervous system. Although AD is considered to be heritable, specific genetic markers remain elusive. Recent genome-wide association studies (GWAS) studies have identified non-catalytic region of tyrosine kinase adaptor protein 1 (NCK1), a gene that codes for an intracellular adaptor protein involved in actin dynamics, as an important gene in the regulation of mood. Using a murine model in which NCK1 is inactivated, we show that male, but not female, mice display increased levels of context-dependent anxiety-like behaviors along with an increase in circulating serum corticosterone relative to control. Treatment of male NCK1 mutant mice with a positive allosteric modulator of the GABAA receptor rescued the anxiety-like behaviors implicating NCK1 in regulating neuronal excitability. These defects are not attributable to apparent defects in gross brain structure or in axon guidance. However, when challenged in an approach-avoidance conflict paradigm, male NCK1-deficient mice have decreased neuronal activation in the prefrontal cortex (PFC), as well as decreased activation of inhibitory interneurons in the basolateral amygdala (BLA). Finally, NCK1 deficiency results in loss of dendritic spine density in principal neurons of the BLA. Taken together, these data implicate NCK1 in the control of E/I balance in BLA. Our work identifies a novel role for NCK1 in the regulation of sex-specific neuronal circuitry necessary for controlling anxiety-like behaviors. Further, our work points to this animal model as a useful preclinical tool for the study of novel anxiolytics and its significance towards understanding sex differences in anxiolytic function.
Collapse
Affiliation(s)
- Antonios Diab
- Department of Pharmacology, Dalhousie University, Canada
| | - Jiansong Qi
- Department of Pharmacology, Dalhousie University, Canada
| | - Ibrahim Shahin
- Department of Pharmacology, Dalhousie University, Canada
| | | | - James P Fawcett
- Department of Pharmacology, Dalhousie University, Canada; Department of Surgery, Dalhousie University, Canada.
| |
Collapse
|
13
|
Diaz MR, Johnson JM, Varlinskaya EI. Increased ethanol intake is associated with social anxiety in offspring exposed to ethanol on gestational day 12. Behav Brain Res 2020; 393:112766. [PMID: 32535179 DOI: 10.1016/j.bbr.2020.112766] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 06/05/2020] [Accepted: 06/07/2020] [Indexed: 01/21/2023]
Abstract
Prenatal alcohol exposure (PAE) can result in physical, cognitive, and neurological deficits termed Fetal Alcohol Spectrum Disorder (FASD). Deficits in social functioning associated with PAE are frequently observed and persist throughout the lifespan. Social impairments, such as social anxiety, are associated with increased alcohol abuse, which is also highly pervasive following PAE. Yet, the relationship between PAE-induced social alterations and alcohol intake later in life is not well understood. In order to test this relationship, we exposed pregnant female Sprague Dawley rats to a single instance of PAE on gestational day 12, a period of substantial neural development, and tested offspring in adulthood (postnatal day 63) in a modified social interaction test followed by alternating alone and social ethanol intake sessions. Consistent with our previous findings, we found that, in general, PAE reduced social preference (measure of social anxiety-like behavior) in female but not male adults. However, ethanol intake was significantly higher in the PAE group regardless of sex. When dividing subjects according to level of social anxiety-like behavior (low, medium, or high), PAE males (under both drinking contexts) and control females (under the social drinking context) with a high social anxiety phenotype showed the highest level of ethanol intake. Taken together, these data indicate that PAE differentially affects the interactions between social anxiety, ethanol intake, and drinking context in males and females. These findings extend our understanding of the complexity and persistence of PAE's sex-dependent effects into adulthood.
Collapse
Affiliation(s)
- Marvin R Diaz
- Department of Psychology, Center for Development and Behavioral Neuroscience Binghamton University, Binghamton, NY13902, United States; Developmental Exposure Alcohol Research Center, Baltimore MD21201, Binghamton NY 13902, Syracuse NY13210, United States.
| | - Julia M Johnson
- Developmental Exposure Alcohol Research Center, Baltimore MD21201, Binghamton NY 13902, Syracuse NY13210, United States
| | - Elena I Varlinskaya
- Department of Psychology, Center for Development and Behavioral Neuroscience Binghamton University, Binghamton, NY13902, United States; Developmental Exposure Alcohol Research Center, Baltimore MD21201, Binghamton NY 13902, Syracuse NY13210, United States
| |
Collapse
|
14
|
DNA Methylation within the Amygdala Early in Life Increases Susceptibility for Depression and Anxiety Disorders. J Neurosci 2020; 39:8828-8830. [PMID: 31694977 DOI: 10.1523/jneurosci.0845-19.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 08/27/2019] [Accepted: 09/11/2019] [Indexed: 12/15/2022] Open
|
15
|
Herrero MJ, Velmeshev D, Hernandez-Pineda D, Sethi S, Sorrells S, Banerjee P, Sullivan C, Gupta AR, Kriegstein AR, Corbin JG. Identification of amygdala-expressed genes associated with autism spectrum disorder. Mol Autism 2020; 11:39. [PMID: 32460837 PMCID: PMC7251751 DOI: 10.1186/s13229-020-00346-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 05/10/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Studies of individuals with autism spectrum disorder (ASD) have revealed a strong multigenic basis with the identification of hundreds of ASD susceptibility genes. ASD is characterized by social deficits and a range of other phenotypes, implicating complex genetics and involvement of a variety of brain regions. However, how mutations and mis-expression of select gene sets are associated with the behavioral components of ASD remains unknown. We reasoned that for genes to be associated with ASD core behaviors they must be: (1) expressed in brain regions relevant to ASD social behaviors and (2) expressed during the ASD susceptible window of brain development. METHODS Focusing on the amygdala, a brain region whose dysfunction has been highly implicated in the social component of ASD, we mined publicly available gene expression databases to identify ASD-susceptibility genes expressed during human and mouse amygdala development. We found that a large cohort of known ASD susceptibility genes is expressed in the developing human and mouse amygdala. We further performed analysis of single-nucleus RNA-seq (snRNA-seq) data from microdissected amygdala tissue from five ASD and five control human postmortem brains ranging in age from 4 to 20 years to elucidate cell type specificity of amygdala-expressed genes and their dysregulation in ASD. RESULTS Our analyses revealed that of the high-ranking ASD susceptibility genes, 80 are expressed in both human and mouse amygdala during fetal to early postnatal stages of development. Our human snRNA-seq analyses revealed cohorts of genes with altered expression in the ASD amygdala postnatally, especially within excitatory neurons, with dysregulated expression of seven genes predicted from our datamining pipeline. LIMITATIONS We were limited by the ages for which we were able to obtain human tissue; therefore, the results from our datamining pipeline approach will require validation, to the extent possible, in human tissue from earlier developmental stages. CONCLUSIONS Our pipeline narrows down the number of amygdala-expressed genes possibly involved in the social pathophysiology of ASD. Our human single-nucleus gene expression analyses revealed that ASD is characterized by changes in gene expression in specific cell types in the early postnatal amygdala.
Collapse
Affiliation(s)
- Maria Jesus Herrero
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Dmitry Velmeshev
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California-San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California-San Francisco, San Francisco, CA, USA
| | - David Hernandez-Pineda
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Saarthak Sethi
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Shawn Sorrells
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California-San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California-San Francisco, San Francisco, CA, USA
- Present Address: Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Payal Banerjee
- Center for Genetic Medicine, Children's Research Institute, Children's National Hospital, Washington, DC, USA
| | - Catherine Sullivan
- Department of Pediatrics and Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Abha R Gupta
- Department of Pediatrics and Child Study Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Arnold R Kriegstein
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California-San Francisco, San Francisco, CA, USA.
- Department of Neurology, University of California-San Francisco, San Francisco, CA, USA.
| | - Joshua G Corbin
- Center for Neuroscience Research, Children's Research Institute, Children's National Hospital, Washington, DC, USA.
| |
Collapse
|
16
|
Daun KA, Fuchigami T, Koyama N, Maruta N, Ikenaka K, Hitoshi S. Early Maternal and Social Deprivation Expands Neural Stem Cell Population Size and Reduces Hippocampus/Amygdala-Dependent Fear Memory. Front Neurosci 2020; 14:22. [PMID: 32063832 PMCID: PMC7000530 DOI: 10.3389/fnins.2020.00022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 01/10/2020] [Indexed: 11/13/2022] Open
Abstract
Early life stress can exert detrimental or beneficial effects on neural development and postnatal behavior depending on the timing, duration, strength, and ability to control the stressors. In this study, we utilized a maternal and social deprivation (MSD) model to investigate the effects of early life stress on neural stem cells (NSCs) and neurogenesis in the adult brain. We found that MSD during the stress-hyporesponsive period (SHRP) (early-MSD), when corticosterone secretion is suppressed, increased the size of the NSC population, whereas the same stress beyond the SHRP abrogated these effects. Early-MSD enhanced neurogenesis not only in the dentate gyrus of the hippocampus, one of the classic neurogenic regions, but also in the amygdala. In addition, mice exposed to early-MSD exhibited a reduction in amygdala/hippocampus-dependent fear memory. These results suggest that animals exposed to early life stress during the SHRP have reinforced stress resilience to cope with perceived stressors to maintain a normal homeostatic state.
Collapse
Affiliation(s)
- Kenny Anak Daun
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, Japan
| | - Takahiro Fuchigami
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, Japan
| | - Natsu Koyama
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, Japan
| | - Noriko Maruta
- Department of Psychiatry, Health Center, Hitotsubashi University, Tokyo, Japan
| | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, School of Life Sciences, The Graduate University for Advanced Studies, Okazaki, Japan
| | - Seiji Hitoshi
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, Japan.,Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan.,Department of Physiological Sciences, School of Life Sciences, The Graduate University for Advanced Studies, Okazaki, Japan
| |
Collapse
|
17
|
Abstract
In utero electroporation is a rapid and powerful technique to study the development of many brain regions. This approach presents several advantages over other methods to study specific steps of brain development in vivo, from proliferation to synaptic integration. Here, we describe in detail the individual steps necessary to carry out the technique. We also highlight the variations that can be implemented to target different cerebral structures and to study specific steps of development.
Collapse
|
18
|
López JM, Morona R, Moreno N, Lozano D, Jiménez S, González A. Pax6 expression highlights regional organization in the adult brain of lungfishes, the closest living relatives of land vertebrates. J Comp Neurol 2019; 528:135-159. [PMID: 31299095 DOI: 10.1002/cne.24744] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/27/2019] [Accepted: 07/05/2019] [Indexed: 12/15/2022]
Abstract
The Pax6 gene encodes a regulatory transcription factor that is key in brain development. The molecular structure of Pax6, the roles it plays and its patterns of expression in the brain have been highly conserved during vertebrate evolution. As neurodevelopment proceeds, the Pax6 expression changes from the mitotic germinal zone in the ventricular zone to become distributed in cell groups in the adult brain. Studies in various vertebrates, from fish to mammals, found that the Pax6 expression is maintained in adults in most regions that express it during development. Specifically, in amphibians, Pax6 is widely expressed in the adult brain and its distribution pattern serves to highlight regional organization of the brain. In the present study, we analyzed the detailed distribution of Pax6 cells in the adult central nervous system of lungfishes, the closest living relatives of all tetrapods. Immunohistochemistry performed using double labeling techniques with several neuronal markers of known distribution patterns served to evaluate the actual location of Pax6 cells. Our results show that the Pax6 expression is maintained in the adult brain of lungfishes, in distinct regions of the telencephalon (pallium and subpallium), diencephalon, mesencephalon, hindbrain, spinal cord, and retina. The pattern of Pax6 expression is largely shared with amphibians and helps to understand the primitive condition that would have characterized the common ancestors to all sarcopterygians (lobe-finned fishes and tetrapods), in which Pax6 would be needed to maintain specific entities of subpopulations of neurons.
Collapse
Affiliation(s)
- Jesús M López
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| | - Ruth Morona
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| | - Nerea Moreno
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| | - Daniel Lozano
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| | - Sara Jiménez
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| | - Agustín González
- Department of Cell Biology, Faculty of Biology, University Complutense of Madrid, Madrid, Spain
| |
Collapse
|
19
|
Medina L, Abellán A, Desfilis E. Evolution of Pallial Areas and Networks Involved in Sociality: Comparison Between Mammals and Sauropsids. Front Physiol 2019; 10:894. [PMID: 31354528 PMCID: PMC6640085 DOI: 10.3389/fphys.2019.00894] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 06/27/2019] [Indexed: 11/19/2022] Open
Abstract
Birds are extremely interesting animals for studying the neurobiological basis of cognition and its evolution. They include species that are highly social and show high cognitive capabilities. Moreover, birds rely more on visual and auditory cues than on olfaction for social behavior and cognition, just like primates. In primates, there are two major brain networks associated to sociality: (1) one related to perception and decision-making, involving the pallial amygdala (with the basolateral complex as a major component), the temporal and temporoparietal neocortex, and the orbitofrontal cortex; (2) another one related to affiliation, including the medial extended amygdala, the ventromedial prefrontal and anterior cingulate cortices, the ventromedial striatum (largely nucleus accumbens), and the ventromedial hypothalamus. In this account, we used an evolutionary developmental neurobiology approach, in combination with published comparative connectivity and functional data, to identify areas and functional networks in the sauropsidian brain comparable to those of mammals that are related to decision-making and affiliation. Both in mammals and sauropsids, there is an important interaction between these networks by way of cross projections between areas of both systems.
Collapse
Affiliation(s)
- Loreta Medina
- Department of Experimental Medicine, Institut de Recerca Biomèdica de Lleida - Fundació Dr. Pifarré (IRBLleida), University of Lleida, Lleida, Spain
| | - Antonio Abellán
- Department of Experimental Medicine, Institut de Recerca Biomèdica de Lleida - Fundació Dr. Pifarré (IRBLleida), University of Lleida, Lleida, Spain
| | - Ester Desfilis
- Department of Experimental Medicine, Institut de Recerca Biomèdica de Lleida - Fundació Dr. Pifarré (IRBLleida), University of Lleida, Lleida, Spain
| |
Collapse
|
20
|
Martin-Lopez E, Xu C, Liberia T, Meller SJ, Greer CA. Embryonic and postnatal development of mouse olfactory tubercle. Mol Cell Neurosci 2019; 98:82-96. [PMID: 31200100 DOI: 10.1016/j.mcn.2019.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 05/09/2019] [Accepted: 06/10/2019] [Indexed: 02/06/2023] Open
Abstract
The olfactory tubercle (OT) is located in the ventral-medial region of the brain where it receives primary input from olfactory bulb (OB) projection neurons and processes olfactory behaviors related to motivation, hedonics of smell and sexual encounters. The OT is part of the dopamine reward system that shares characteristics with the striatum. Together with the nucleus accumbens, the OT has been referred to as the "ventral striatum". However, despite its functional importance little is known about the embryonic development of the OT and the phenotypic properties of the OT cells. Here, using thymidine analogs, we establish that mouse OT neurogenesis occurs predominantly between E11-E15 in a lateral-to-medial gradient. Then, using a piggyBac multicolor technique we characterized the migratory route of OT neuroblasts from their embryonic point of origin. Following neurogenesis in the ventral lateral ganglionic eminence (vLGE), neuroblasts destined for the OT followed a dorsal-ventral pathway we named "ventral migratory course" (VMC). Upon reaching the nascent OT, neurons established a prototypical laminar distribution that was determined, in part, by the progenitor cell of origin. A phenotypic analysis of OT neuroblasts using a single-color piggyBac technique, showed that OT shared the molecular specification of striatal neurons. In addition to primary afferent input from the OB, the OT also receives a robust dopaminergic input from ventral tegmentum (Ikemoto, 2007). We used tyrosine hydroxylase (TH) expression as a proxy for dopaminergic innervation and showed that TH onset occurs at E13 and progressively increased until postnatal stages following an 'inside-out' pattern. Postnatally, we established the myelination in the OT occurring between P7 and P14, as shown with CNPase staining, and we characterized the cellular phenotypes populating the OT by immunohistochemistry. Collectively, this work provides the first detailed analysis of the developmental and maturation processes occurring in mouse OT, and demonstrates the striatal nature of the OT as part of the ventral striatum (vST).
Collapse
Affiliation(s)
- Eduardo Martin-Lopez
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Christine Xu
- Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Teresa Liberia
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Sarah J Meller
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; The Interdepartmental Neuroscience Graduate Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Charles A Greer
- Department of Neuroscience, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; Department of Neurosurgery, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA; The Interdepartmental Neuroscience Graduate Program, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| |
Collapse
|
21
|
The role of Pax6 in brain development and its impact on pathogenesis of autism spectrum disorder. Brain Res 2019; 1705:95-103. [DOI: 10.1016/j.brainres.2018.02.041] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/23/2018] [Accepted: 02/24/2018] [Indexed: 12/14/2022]
|
22
|
Kawasaki H. Molecular Investigations of the Development and Diseases of Cerebral Cortex Folding using Gyrencephalic Mammal Ferrets. Biol Pharm Bull 2018; 41:1324-1329. [DOI: 10.1248/bpb.b18-00142] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University
| |
Collapse
|
23
|
Cwetsch AW, Pinto B, Savardi A, Cancedda L. In vivo methods for acute modulation of gene expression in the central nervous system. Prog Neurobiol 2018; 168:69-85. [PMID: 29694844 PMCID: PMC6080705 DOI: 10.1016/j.pneurobio.2018.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 04/17/2018] [Accepted: 04/20/2018] [Indexed: 12/17/2022]
Abstract
Accurate and timely expression of specific genes guarantees the healthy development and function of the brain. Indeed, variations in the correct amount or timing of gene expression lead to improper development and/or pathological conditions. Almost forty years after the first successful gene transfection in in vitro cell cultures, it is currently possible to regulate gene expression in an area-specific manner at any step of central nervous system development and in adulthood in experimental animals in vivo, even overcoming the very poor accessibility of the brain. Here, we will review the diverse approaches for acute gene transfer in vivo, highlighting their advantages and disadvantages with respect to the efficiency and specificity of transfection as well as to brain accessibility. In particular, we will present well-established chemical, physical and virus-based approaches suitable for different animal models, pointing out their current and future possible applications in basic and translational research as well as in gene therapy.
Collapse
Affiliation(s)
- Andrzej W Cwetsch
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Università degli Studi di Genova, Via Balbi, 5, 16126 Genova, Italy
| | - Bruno Pinto
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126, Pisa, Italy
| | - Annalisa Savardi
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Università degli Studi di Genova, Via Balbi, 5, 16126 Genova, Italy
| | - Laura Cancedda
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; DulbeccoTelethon Institute, Italy.
| |
Collapse
|
24
|
Sato M, Ohtsuka M, Nakamura S, Sakurai T, Watanabe S, Gurumurthy CB. In vivo genome editing targeted towards the female reproductive system. Arch Pharm Res 2018; 41:898-910. [PMID: 29974342 DOI: 10.1007/s12272-018-1053-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 06/27/2018] [Indexed: 11/30/2022]
Abstract
The discovery of sequence specific nucleases such as ZFNs, TALENs, and CRISPR/Cas9 has revolutionized genome editing. The CRISPR/Cas9 system has particularly emerged as a highly simple and efficient approach towards generating genome-edited animal models of most of the experimental species. The limitation of these novel genome editing tools is that, till date, they depend on traditional pronuclear injection (PI)-based transgenic technologies developed over the last three decades. PI requires expensive micromanipulator systems and the equipment operators must possess a high level of skill. Therefore, since the establishment of PI-based transgenesis, various research groups worldwide have attempted to develop alternative and simple gene delivery methods. However, owing to the failure of chromosomal integration of the transgene, none of these methods gained the level of confidence as that by the PI method in order to be adapted as a routine approach. The recently developed genome editing systems do not require complicated techniques. Therefore, presently, attention is being focused on non-PI-based gene delivery into germ cells for simple and rapid production of genetically engineered animals. For example, a few reports during the previous 1-2 years demonstrated the use of electroporation (EP) in isolated zygotes that helped to overcome the absolute dependency on PI techniques. Recently, another breakthrough technology called genome editing via oviductal nucleic acids delivery (GONAD) that directly delivers nucleic acids into zygotes within the oviducts in situ was developed. This technology completely relieves the bottlenecks of animal transgenesis as it does not require PI and ex vivo handling of embryos. This review discusses in detail the in vivo gene delivery methods targeted towards female reproductive tissues as these methods that have been developed over the past 2-3 decades can now be re-evaluated for their suitability to deliver the CRISPR/Cas9 components to produce transgenic animals. This review also provides an overview of the latest advances in CRISPR-enabled delivery technologies that have caused paradigm shifts in animal transgenesis methodologies.
Collapse
Affiliation(s)
- Masahiro Sato
- Section of Gene Expression Regulation, Frontier Science Research Center, Kagoshima University, Kagoshima, 890-8544, Japan.
| | - Masato Ohtsuka
- Division of Basic Medical Science and Molecular Medicine, School of Medicine, Tokai University, Kanagawa, 259-1193, Japan.,Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, Kanagawa, 259-1193, Japan.,The Institute of Medical Sciences, Tokai University, Kanagawa, 259-1193, Japan
| | - Shingo Nakamura
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Saitama, 359-8513, Japan
| | - Takayuki Sakurai
- Department of Cardiovascular Research, Graduate School of Medicine, Shinshu University, Nagano, 390-8621, Japan.,Basic Research Division for Next-Generation Disease Models and Fundamental Technology, Research Center for Next Generation Medicine, Shinshu University, Nagano, 390-8621, Japan
| | - Satoshi Watanabe
- Animal Genome Research Unit, Division of Animal Science, National Institute of Agrobiological Sciences, Ibaraki, 305-8602, Japan
| | - Channabasavaiah B Gurumurthy
- Mouse Genome Engineering Core Facility, University of Nebraska Medical Center, Omaha, NE, 68198, USA.,Developmental Neuroscience, Munro Meyer Institute for Genetics and Rehabilitation, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| |
Collapse
|
25
|
Ruiz-Reig N, Andres B, Lamonerie T, Theil T, Fairén A, Studer M. The caudo-ventral pallium is a novel pallial domain expressing Gdf10 and generating Ebf3-positive neurons of the medial amygdala. Brain Struct Funct 2018; 223:3279-3295. [PMID: 29869132 DOI: 10.1007/s00429-018-1687-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 05/18/2018] [Indexed: 12/16/2022]
Abstract
In rodents, the medial nucleus of the amygdala receives direct inputs from the accessory olfactory bulbs and is mainly implicated in pheromone-mediated reproductive and defensive behaviors. The principal neurons of the medial amygdala are GABAergic neurons generated principally in the caudo-ventral medial ganglionic eminence and preoptic area. Beside GABAergic neurons, the medial amygdala also contains glutamatergic Otp-expressing neurons cells generated in the lateral hypothalamic neuroepithelium and a non-well characterized Pax6-positive population. In the present work, we describe a novel glutamatergic Ebf3-expressing neuronal subpopulation distributed within the periphery of the postero-ventral medial amygdala. These neurons are generated in a pallial domain characterized by high expression of Gdf10. This territory is topologically the most caudal tier of the ventral pallium and accordingly, we named it Caudo-Ventral Pallium (CVP). In the absence of Pax6, the CVP is disrupted and Ebf3-expressing neurons fail to be generated. Overall, this work proposes a novel model of the neuronal composition of the medial amygdala and unravels for the first time a new novel pallial subpopulation originating from the CVP and expressing the transcription factor Ebf3.
Collapse
Affiliation(s)
- Nuria Ruiz-Reig
- Université Côte d'Azur (UCA), CNRS, Inserm, Institut de Biologie Valrose (iBV), 06108, Nice, France.
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), 03550, San Juan de Alicante, Spain.
| | - Belen Andres
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), 03550, San Juan de Alicante, Spain
| | - Thomas Lamonerie
- Université Côte d'Azur (UCA), CNRS, Inserm, Institut de Biologie Valrose (iBV), 06108, Nice, France
| | - Thomas Theil
- Centre for Discovery Brain Sciences, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Alfonso Fairén
- Instituto de Neurociencias (Consejo Superior de Investigaciones Científicas-Universidad Miguel Hernández, CSIC-UMH), 03550, San Juan de Alicante, Spain
- , Palau 11, 03550, San Juan de Alicante, Spain
| | - Michèle Studer
- Université Côte d'Azur (UCA), CNRS, Inserm, Institut de Biologie Valrose (iBV), 06108, Nice, France.
| |
Collapse
|
26
|
Ruiz-Reig N, Studer M. Rostro-Caudal and Caudo-Rostral Migrations in the Telencephalon: Going Forward or Backward? Front Neurosci 2017; 11:692. [PMID: 29311773 PMCID: PMC5742585 DOI: 10.3389/fnins.2017.00692] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/23/2017] [Indexed: 11/13/2022] Open
Abstract
The generation and differentiation of an appropriate number of neurons, as well as its distribution in different parts of the brain, is crucial for the proper establishment, maintenance and plasticity of neural circuitries. Newborn neurons travel along the brain in a process known as neuronal migration, to finalize their correct position in the nervous system. Defects in neuronal migration produce abnormalities in the brain that can generate neurodevelopmental pathologies, such as autism, schizophrenia and intellectual disability. In this review, we present an overview of the developmental origin of the different telencephalic subdivisions and a description of migratory pathways taken by distinct neural populations traveling long distances before reaching their target position in the brain. In addition, we discuss some of the molecules implicated in the guidance of these migratory paths and transcription factors that contribute to the correct migration and integration of these neurons.
Collapse
|
27
|
Rouzer SK, Cole JM, Johnson JM, Varlinskaya EI, Diaz MR. Moderate Maternal Alcohol Exposure on Gestational Day 12 Impacts Anxiety-Like Behavior in Offspring. Front Behav Neurosci 2017; 11:183. [PMID: 29033803 PMCID: PMC5626811 DOI: 10.3389/fnbeh.2017.00183] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 09/14/2017] [Indexed: 12/29/2022] Open
Abstract
Among the numerous consequences of prenatal alcohol exposure (PAE) is an increase in anxiety-like behavior that can prove debilitating to daily functioning. A significant body of literature has linked gestational day 12 (G12) heavy ethanol exposure with social anxiety, evident in adolescent males and females. However, the association between non-social anxiety-like behavior and moderate alcohol exposure, a more common pattern of drinking in pregnant women, is yet unidentified. To model moderate PAE (mPAE), we exposed pregnant Sprague-Dawley rats to either room air or vaporized ethanol for 6 h on G12. Adolescent offspring were then tested on postnatal days (P) 41-47 in one of the following four anxiety assays: novelty-induced hypophagia (NIH), elevated plus maze (EPM), light-dark box (LDB) and open-field (OF). Our findings revealed significant increases in measures of anxiety-like behavior in male PAE offspring in the NIH, LDB and OF, with no differences observed in females on any test. Additionally, male offspring who demonstrated heightened anxiety-like behavior as adolescents demonstrated decreased anxiety-like behavior in adulthood, as measured by a marble-burying test (MBT), while females continued to be unaffected in adulthood. These results suggest that mPAE leads to dynamic changes in anxiety-like behavior exclusively in male offspring.
Collapse
Affiliation(s)
- Siara K Rouzer
- Department of Psychology, Center for Development and Behavioral Neuroscience, Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY, United States
| | - Jesse M Cole
- Department of Psychology, Center for Development and Behavioral Neuroscience, Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY, United States
| | - Julia M Johnson
- Department of Psychology, Center for Development and Behavioral Neuroscience, Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY, United States
| | - Elena I Varlinskaya
- Department of Psychology, Center for Development and Behavioral Neuroscience, Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY, United States
| | - Marvin R Diaz
- Department of Psychology, Center for Development and Behavioral Neuroscience, Developmental Exposure Alcohol Research Center, Binghamton University, Binghamton, NY, United States
| |
Collapse
|
28
|
Lischinsky JE, Sokolowski K, Li P, Esumi S, Kamal Y, Goodrich M, Oboti L, Hammond TR, Krishnamoorthy M, Feldman D, Huntsman M, Liu J, Corbin JG. Embryonic transcription factor expression in mice predicts medial amygdala neuronal identity and sex-specific responses to innate behavioral cues. eLife 2017; 6. [PMID: 28244870 PMCID: PMC5384829 DOI: 10.7554/elife.21012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 02/26/2017] [Indexed: 11/14/2022] Open
Abstract
The medial subnucleus of the amygdala (MeA) plays a central role in processing sensory cues required for innate behaviors. However, whether there is a link between developmental programs and the emergence of inborn behaviors remains unknown. Our previous studies revealed that the telencephalic preoptic area (POA) embryonic niche is a novel source of MeA destined progenitors. Here, we show that the POA is comprised of distinct progenitor pools complementarily marked by the transcription factors Dbx1 and Foxp2. As determined by molecular and electrophysiological criteria this embryonic parcellation predicts postnatal MeA inhibitory neuronal subtype identity. We further find that Dbx1-derived and Foxp2+ cells in the MeA are differentially activated in response to innate behavioral cues in a sex-specific manner. Thus, developmental transcription factor expression is predictive of MeA neuronal identity and sex-specific neuronal responses, providing a potential developmental logic for how innate behaviors could be processed by different MeA neuronal subtypes. DOI:http://dx.doi.org/10.7554/eLife.21012.001 Within the brain, a set of interconnected structures called the limbic system is involved in emotion, motivation and memory. This system – and in particular a structure called the medial amygdala – also contributes to behavioral drives that help an animal to survive and reproduce. These include the drive to avoid predators, to defend territory, and to find a mate. Such behaviors are thought to be inborn or innate. This means that animals display them instinctively whenever specific triggers are present, without the need to learn them beforehand. However, just as a computer must be programmed to perform specific tasks, these innate behavioral responses must also be programmed into the brain. Given that animals do not learn these behaviors, Lischinsky et al. reasoned that specific events during the development of the brain must provide the animal’s brain with the necessary instructions. To test this idea, they studied how the development of the medial amygdala in mouse embryos may give rise to differences in innate mating behavior seen between male and female mice. The medial amygdala contains many subtypes of neurons, which show different responses to sex hormones such as estrogen and androgen. Lischinsky et al. show that two sets of cells give rise to some of the different neurons of the adult medial amygdala. One set of these precursor cells makes a protein called Dbx1 and the other makes a protein called Foxp2. These two sets of precursors generate medial amygdala neurons with different arrays of sex hormone receptors in male and female mice. Moreover, while the two sets of medial amygdala neurons are activated during aggressive encounters, they show different patterns of activation in male and female animals during mating. These findings suggest that the development of Dbx1-derived and Foxp2+ neurons in the medial amygdala helps program innate reproductive and aggressive behaviors into the brain. The new findings also provide insights into why these behaviors differ in male and female mice. The next challenge is to identify the inputs and outputs of these two distinct subpopulations of medial amygdala neurons. This should make it possible to work out exactly how these populations of cells control innate behaviors in male and female animals. DOI:http://dx.doi.org/10.7554/eLife.21012.002
Collapse
Affiliation(s)
- Julieta E Lischinsky
- Institute for Biomedical Sciences, The George Washington University, Washington DC, United States.,Center for Neuroscience Research, Children's National Medical Center, Washington, DC, United States
| | - Katie Sokolowski
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, United States
| | - Peijun Li
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, United States
| | - Shigeyuki Esumi
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, United States.,Graduate School of Medical Sciences, Kumamoto-University, Kumamoto City, Japan
| | - Yasmin Kamal
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, United States
| | - Meredith Goodrich
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, United States
| | - Livio Oboti
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, United States
| | - Timothy R Hammond
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, United States
| | - Meera Krishnamoorthy
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, United States
| | - Daniel Feldman
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, United States
| | - Molly Huntsman
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado
| | - Judy Liu
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, United States
| | - Joshua G Corbin
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC, United States
| |
Collapse
|
29
|
KAWASAKI H. Molecular investigations of development and diseases of the brain of higher mammals using the ferret. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2017; 93:259-269. [PMID: 28496051 PMCID: PMC5489433 DOI: 10.2183/pjab.93.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 02/14/2017] [Indexed: 06/07/2023]
Abstract
The brains of higher mammals such as primates and carnivores contain well-developed unique brain structures. Uncovering the physiological functions, developmental mechanisms and evolution of these brain structures would greatly facilitate our understanding of the human brain and its diseases. Although the anatomical and electrophysiological features of these brain structures have been intensively investigated, our knowledge about their molecular bases is still limited. To overcome this limitation, genetic techniques for the brains of carnivores and primates have been established, and molecules whose expression patterns correspond to these brain structures were identified recently. To investigate the functional roles of these molecules, rapid and efficient genetic manipulation methods for higher mammals have been explored. In this review, recent advances in molecular investigations of the brains of higher mammals are discussed, mainly focusing on ferrets (Mustela putorius furo).
Collapse
Affiliation(s)
- Hiroshi KAWASAKI
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Ishikawa, Japan
- Brain/Liver Interface Medicine Research Center, Kanazawa University, Ishikawa, Japan
| |
Collapse
|
30
|
Luo W, Mizuno H, Iwata R, Nakazawa S, Yasuda K, Itohara S, Iwasato T. Supernova: A Versatile Vector System for Single-Cell Labeling and Gene Function Studies in vivo. Sci Rep 2016; 6:35747. [PMID: 27775045 PMCID: PMC5075795 DOI: 10.1038/srep35747] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/30/2016] [Indexed: 11/29/2022] Open
Abstract
Here we describe “Supernova” series of vector systems that enable single-cell labeling and labeled cell-specific gene manipulation, when introduced by in utero electroporation (IUE) or adeno-associated virus (AAV)-mediated gene delivery. In Supernova, sparse labeling relies on low TRE leakage. In a small population of cells with over-threshold leakage, initial tTA-independent weak expression is enhanced by tTA/TRE-positive feedback along with a site-specific recombination system (e.g., Cre/loxP, Flpe/FRT). Sparse and bright labeling by Supernova with little background enables the visualization of the morphological details of individual neurons in densely packed brain areas such as the cortex and hippocampus, both during development and in adulthood. Sparseness levels are adjustable. Labeled cell-specific gene knockout was accomplished by introducing Cre/loxP-based Supernova vectors into floxed mice. Furthermore, by combining with RNAi, TALEN, and CRISPR/Cas9 technologies, IUE-based Supernova achieved labeled cell-specific gene knockdown and editing/knockout without requiring genetically altered mice. Thus, Supernova system is highly extensible and widely applicable for single-cell analyses in complex organs, such as the mammalian brain.
Collapse
Affiliation(s)
- Wenshu Luo
- Division of Neurogenetics, National Institute of Genetics, Mishima, 411-8540, Japan.,Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, 411-8540, Japan
| | - Hidenobu Mizuno
- Division of Neurogenetics, National Institute of Genetics, Mishima, 411-8540, Japan.,Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, 411-8540, Japan
| | - Ryohei Iwata
- Division of Neurogenetics, National Institute of Genetics, Mishima, 411-8540, Japan.,Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, 411-8540, Japan
| | - Shingo Nakazawa
- Division of Neurogenetics, National Institute of Genetics, Mishima, 411-8540, Japan.,Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, 411-8540, Japan
| | - Kosuke Yasuda
- Laboratory for Behavioral Genetics, RIKEN Brain Science Institute, Wako, 351-0198, Japan
| | - Shigeyoshi Itohara
- Laboratory for Behavioral Genetics, RIKEN Brain Science Institute, Wako, 351-0198, Japan
| | - Takuji Iwasato
- Division of Neurogenetics, National Institute of Genetics, Mishima, 411-8540, Japan.,Department of Genetics, SOKENDAI (The Graduate University for Advanced Studies), Mishima, 411-8540, Japan
| |
Collapse
|
31
|
Huilgol D, Tole S. Cell migration in the developing rodent olfactory system. Cell Mol Life Sci 2016; 73:2467-90. [PMID: 26994098 PMCID: PMC4894936 DOI: 10.1007/s00018-016-2172-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 02/08/2016] [Accepted: 03/01/2016] [Indexed: 02/06/2023]
Abstract
The components of the nervous system are assembled in development by the process of cell migration. Although the principles of cell migration are conserved throughout the brain, different subsystems may predominantly utilize specific migratory mechanisms, or may display unusual features during migration. Examining these subsystems offers not only the potential for insights into the development of the system, but may also help in understanding disorders arising from aberrant cell migration. The olfactory system is an ancient sensory circuit that is essential for the survival and reproduction of a species. The organization of this circuit displays many evolutionarily conserved features in vertebrates, including molecular mechanisms and complex migratory pathways. In this review, we describe the elaborate migrations that populate each component of the olfactory system in rodents and compare them with those described in the well-studied neocortex. Understanding how the components of the olfactory system are assembled will not only shed light on the etiology of olfactory and sexual disorders, but will also offer insights into how conserved migratory mechanisms may have shaped the evolution of the brain.
Collapse
Affiliation(s)
- Dhananjay Huilgol
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
- Cold Spring Harbor Laboratory, Cold Spring Harbor, USA
| | - Shubha Tole
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India.
| |
Collapse
|
32
|
Mikuni T, Nishiyama J, Sun Y, Kamasawa N, Yasuda R. High-Throughput, High-Resolution Mapping of Protein Localization in Mammalian Brain by In Vivo Genome Editing. Cell 2016; 165:1803-1817. [PMID: 27180908 DOI: 10.1016/j.cell.2016.04.044] [Citation(s) in RCA: 152] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/14/2016] [Accepted: 04/13/2016] [Indexed: 12/25/2022]
Abstract
A scalable and high-throughput method to identify precise subcellular localization of endogenous proteins is essential for integrative understanding of a cell at the molecular level. Here, we developed a simple and generalizable technique to image endogenous proteins with high specificity, resolution, and contrast in single cells in mammalian brain tissue. The technique, single-cell labeling of endogenous proteins by clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9-mediated homology-directed repair (SLENDR), uses in vivo genome editing to insert a sequence encoding an epitope tag or a fluorescent protein to a gene of interest by CRISPR-Cas9-mediated homology-directed repair (HDR). Single-cell, HDR-mediated genome editing was achieved by delivering the editing machinery to dividing neuronal progenitors through in utero electroporation. We demonstrate that SLENDR allows rapid determination of the localization and dynamics of many endogenous proteins in various cell types, regions, and ages in the brain. Thus, SLENDR provides a high-throughput platform to map the subcellular localization of endogenous proteins with the resolution of micro- to nanometers in the brain.
Collapse
Affiliation(s)
- Takayasu Mikuni
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Jun Nishiyama
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA.
| | - Ye Sun
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA; Integrative Program in Biology and Neuroscience, Florida Atlantic University, Jupiter, FL 33458, USA
| | - Naomi Kamasawa
- Electron Microscopy Core Facility, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA
| | - Ryohei Yasuda
- Neuronal Signal Transduction Group, Max Planck Florida Institute for Neuroscience, Jupiter, FL 33458, USA.
| |
Collapse
|
33
|
Vicario A, Mendoza E, Abellán A, Scharff C, Medina L. Genoarchitecture of the extended amygdala in zebra finch, and expression of FoxP2 in cell corridors of different genetic profile. Brain Struct Funct 2016; 222:481-514. [PMID: 27160258 PMCID: PMC5225162 DOI: 10.1007/s00429-016-1229-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 04/21/2016] [Indexed: 02/01/2023]
Abstract
We used a battery of genes encoding transcription factors (Pax6, Islet1, Nkx2.1, Lhx6, Lhx5, Lhx9, FoxP2) and neuropeptides to study the extended amygdala in developing zebra finches. We identified different components of the central extended amygdala comparable to those found in mice and chickens, including the intercalated amygdalar cells, the central amygdala, and the lateral bed nucleus of the stria terminalis. Many cells likely originate in the dorsal striatal domain, ventral striatal domain, or the pallidal domain, as is the case in mice and chickens. Moreover, a cell subpopulation of the central extended amygdala appears to originate in the prethalamic eminence. As a general principle, these different cells with specific genetic profiles and embryonic origin form separate or partially intermingled cell corridors along the extended amygdala, which may be involved in different functional pathways. In addition, we identified the medial amygdala of the zebra finch. Like in the chickens and mice, it is located in the subpallium and is rich in cells of pallido-preoptic origin, containing minor subpopulations of immigrant cells from the ventral pallium, alar hypothalamus and prethalamic eminence. We also proposed that the medial bed nucleus of the stria terminalis is composed of several parallel cell corridors with different genetic profile and embryonic origin: preoptic, pallidal, hypothalamic, and prethalamic. Several of these cell corridors with distinct origin express FoxP2, a transcription factor implicated in synaptic plasticity. Our results pave the way for studies using zebra finches to understand the neural basis of social behavior, in which the extended amygdala is involved.
Collapse
Affiliation(s)
- Alba Vicario
- Laboratory of Brain Development and Evolution, Department of Experimental Medicine, Faculty of Medicine, University of Lleida, Institute of Biomedical Research of Lleida (IRBLleida), Avda. Alcalde Rovira Roure 80, Catalunya, 25198, Lleida, Spain
| | | | - Antonio Abellán
- Laboratory of Brain Development and Evolution, Department of Experimental Medicine, Faculty of Medicine, University of Lleida, Institute of Biomedical Research of Lleida (IRBLleida), Avda. Alcalde Rovira Roure 80, Catalunya, 25198, Lleida, Spain
| | | | - Loreta Medina
- Laboratory of Brain Development and Evolution, Department of Experimental Medicine, Faculty of Medicine, University of Lleida, Institute of Biomedical Research of Lleida (IRBLleida), Avda. Alcalde Rovira Roure 80, Catalunya, 25198, Lleida, Spain.
| |
Collapse
|
34
|
Abstract
UNLABELLED Human mutations in ZIC2 have been identified in patients with holoprosencephaly and schizophrenia. Similarly, Zic2 mutant mice exhibit holoprosencephaly in homozygosis and behavioral and morphological schizophrenic phenotypes associated with forebrain defects in heterozygosis. Despite the devastating effects of mutations in Zic2, the cellular and molecular mechanisms that provoke Zic2-deficiency phenotypes are yet unclear. Here, we report a novel role for this transcription factor in the migration of three different types of forebrain neurons: the Cajal-Retzius cells that populate the surface of the telencephalic vesicles, an amygdaloid group of cells originated in the caudal pole of the telencephalic pallium, and a cell population that travels from the prethalamic neuroepithelium to the ventral lateral geniculate nucleus. Our results also suggest that the receptor EphB1, previously identified as a Zic2 target, may mediate, at least partially, Zic2-dependent migratory events. According to these results, we propose that deficiencies in cell motility and guidance contribute to most of the forebrain pathologies associated with Zic2 mutations. SIGNIFICANCE STATEMENT Although the phenotype of Zic2 mutant individuals was reported more than 10 years ago, until now, the main function of this transcription factor during early development has not been precisely defined. Here, we reveal a previously unknown role for Zic2 in the migration of forebrain neurons such as Cajal-Retzius cells, interneurons moving to the ventral lateral geniculate nucleus, and neocortical cells going to the amygdala. We believe that the role of this transcription factor in certain populations of migratory cells contributes to defects in cortical layering and hypocellularity in the ventral LGN and amygdala and will contribute to our understanding of the devastating phenotypes associated with Zic2 mutations in both humans and mice.
Collapse
|
35
|
The COUP-TFII/Neuropilin-2 is a molecular switch steering diencephalon-derived GABAergic neurons in the developing mouse brain. Proc Natl Acad Sci U S A 2015; 112:E4985-94. [PMID: 26305926 DOI: 10.1073/pnas.1420701112] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
The preoptic area (POa) of the rostral diencephalon supplies the neocortex and the amygdala with GABAergic neurons in the developing mouse brain. However, the molecular mechanisms that determine the pathway and destinations of POa-derived neurons have not yet been identified. Here we show that Chicken ovalbumin upstream promoter transcription factor II (COUP-TFII)-induced expression of Neuropilin-2 (Nrp2) and its down-regulation control the destination of POa-derived GABAergic neurons. Initially, a majority of the POa-derived migrating neurons express COUP-TFII and form a caudal migratory stream toward the caudal subpallium. When a subpopulation of cells steers toward the neocortex, they exhibit decreased expression of COUP-TFII and Nrp2. The present findings show that suppression of COUP-TFII/Nrp2 changed the destination of the cells into the neocortex, whereas overexpression of COUP-TFII/Nrp2 caused cells to end up in the medial part of the amygdala. Taken together, these results reveal that COUP-TFII/Nrp2 is a molecular switch determining the pathway and destination of migrating GABAergic neurons born in the POa.
Collapse
|
36
|
Pombero A, Martinez S. The α2-subunit of the nicotinic cholinergic receptor is specifically expressed in medial subpallium-derived cells of mammalian amygdala. J Comp Neurol 2015; 523:1608-21. [PMID: 25641263 DOI: 10.1002/cne.23754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 01/22/2015] [Accepted: 01/25/2015] [Indexed: 02/03/2023]
Abstract
Nicotinic acetylcholine receptor (nAChR) subtypes are expressed in specific neuronal populations, which are involved in numerous neural functions such as sleep, fatigue, anxiety, and cognition, as well as the central processing of pain and food intake. Moreover, mutations in nAChRs subunits have been related to frontal lobe epilepsy, neurodegenerative diseases, and other neurological disorders, including schizophrenia and attention deficit and hyperactivity disorder (ADHD). Previous studies have shown that the α2-subunit of the AChR (Chrna2) is expressed in the basal forebrain, in the septum, and in some amygdalar nuclei in the adult rodent brain. However, although the importance of this amygdalar expression in emotion-related behavior and the physiopathology of neuropsychiatric disorders has been accepted, a detailed study of the Chrna2 expression pattern during development has been lacking. In this study we found that Chrna2 is specifically expressed in medial subpallium-derived amygdalar nuclei from early developmental stages to adult. This finding could help us to better understand the role of Chrna2 in the differentiation and functional maturation of amygdalar neurons involved in cholinergic-regulated emotional behavior.
Collapse
Affiliation(s)
- Ana Pombero
- Neurosciences Institute, University Miguel Hernandez-Spanish National Research Council (CSIC), San Juan de Alicante, 03550, Spain
| | - Salvador Martinez
- Neurosciences Institute, University Miguel Hernandez-Spanish National Research Council (CSIC), San Juan de Alicante, 03550, Spain.,Murcia Biomedical Research Institute (IMIB)-Arraixaca, University of Murcia, 30071, Spain.,Biomedical Research Center, Mental Healthe Network (CIBERSAM), Institute of Health Carlos III, Madrid, 28029, Spain
| |
Collapse
|
37
|
Jahan E, Rafiq AM, Otani H. In utero and exo utero fetal surgery on histogenesis of organs in animals. World J Surg Proced 2015; 5:198-207. [DOI: 10.5412/wjsp.v5.i2.198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Revised: 01/22/2015] [Accepted: 03/18/2015] [Indexed: 02/06/2023] Open
Abstract
Until recently, fetal surgery was only used for fetuses with very poor prognosis who were likely to die without intervention. With advances in imaging, endoscopic techniques, anesthesia and novel interventions, fetal surgery is becoming a realistic option for conditions with less severe prognoses, where the aim is now to improve quality of life rather than simply allow survival. Until forty years ago, the uterus shielded the fetus from observation and therapy. Rapid changes in the diagnosis and treatment of human fetal anatomical abnormalities are due to improved fetal imaging studies, fetal sampling techniques (e.g., amniocentesis and chorionic villus sampling), and a better understanding of fetal pathophysiology derived from laboratory animals. Fetal therapy is the logical culmination of progress in fetal diagnosis. In other words, the fetus is now a patient. Now-a-days, in utero (IU) and exo utero (EU) surgical methods are popular for experimental analyses of the histogenesis of organ development. Using these surgical methods, developmental anomalies can be created and then repaired. By applying microinjection and/or fetal surgery with these methods, models of developmental anomalies such as neural tube defects, temporomandibular joint defects, hip joint defects, digit amputation, limb and digit development and regeneration, and tooth germ transplantation in the jaw could be created and later observed. After observing different types of anomalies, novel IU and EU surgical techniques would be the best approach for repairing or treating those anomalies or diseases. This review will focus on the rationale for the IU and EU creation of animal models of different organ defects or anomalies and their repair, based on analyses of organ histogenesis and pathologic observations. It will also focus in detail on the surgical techniques of both IU and EU methods.
Collapse
|
38
|
Hayashi S, Inoue Y, Kiyonari H, Abe T, Misaki K, Moriguchi H, Tanaka Y, Takeichi M. Protocadherin-17 mediates collective axon extension by recruiting actin regulator complexes to interaxonal contacts. Dev Cell 2014; 30:673-87. [PMID: 25199687 DOI: 10.1016/j.devcel.2014.07.015] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2013] [Revised: 06/07/2014] [Accepted: 07/16/2014] [Indexed: 11/17/2022]
Abstract
In the process of neuronal wiring, axons derived from the same functional group typically extend together, resulting in fascicle formation. How these axons communicate with one another remains largely unknown. Here, we show that protocadherin-17 (Pcdh17) supports this group extension by recruiting actin polymerization regulators to interaxonal contact sites. Pcdh17 is expressed by a subset of amygdala neurons, and it accumulates at axon-axon boundaries because of homophilic binding. Pcdh17 knockout in mice suppressed the extension of these axons. Ectopically expressed Pcdh17 altered the pattern of axon extension. In in-vitro cultures, wild-type growth cones normally migrate along other axons, whereas Pcdh17 null growth cones do not. Pcdh17 recruits the WAVE complex, Lamellipodin, and Ena/VASP to cell-cell contacts, converting these sites into motile structures. We propose that, through these mechanisms, Pcdh17 maintains the migration of growth cones that are in contact with other axons, thereby supporting their collective extension.
Collapse
Affiliation(s)
- Shuichi Hayashi
- Laboratory for Cell Adhesion and Tissue Patterning, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Yoko Inoue
- Laboratory for Cell Adhesion and Tissue Patterning, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Takaya Abe
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Kazuyo Misaki
- Laboratory for Electron Microscope, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Hiroyuki Moriguchi
- Laboratory for Integrated Biodevice, RIKEN Quantitative Biology Center, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Yo Tanaka
- Laboratory for Integrated Biodevice, RIKEN Quantitative Biology Center, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan
| | - Masatoshi Takeichi
- Laboratory for Cell Adhesion and Tissue Patterning, RIKEN Center for Developmental Biology, 2-2-3 Minatojima-Minamimachi, Chuo-ku, Kobe 650-0047, Japan.
| |
Collapse
|
39
|
Kawasaki H. Molecular investigations of the brain of higher mammals using gyrencephalic carnivore ferrets. Neurosci Res 2014; 86:59-65. [PMID: 24983876 DOI: 10.1016/j.neures.2014.06.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 06/16/2014] [Accepted: 06/17/2014] [Indexed: 11/17/2022]
Abstract
The brains of mammals such as carnivores and primates contain developed structures not found in the brains of mice. Uncovering the physiological importance, developmental mechanisms and evolution of these structures using carnivores and primates would greatly contribute to our understanding of the human brain and its diseases. Although the anatomical and physiological properties of the brains of carnivores and primates have been intensively examined, molecular investigations are still limited. Recently, genetic techniques that can be applied to carnivores and primates have been explored, and molecules whose expression patterns correspond to these structures were reported. Furthermore, to investigate the functional importance of these molecules, rapid and efficient genetic manipulation methods were established by applying electroporation to gyrencephalic carnivore ferrets. In this article, I review recent advances in molecular investigations of the brains of carnivores and primates, mainly focusing on ferrets (Mustela putorius furo).
Collapse
Affiliation(s)
- Hiroshi Kawasaki
- Graduate School of Medical Sciences, Kanazawa University, Ishikawa 920-8640, Japan; Brain/Liver Interface Medicine Research Center, Kanazawa University, Ishikawa 920-8640, Japan.
| |
Collapse
|
40
|
Chen CC, Winkler CM, Pfenning AR, Jarvis ED. Molecular profiling of the developing avian telencephalon: regional timing and brain subdivision continuities. J Comp Neurol 2014; 521:3666-701. [PMID: 23818174 PMCID: PMC3863995 DOI: 10.1002/cne.23406] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 06/19/2013] [Accepted: 06/21/2013] [Indexed: 12/30/2022]
Abstract
In our companion study (Jarvis et al. [2013] J Comp Neurol. doi: 10.1002/cne.23404) we used quantitative brain molecular profiling to discover that distinct subdivisions in the avian pallium above and below the ventricle and the associated mesopallium lamina have similar molecular profiles, leading to a hypothesis that they may form as continuous subdivisions around the lateral ventricle. To explore this hypothesis, here we profiled the expression of 16 genes at eight developmental stages. The genes included those that define brain subdivisions in the adult and some that are also involved in brain development. We found that phyletic hierarchical cluster and linear regression network analyses of gene expression profiles implicated single and mixed ancestry of these brain regions at early embryonic stages. Most gene expression-defined pallial subdivisions began as one ventral or dorsal domain that later formed specific folds around the lateral ventricle. Subsequently a clear ventricle boundary formed, partitioning them into dorsal and ventral pallial subdivisions surrounding the mesopallium lamina. These subdivisions each included two parts of the mesopallium, the nidopallium and hyperpallium, and the arcopallium and hippocampus, respectively. Each subdivision expression profile had a different temporal order of appearance, similar in timing to the order of analogous cell types of the mammalian cortex. Furthermore, like the mammalian pallium, expression in the ventral pallial subdivisions became distinct during prehatch development, whereas the dorsal portions did so during posthatch development. These findings support the continuum hypothesis of avian brain subdivision development around the ventricle and influence hypotheses on homologies of the avian pallium with other vertebrates.
Collapse
Affiliation(s)
- Chun-Chun Chen
- Department of Neurobiology, Howard Hughes Medical Institute, Duke University Medical Center, Durham, North Carolina, 27710
| | | | | | | |
Collapse
|
41
|
Jarvis ED, Yu J, Rivas MV, Horita H, Feenders G, Whitney O, Jarvis SC, Jarvis ER, Kubikova L, Puck AEP, Siang-Bakshi C, Martin S, McElroy M, Hara E, Howard J, Pfenning A, Mouritsen H, Chen CC, Wada K. Global view of the functional molecular organization of the avian cerebrum: mirror images and functional columns. J Comp Neurol 2014; 521:3614-65. [PMID: 23818122 DOI: 10.1002/cne.23404] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 06/19/2013] [Accepted: 06/21/2013] [Indexed: 11/06/2022]
Abstract
Based on quantitative cluster analyses of 52 constitutively expressed or behaviorally regulated genes in 23 brain regions, we present a global view of telencephalic organization of birds. The patterns of constitutively expressed genes revealed a partial mirror image organization of three major cell populations that wrap above, around, and below the ventricle and adjacent lamina through the mesopallium. The patterns of behaviorally regulated genes revealed functional columns of activation across boundaries of these cell populations, reminiscent of columns through layers of the mammalian cortex. The avian functionally regulated columns were of two types: those above the ventricle and associated mesopallial lamina, formed by our revised dorsal mesopallium, hyperpallium, and intercalated hyperpallium; and those below the ventricle, formed by our revised ventral mesopallium, nidopallium, and intercalated nidopallium. Based on these findings and known connectivity, we propose that the avian pallium has four major cell populations similar to those in mammalian cortex and some parts of the amygdala: 1) a primary sensory input population (intercalated pallium); 2) a secondary intrapallial population (nidopallium/hyperpallium); 3) a tertiary intrapallial population (mesopallium); and 4) a quaternary output population (the arcopallium). Each population contributes portions to columns that control different sensory or motor systems. We suggest that this organization of cell groups forms by expansion of contiguous developmental cell domains that wrap around the lateral ventricle and its extension through the middle of the mesopallium. We believe that the position of the lateral ventricle and its associated mesopallium lamina has resulted in a conceptual barrier to recognizing related cell groups across its border, thereby confounding our understanding of homologies with mammals.
Collapse
Affiliation(s)
- Erich D Jarvis
- Department of Neurobiology, Howard Hughes Medical Institute, Duke University Medical Center, Durham, North Carolina, 27710
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Hirata-Fukae C, Hirata T. The zinc finger geneFezf2is required for the development of excitatory neurons in the basolateral complex of the amygdala. Dev Dyn 2014; 243:1030-6. [DOI: 10.1002/dvdy.24137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 04/06/2014] [Accepted: 04/06/2014] [Indexed: 11/08/2022] Open
Affiliation(s)
| | - Tsutomu Hirata
- Senior Research Fellow Center; Ehime University; Ehime Japan
- Proteo-Science Center; Ehime University; Ehime Japan
| |
Collapse
|
43
|
Tekko T, Lilleväli K, Luuk H, Sütt S, Truu L, Örd T, Möls M, Vasar E. Initiation and developmental dynamics of Wfs1 expression in the context of neural differentiation and ER stress in mouse forebrain. Int J Dev Neurosci 2014; 35:80-8. [PMID: 24694561 DOI: 10.1016/j.ijdevneu.2014.03.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2013] [Revised: 03/22/2014] [Accepted: 03/23/2014] [Indexed: 02/04/2023] Open
Abstract
Wolframin (Wfs1) is a membrane glycoprotein that resides in the endoplasmic reticulum (ER) and regulates cellular Ca(2+) homeostasis. In pancreas Wfs1 attenuates unfolded protein response (UPR) and protects cells from apoptosis. Loss of Wfs1 function results in Wolfram syndrome (OMIM 222300) characterized by early-onset diabetes mellitus, progressive optic atrophy, diabetes insipidus, deafness, and psychiatric disorders. Similarly, Wfs1-/- mice exhibit diabetes and increased basal anxiety. In the adult central nervous system Wfs1 is prominent in central extended amygdala, striatum and hippocampus, brain structures largely involved in behavioral adaptation of the organism. Here, we describe the initiation pattern of Wfs1 expression in mouse forebrain using mRNA in situ hybridization and compare it with Synaptophysin (Syp1), a gene encoding synaptic vesicle protein widely used as neuronal differentiation marker. We show that the expression of Wfs1 starts during late embryonic development in the dorsal striatum and amygdala, then expands broadly at birth, possessing several transitory regions during maturation. Syp1 expression precedes Wfs1 and it is remarkably upregulated during the period of Wfs1 expression initiation and maturation, suggesting relationship between neural activation and Wfs1 expression. Using in situ hybridization and quantitative real-time PCR we show that UPR-related genes (Grp78, Grp94, and Chop) display dynamic expression in the perinatal brain when Wfs1 is initiated and their expression pattern is not altered in the brain lacking functional Wfs1.
Collapse
Affiliation(s)
- Triin Tekko
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Kersti Lilleväli
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; Department of Developmental Biology, Institute of Molecular and Cell Biology, Faculty of Science and Technology, University of Tartu, 46 Vanemuise Street, 51014 Tartu, Estonia.
| | - Hendrik Luuk
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Silva Sütt
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Laura Truu
- Department of Developmental Biology, Institute of Molecular and Cell Biology, Faculty of Science and Technology, University of Tartu, 46 Vanemuise Street, 51014 Tartu, Estonia; Competence Centre for Cancer Research, Akadeemia tee 15, Tallinn 12618, Estonia
| | - Tiit Örd
- Institute of Molecular and Cell Biology, University of Tartu, Riia 23, 51010 Tartu, Estonia
| | - Märt Möls
- Institute of Mathematical Statistics, University of Tartu, J. Liivi 2, 50409 Tartu, Estonia
| | - Eero Vasar
- Department of Physiology, Institute of Biomedicine and Translational Medicine, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| |
Collapse
|
44
|
Alfano C, Magrinelli E, Harb K, Studer M. The nuclear receptors COUP-TF: a long-lasting experience in forebrain assembly. Cell Mol Life Sci 2014; 71:43-62. [PMID: 23525662 PMCID: PMC11114017 DOI: 10.1007/s00018-013-1320-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Revised: 02/14/2013] [Accepted: 03/04/2013] [Indexed: 12/16/2022]
Abstract
Chicken ovalbumin upstream promoter transcription factors (COUP-TFs) are nuclear receptors belonging to the superfamily of the steroid/thyroid hormone receptors. Members of this family are internalized to the nucleus both in a ligand-dependent or -independent manner and act as strong transcriptional regulators by binding to the DNA of their target genes. COUP-TFs are defined as orphan receptors, since ligands regulating their activity have not so far been identified. From the very beginning of metazoan evolution, these molecules have been involved in various key events during embryonic development and organogenesis. In this review, we will mainly focus on their function during development and maturation of the central nervous system, which has been well characterized in various animal classes ranging from ctenophores to mammals. We will start by introducing the current knowledge on COUP-TF mechanisms of action and then focus our discussion on the crucial processes underlying forebrain ontogenesis, with special emphasis on mammalian development. Finally, the conserved roles of COUP-TFs along phylogenesis will be highlighted, and some hypotheses, worth exploring in future years to gain more insight into the mechanisms controlled by these factors, will be proposed.
Collapse
Affiliation(s)
- Christian Alfano
- Institute of Biology Valrose, iBV, UMR INSERM1091/CNRS7277/UNS, 06108 Nice, France
- University of Nice-Sophia Antipolis, UFR Sciences, 06108 Nice, France
| | - Elia Magrinelli
- Institute of Biology Valrose, iBV, UMR INSERM1091/CNRS7277/UNS, 06108 Nice, France
- University of Nice-Sophia Antipolis, UFR Sciences, 06108 Nice, France
| | - Kawssar Harb
- Institute of Biology Valrose, iBV, UMR INSERM1091/CNRS7277/UNS, 06108 Nice, France
- University of Nice-Sophia Antipolis, UFR Sciences, 06108 Nice, France
| | - Michèle Studer
- Institute of Biology Valrose, iBV, UMR INSERM1091/CNRS7277/UNS, 06108 Nice, France
- University of Nice-Sophia Antipolis, UFR Sciences, 06108 Nice, France
| |
Collapse
|
45
|
Abstract
In utero electroporation is a rapid and powerful technique to study the development of many brain regions. This approach presents several advantages over other methods to study specific steps of brain development in vivo, from proliferation to synaptic integration. Here, we describe in detail the individual steps necessary to carry out the technique. We also highlight the variations that can be implemented to target different cerebral structures and to study specific steps of development.
Collapse
|
46
|
Shinohara M, Zhu Y, Murakami F. Four-dimensional analysis of nucleogenesis of the pontine nucleus in the hindbrain. J Comp Neurol 2013; 521:3340-57. [DOI: 10.1002/cne.23353] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 04/09/2013] [Accepted: 04/25/2013] [Indexed: 01/22/2023]
Affiliation(s)
- Masaki Shinohara
- Graduate School of Frontier Biosciences, Osaka University; Suita; Osaka; 560-8531; Japan
| | - Yan Zhu
- Graduate School of Frontier Biosciences, Osaka University; Suita; Osaka; 560-8531; Japan
| | - Fujio Murakami
- Graduate School of Frontier Biosciences, Osaka University; Suita; Osaka; 560-8531; Japan
| |
Collapse
|
47
|
Hertel N, Redies C, Medina L. Cadherin expression delineates the divisions of the postnatal and adult mouse amygdala. J Comp Neurol 2013; 520:3982-4012. [PMID: 22592879 DOI: 10.1002/cne.23140] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The amygdaloid complex represents a group of telencephalic nuclei and cortical areas that control emotional and social behavior. Amygdalar development is poorly understood. It is generally accepted that the structures of the amygdala originate from the neuroepithelium at both sides of the pallial-subpallial boundary. In the present study, we mapped the expression of 13 members of the cadherin superfamily of cell adhesion molecules, which provide an adhesive code for the development and maintenance of functional structures in the central nervous system (CNS). Five classic cadherins (Cdh4, Cdh6, Cdh7, Cdh8, Cdh11) and eight delta-protocadherins (Pcdh1, Pcdh7, Pcdh8, Pcdh9, Pcdh10, Pcdh11, PCdh17, PCdh19) were studied by in situ hybridization in the postnatal (P5) and adult mouse amygdala. In the different parts of the amygdala, each of these (proto-) cadherins shows a distinct and spatially restricted expression pattern that is highly similar at postnatal and adult stages. The combinatorial expression of (proto-) cadherins allows the distinction of multiple molecular subdivisions within the amygdala that partially coincide with previously described morphological divisions. Beyond these expected results, a number of novel molecular subdivisions and subpopulations of cells were identified; for example, additional molecular subdomains, patches, or cell aggregates with distinct (proto-) cadherin expression in several nuclei/areas of the amygdala. We also show that several cadherins are molecular markers for particular functional subsystems within the amygdala, such as in the olfactory projections. In summary, (proto-) cadherins provide a code of potentially adhesive cues that can aid the understanding of functional organization in the amygdala.
Collapse
Affiliation(s)
- Nicole Hertel
- Institute of Anatomy I, Friedrich Schiller University School of Medicine, Jena University Hospital, 07743 Jena, Germany
| | | | | |
Collapse
|
48
|
Dynamic expression of tyrosine hydroxylase mRNA and protein in neurons of the striatum and amygdala of mice, and experimental evidence of their multiple embryonic origin. Brain Struct Funct 2013; 219:751-76. [PMID: 23479178 PMCID: PMC4023077 DOI: 10.1007/s00429-013-0533-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Accepted: 02/21/2013] [Indexed: 12/22/2022]
Abstract
Emotional and motivational dysfunctions observed in Parkinson's disease, schizophrenia, and drug addiction are associated to an alteration of the mesocortical and mesolimbic dopaminergic pathways, which include axons projecting to the prefrontal cortex, the ventral striatum, and the amygdala. Subpopulations of catecholaminergic neurons have been described in the cortex and striatum of several mammals, but the presence of such cells in the adult amygdala is unclear in murine rodents, and in other rodents appears to show variations depending on the species. Moreover, the embryonic origin of telencephalic tyrosine hydroxylase (TH) cells is unknown, which is essential for trying to understand aspects of their evolution, distribution and function. Herein we investigated the expression of TH mRNA and protein in cells of the striatum and amygdala of developing and adult mice, and analyzed the embryonic origin of such cells using in vitro migration assays. Our results showed the presence of TH mRNA and protein expressing cells in the striatum (including nucleus accumbens), central and medial extended amygdala during development, which are persistent in adulthood although they are less numerous, generally show weak mRNA expression, and some appear to lack the protein. Fate mapping analysis showed that these cells include at least two subpopulations with different embryonic origin in either the commissural preoptic area of the subpallium or the supraopto-paraventricular domain of the alar hypothalamus. These data are important for future studies trying to understand the role of catecholamines in modulation of emotion, motivation, and reward.
Collapse
|
49
|
Maximino C, Lima MG, Oliveira KRM, Batista EDJO, Herculano AM. “Limbic associative” and “autonomic” amygdala in teleosts: A review of the evidence. J Chem Neuroanat 2013; 48-49:1-13. [DOI: 10.1016/j.jchemneu.2012.10.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 10/04/2012] [Accepted: 10/05/2012] [Indexed: 12/31/2022]
|
50
|
Dual origins of the mammalian accessory olfactory bulb revealed by an evolutionarily conserved migratory stream. Nat Neurosci 2013; 16:157-65. [PMID: 23292680 DOI: 10.1038/nn.3297] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 12/05/2012] [Indexed: 11/09/2022]
Abstract
The accessory olfactory bulb (AOB) is a critical olfactory structure that has been implicated in mediating social behavior. It receives input from the vomeronasal organ and projects to targets in the amygdaloid complex. Its anterior and posterior components (aAOB and pAOB) display molecular, connectional and functional segregation in processing reproductive and defensive and aggressive behaviors, respectively. We observed a dichotomy in the development of the projection neurons of the aAOB and pAOB in mice. We found that they had distinct sites of origin and that different regulatory molecules were required for their specification and migration. aAOB neurons arose locally in the rostral telencephalon, similar to main olfactory bulb neurons. In contrast, pAOB neurons arose caudally, from the neuroepithelium of the diencephalic-telencephalic boundary, from which they migrated rostrally to reach their destination. This unusual origin and migration is conserved in Xenopus, providing an insight into the origin of a key component of this system in evolution.
Collapse
|