1
|
Placzek M, Chinnaiya K, Kim DW, Blackshaw S. Control of tuberal hypothalamic development and its implications in metabolic disorders. Nat Rev Endocrinol 2024:10.1038/s41574-024-01036-1. [PMID: 39313573 DOI: 10.1038/s41574-024-01036-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2024] [Indexed: 09/25/2024]
Abstract
The tuberal hypothalamus regulates a range of crucial physiological processes, including energy homeostasis and metabolism. In this Review, we explore the intricate molecular mechanisms and signalling pathways that control the development of the tuberal hypothalamus, focusing on aspects that shape metabolic outcomes. Major developmental events are discussed in the context of their effect on the establishment of both functional hypothalamic neuronal circuits and brain-body interfaces that are pivotal to the control of metabolism. Emerging evidence indicates that aberrations in molecular pathways during tuberal hypothalamic development contribute to metabolic dysregulation. Understanding the molecular underpinnings of tuberal hypothalamic development provides a comprehensive view of neurodevelopmental processes and offers a promising avenue for future targeted interventions to prevent and treat metabolic disorders.
Collapse
Affiliation(s)
- Marysia Placzek
- School of Biosciences, University of Sheffield, Sheffield, UK.
- Bateson Centre, University of Sheffield, Sheffield, UK.
- Neuroscience Institute, University of Sheffield, Sheffield, UK.
| | | | - Dong Won Kim
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Bock M, Hong SJ, Zhang S, Yu Y, Lee S, Shin H, Choi BH, Han I. Morphogenetic Designs, and Disease Models in Central Nervous System Organoids. Int J Mol Sci 2024; 25:7750. [PMID: 39062993 PMCID: PMC11276855 DOI: 10.3390/ijms25147750] [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: 06/24/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Since the emergence of the first cerebral organoid (CO) in 2013, advancements have transformed central nervous system (CNS) research. Initial efforts focused on studying the morphogenesis of COs and creating reproducible models. Numerous methodologies have been proposed, enabling the design of the brain organoid to represent specific regions and spinal cord structures. CNS organoids now facilitate the study of a wide range of CNS diseases, from infections to tumors, which were previously difficult to investigate. We summarize the major advancements in CNS organoids, concerning morphogenetic designs and disease models. We examine the development of fabrication procedures and how these advancements have enabled the generation of region-specific brain organoids and spinal cord models. We highlight the application of these organoids in studying various CNS diseases, demonstrating the versatility and potential of organoid models in advancing our understanding of complex conditions. We discuss the current challenges in the field, including issues related to reproducibility, scalability, and the accurate recapitulation of the in vivo environment. We provide an outlook on prospective studies and future directions. This review aims to provide a comprehensive overview of the state-of-the-art CNS organoid research, highlighting key developments, current challenges, and prospects in the field.
Collapse
Affiliation(s)
- Minsung Bock
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Sung Jun Hong
- Research Competency Milestones Program, School of Medicine, CHA University, Seongnam-si 13488, Republic of Korea;
- Department of Medicine, School of Medicine, CHA University, Seongnam-si 13496, Republic of Korea
| | - Songzi Zhang
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Yerin Yu
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Somin Lee
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Haeeun Shin
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
| | - Byung Hyune Choi
- Department of Biomedical Science, Inha University College of Medicine, Incheon 22212, Republic of Korea;
| | - Inbo Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam-si 13496, Republic of Korea; (M.B.); (S.Z.); (Y.Y.); (S.L.); (H.S.)
- Advanced Regenerative Medicine Research Center, CHA Future Medicine Research Institute, Seongnam-si 13488, Republic of Korea
| |
Collapse
|
3
|
Chinnaiya K, Burbridge S, Jones A, Kim DW, Place E, Manning E, Groves I, Sun C, Towers M, Blackshaw S, Placzek M. A neuroepithelial wave of BMP signalling drives anteroposterior specification of the tuberal hypothalamus. eLife 2023; 12:e83133. [PMID: 36718990 PMCID: PMC9917434 DOI: 10.7554/elife.83133] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 01/29/2023] [Indexed: 02/01/2023] Open
Abstract
The tuberal hypothalamus controls life-supporting homeostatic processes, but despite its fundamental role, the cells and signalling pathways that specify this unique region of the central nervous system in embryogenesis are poorly characterised. Here, we combine experimental and bioinformatic approaches in the embryonic chick to show that the tuberal hypothalamus is progressively generated from hypothalamic floor plate-like cells. Fate-mapping studies show that a stream of tuberal progenitors develops in the anterior-ventral neural tube as a wave of neuroepithelial-derived BMP signalling sweeps from anterior to posterior through the hypothalamic floor plate. As later-specified posterior tuberal progenitors are generated, early specified anterior tuberal progenitors become progressively more distant from these BMP signals and differentiate into tuberal neurogenic cells. Gain- and loss-of-function experiments in vivo and ex vivo show that BMP signalling initiates tuberal progenitor specification, but must be eliminated for these to progress to anterior neurogenic progenitors. scRNA-Seq profiling shows that tuberal progenitors that are specified after the major period of anterior tuberal specification begin to upregulate genes that characterise radial glial cells. This study provides an integrated account of the development of the tuberal hypothalamus.
Collapse
Affiliation(s)
- Kavitha Chinnaiya
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Sarah Burbridge
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Aragorn Jones
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Elsie Place
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Elizabeth Manning
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Ian Groves
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Changyu Sun
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Matthew Towers
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Psychiatry and Behavioral Science, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Ophthalmology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Institute for Cell Engineering, Johns Hopkins University School of MedicineBaltimoreUnited States
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Marysia Placzek
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
- Bateson Centre, University of SheffieldSheffieldUnited Kingdom
- Neuroscience Institute, University of SheffieldSheffieldUnited Kingdom
| |
Collapse
|
4
|
López-González L, Martínez-de-la-Torre M, Puelles L. Populational heterogeneity and partial migratory origin of the ventromedial hypothalamic nucleus: genoarchitectonic analysis in the mouse. Brain Struct Funct 2023; 228:537-576. [PMID: 36598560 PMCID: PMC9944059 DOI: 10.1007/s00429-022-02601-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/27/2022] [Indexed: 01/05/2023]
Abstract
The ventromedial hypothalamic nucleus (VMH) is one of the most distinctive hypothalamic tuberal structures, subject of numerous classic and modern functional studies. Commonly, the adult VMH has been divided in several portions, attending to differences in cell aggregation, cell type, connectivity, and function. Consensus VMH partitions in the literature comprise the dorsomedial (VMHdm), and ventrolateral (VMHvl) subnuclei, which are separated by an intermediate or central (VMHc) population (topographic names based on the columnar axis). However, some recent transcriptome analyses have identified a higher number of different cell types in the VMH, suggesting additional subdivisions, as well as the possibility of separate origins. We offer a topologic and genoarchitectonic developmental study of the mouse VMH complex using the prosomeric axis as a reference. We analyzed genes labeling specific VMH subpopulations, with particular focus upon the Nkx2.2 transcription factor, a marker of the alar-basal boundary territory of the prosencephalon, from where some cells seem to migrate dorsoventrally into VMH. We also identified separate neuroepithelial origins of a Nr2f1-positive subpopulation, and a new Six3-positive component, as well as subtle differences in origin of Nr5a1 positive versus Nkx2.2-positive cell populations entering dorsoventrally the VMH. Several of these migrating cell types are born in the dorsal tuberal domain and translocate ventralwards to reach the intermediate tuberal domain, where the adult VMH mass is located in the adult. This work provides a more detailed area map on the intrinsic organization of the postmigratory VMH complex, helpful for deeper functional studies of this basal hypothalamic entity.
Collapse
Affiliation(s)
- Lara López-González
- grid.10586.3a0000 0001 2287 8496University of Murcia, IMIB-Arrixaca Institute of Biomedical Research, El Palmar, 30120 Murcia, Spain
| | - Margaret Martínez-de-la-Torre
- grid.10586.3a0000 0001 2287 8496University of Murcia, IMIB-Arrixaca Institute of Biomedical Research, El Palmar, 30120 Murcia, Spain
| | - Luis Puelles
- University of Murcia, IMIB-Arrixaca Institute of Biomedical Research, El Palmar, 30120, Murcia, Spain.
| |
Collapse
|
5
|
Place E, Manning E, Kim DW, Kinjo A, Nakamura G, Ohyama K. SHH and Notch regulate SOX9+ progenitors to govern arcuate POMC neurogenesis. Front Neurosci 2022; 16:855288. [PMID: 36033614 PMCID: PMC9404380 DOI: 10.3389/fnins.2022.855288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 07/20/2022] [Indexed: 12/05/2022] Open
Abstract
Pro-opiomelanocortin (POMC)-expressing neurons in the hypothalamic arcuate nucleus (ARC) play key roles in feeding and energy homoeostasis, hence their development is of great research interest. As the process of neurogenesis is accompanied by changes in adhesion, polarity, and migration that resemble aspects of epithelial-to-mesenchymal transitions (EMTs), we have characterised the expression and regulation within the prospective ARC of transcription factors with context-dependent abilities to regulate aspects of EMT. Informed by pseudotime meta-analysis of recent scRNA-seq data, we use immunohistochemistry and multiplex in situ hybridisation to show that SOX2, SRY-Box transcription factor 9 (SOX9), PROX1, Islet1 (ISL1), and SOX11 are sequentially expressed over the course of POMC neurogenesis in the embryonic chick. Through pharmacological studies ex vivo, we demonstrate that while inhibiting either sonic hedgehog (SHH) or Notch signalling reduces the number of SOX9+ neural progenitor cells, these treatments lead, respectively, to lesser and greater numbers of differentiating ISL1+/POMC+ neurons. These results are consistent with a model in which SHH promotes the formation of SOX9+ progenitors, and Notch acts to limit their differentiation. Both pathways are also required to maintain normal levels of proliferation and to suppress apoptosis. Together our findings demonstrate that hypothalamic neurogenesis is accompanied by dynamic expression of transcription factors (TFs) that mediate EMTs, and that SHH and Notch signalling converge to regulate hypothalamic cellular homoeostasis.
Collapse
Affiliation(s)
- Elsie Place
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
| | - Elizabeth Manning
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Arisa Kinjo
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Go Nakamura
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| | - Kyoji Ohyama
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
6
|
Cohen NT, Cross JH, Arzimanoglou A, Berkovic SF, Kerrigan JF, Miller IP, Webster E, Soeby L, Cukiert A, Hesdorffer DK, Kroner BL, Saper CB, Schulze-Bonhage A, Gaillard WD. Hypothalamic Hamartomas: Evolving Understanding and Management. Neurology 2021; 97:864-873. [PMID: 34607926 PMCID: PMC8610628 DOI: 10.1212/wnl.0000000000012773] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 08/31/2021] [Indexed: 11/15/2022] Open
Abstract
Hypothalamic hamartomas (HH) are rare, basilar developmental lesions with widespread comorbidities often associated with refractory epilepsy and encephalopathy. Imaging advances allow for early, even prenatal, detection. Genetic studies suggest mutations in GLI3 and other patterning genes are involved in HH pathogenesis. About 50%-80% of children with HH have severe rage and aggression and a majority of patients exhibit externalizing disorders. Behavioral disruption and intellectual disability may predate epilepsy. Neuropsychological, sleep, and endocrine disorders are typical. The purpose of this article is to provide a summary of the current understanding of HH and to highlight opportunities for future research.
Collapse
Affiliation(s)
- Nathan T Cohen
- From the Center for Neuroscience Research (N.T.C., W.D.G.), Children's National Hospital, The George Washington University School of Medicine, Washington, DC; UCL NIHR BRC Great Ormond Street Institute of Child Health (J.H.C.), Member of ERN-EpiCARE, London; Great Ormond Street Hospital for Children (J.H.C.), NHS Trust, London; Young Epilepsy (J.H.C.), Lingfield, Surrey, UK; Department of Pediatric Clinical Epileptology (A.A.), Sleep Disorders and Functional Neurology, Member of ERN-EpiCARE; HFME (A.A.), Hospices Civils de Lyon, France; Epilepsy Research Unit (A.A.), Barcelona's Children Hospital San Juan de Dios, Member of the ERN EpiCARE, Spain; Epilepsy Research Centre (S.F.B.), University of Melbourne, Australia; Division of Pediatric Neurology (J.F.K.), Barrow Neurological Institute at Phoenix Children's Hospital; Hope for Hypothalamic Hamartomas (I.P.M., E.W., L.S.), Phoenix, AZ; Epilepsy Surgery Program (A.C.), Clinica de Epilepsia de Sao Paulo, Brazil; Department of Epidemiology (D.K.H.), Columbia University Medical Center, New York, NY; RTI International (B.L.K.), Rockville, MD; Department of Neurology (C.B.S.), Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and Epilepsy Center (A.S.-B.), Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Germany.
| | - J Helen Cross
- From the Center for Neuroscience Research (N.T.C., W.D.G.), Children's National Hospital, The George Washington University School of Medicine, Washington, DC; UCL NIHR BRC Great Ormond Street Institute of Child Health (J.H.C.), Member of ERN-EpiCARE, London; Great Ormond Street Hospital for Children (J.H.C.), NHS Trust, London; Young Epilepsy (J.H.C.), Lingfield, Surrey, UK; Department of Pediatric Clinical Epileptology (A.A.), Sleep Disorders and Functional Neurology, Member of ERN-EpiCARE; HFME (A.A.), Hospices Civils de Lyon, France; Epilepsy Research Unit (A.A.), Barcelona's Children Hospital San Juan de Dios, Member of the ERN EpiCARE, Spain; Epilepsy Research Centre (S.F.B.), University of Melbourne, Australia; Division of Pediatric Neurology (J.F.K.), Barrow Neurological Institute at Phoenix Children's Hospital; Hope for Hypothalamic Hamartomas (I.P.M., E.W., L.S.), Phoenix, AZ; Epilepsy Surgery Program (A.C.), Clinica de Epilepsia de Sao Paulo, Brazil; Department of Epidemiology (D.K.H.), Columbia University Medical Center, New York, NY; RTI International (B.L.K.), Rockville, MD; Department of Neurology (C.B.S.), Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and Epilepsy Center (A.S.-B.), Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Germany
| | - Alexis Arzimanoglou
- From the Center for Neuroscience Research (N.T.C., W.D.G.), Children's National Hospital, The George Washington University School of Medicine, Washington, DC; UCL NIHR BRC Great Ormond Street Institute of Child Health (J.H.C.), Member of ERN-EpiCARE, London; Great Ormond Street Hospital for Children (J.H.C.), NHS Trust, London; Young Epilepsy (J.H.C.), Lingfield, Surrey, UK; Department of Pediatric Clinical Epileptology (A.A.), Sleep Disorders and Functional Neurology, Member of ERN-EpiCARE; HFME (A.A.), Hospices Civils de Lyon, France; Epilepsy Research Unit (A.A.), Barcelona's Children Hospital San Juan de Dios, Member of the ERN EpiCARE, Spain; Epilepsy Research Centre (S.F.B.), University of Melbourne, Australia; Division of Pediatric Neurology (J.F.K.), Barrow Neurological Institute at Phoenix Children's Hospital; Hope for Hypothalamic Hamartomas (I.P.M., E.W., L.S.), Phoenix, AZ; Epilepsy Surgery Program (A.C.), Clinica de Epilepsia de Sao Paulo, Brazil; Department of Epidemiology (D.K.H.), Columbia University Medical Center, New York, NY; RTI International (B.L.K.), Rockville, MD; Department of Neurology (C.B.S.), Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and Epilepsy Center (A.S.-B.), Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Germany
| | - Samuel F Berkovic
- From the Center for Neuroscience Research (N.T.C., W.D.G.), Children's National Hospital, The George Washington University School of Medicine, Washington, DC; UCL NIHR BRC Great Ormond Street Institute of Child Health (J.H.C.), Member of ERN-EpiCARE, London; Great Ormond Street Hospital for Children (J.H.C.), NHS Trust, London; Young Epilepsy (J.H.C.), Lingfield, Surrey, UK; Department of Pediatric Clinical Epileptology (A.A.), Sleep Disorders and Functional Neurology, Member of ERN-EpiCARE; HFME (A.A.), Hospices Civils de Lyon, France; Epilepsy Research Unit (A.A.), Barcelona's Children Hospital San Juan de Dios, Member of the ERN EpiCARE, Spain; Epilepsy Research Centre (S.F.B.), University of Melbourne, Australia; Division of Pediatric Neurology (J.F.K.), Barrow Neurological Institute at Phoenix Children's Hospital; Hope for Hypothalamic Hamartomas (I.P.M., E.W., L.S.), Phoenix, AZ; Epilepsy Surgery Program (A.C.), Clinica de Epilepsia de Sao Paulo, Brazil; Department of Epidemiology (D.K.H.), Columbia University Medical Center, New York, NY; RTI International (B.L.K.), Rockville, MD; Department of Neurology (C.B.S.), Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and Epilepsy Center (A.S.-B.), Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Germany
| | - John F Kerrigan
- From the Center for Neuroscience Research (N.T.C., W.D.G.), Children's National Hospital, The George Washington University School of Medicine, Washington, DC; UCL NIHR BRC Great Ormond Street Institute of Child Health (J.H.C.), Member of ERN-EpiCARE, London; Great Ormond Street Hospital for Children (J.H.C.), NHS Trust, London; Young Epilepsy (J.H.C.), Lingfield, Surrey, UK; Department of Pediatric Clinical Epileptology (A.A.), Sleep Disorders and Functional Neurology, Member of ERN-EpiCARE; HFME (A.A.), Hospices Civils de Lyon, France; Epilepsy Research Unit (A.A.), Barcelona's Children Hospital San Juan de Dios, Member of the ERN EpiCARE, Spain; Epilepsy Research Centre (S.F.B.), University of Melbourne, Australia; Division of Pediatric Neurology (J.F.K.), Barrow Neurological Institute at Phoenix Children's Hospital; Hope for Hypothalamic Hamartomas (I.P.M., E.W., L.S.), Phoenix, AZ; Epilepsy Surgery Program (A.C.), Clinica de Epilepsia de Sao Paulo, Brazil; Department of Epidemiology (D.K.H.), Columbia University Medical Center, New York, NY; RTI International (B.L.K.), Rockville, MD; Department of Neurology (C.B.S.), Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and Epilepsy Center (A.S.-B.), Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Germany
| | - Ilene Penn Miller
- From the Center for Neuroscience Research (N.T.C., W.D.G.), Children's National Hospital, The George Washington University School of Medicine, Washington, DC; UCL NIHR BRC Great Ormond Street Institute of Child Health (J.H.C.), Member of ERN-EpiCARE, London; Great Ormond Street Hospital for Children (J.H.C.), NHS Trust, London; Young Epilepsy (J.H.C.), Lingfield, Surrey, UK; Department of Pediatric Clinical Epileptology (A.A.), Sleep Disorders and Functional Neurology, Member of ERN-EpiCARE; HFME (A.A.), Hospices Civils de Lyon, France; Epilepsy Research Unit (A.A.), Barcelona's Children Hospital San Juan de Dios, Member of the ERN EpiCARE, Spain; Epilepsy Research Centre (S.F.B.), University of Melbourne, Australia; Division of Pediatric Neurology (J.F.K.), Barrow Neurological Institute at Phoenix Children's Hospital; Hope for Hypothalamic Hamartomas (I.P.M., E.W., L.S.), Phoenix, AZ; Epilepsy Surgery Program (A.C.), Clinica de Epilepsia de Sao Paulo, Brazil; Department of Epidemiology (D.K.H.), Columbia University Medical Center, New York, NY; RTI International (B.L.K.), Rockville, MD; Department of Neurology (C.B.S.), Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and Epilepsy Center (A.S.-B.), Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Germany
| | - Erica Webster
- From the Center for Neuroscience Research (N.T.C., W.D.G.), Children's National Hospital, The George Washington University School of Medicine, Washington, DC; UCL NIHR BRC Great Ormond Street Institute of Child Health (J.H.C.), Member of ERN-EpiCARE, London; Great Ormond Street Hospital for Children (J.H.C.), NHS Trust, London; Young Epilepsy (J.H.C.), Lingfield, Surrey, UK; Department of Pediatric Clinical Epileptology (A.A.), Sleep Disorders and Functional Neurology, Member of ERN-EpiCARE; HFME (A.A.), Hospices Civils de Lyon, France; Epilepsy Research Unit (A.A.), Barcelona's Children Hospital San Juan de Dios, Member of the ERN EpiCARE, Spain; Epilepsy Research Centre (S.F.B.), University of Melbourne, Australia; Division of Pediatric Neurology (J.F.K.), Barrow Neurological Institute at Phoenix Children's Hospital; Hope for Hypothalamic Hamartomas (I.P.M., E.W., L.S.), Phoenix, AZ; Epilepsy Surgery Program (A.C.), Clinica de Epilepsia de Sao Paulo, Brazil; Department of Epidemiology (D.K.H.), Columbia University Medical Center, New York, NY; RTI International (B.L.K.), Rockville, MD; Department of Neurology (C.B.S.), Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and Epilepsy Center (A.S.-B.), Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Germany
| | - Lisa Soeby
- From the Center for Neuroscience Research (N.T.C., W.D.G.), Children's National Hospital, The George Washington University School of Medicine, Washington, DC; UCL NIHR BRC Great Ormond Street Institute of Child Health (J.H.C.), Member of ERN-EpiCARE, London; Great Ormond Street Hospital for Children (J.H.C.), NHS Trust, London; Young Epilepsy (J.H.C.), Lingfield, Surrey, UK; Department of Pediatric Clinical Epileptology (A.A.), Sleep Disorders and Functional Neurology, Member of ERN-EpiCARE; HFME (A.A.), Hospices Civils de Lyon, France; Epilepsy Research Unit (A.A.), Barcelona's Children Hospital San Juan de Dios, Member of the ERN EpiCARE, Spain; Epilepsy Research Centre (S.F.B.), University of Melbourne, Australia; Division of Pediatric Neurology (J.F.K.), Barrow Neurological Institute at Phoenix Children's Hospital; Hope for Hypothalamic Hamartomas (I.P.M., E.W., L.S.), Phoenix, AZ; Epilepsy Surgery Program (A.C.), Clinica de Epilepsia de Sao Paulo, Brazil; Department of Epidemiology (D.K.H.), Columbia University Medical Center, New York, NY; RTI International (B.L.K.), Rockville, MD; Department of Neurology (C.B.S.), Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and Epilepsy Center (A.S.-B.), Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Germany
| | - Arthur Cukiert
- From the Center for Neuroscience Research (N.T.C., W.D.G.), Children's National Hospital, The George Washington University School of Medicine, Washington, DC; UCL NIHR BRC Great Ormond Street Institute of Child Health (J.H.C.), Member of ERN-EpiCARE, London; Great Ormond Street Hospital for Children (J.H.C.), NHS Trust, London; Young Epilepsy (J.H.C.), Lingfield, Surrey, UK; Department of Pediatric Clinical Epileptology (A.A.), Sleep Disorders and Functional Neurology, Member of ERN-EpiCARE; HFME (A.A.), Hospices Civils de Lyon, France; Epilepsy Research Unit (A.A.), Barcelona's Children Hospital San Juan de Dios, Member of the ERN EpiCARE, Spain; Epilepsy Research Centre (S.F.B.), University of Melbourne, Australia; Division of Pediatric Neurology (J.F.K.), Barrow Neurological Institute at Phoenix Children's Hospital; Hope for Hypothalamic Hamartomas (I.P.M., E.W., L.S.), Phoenix, AZ; Epilepsy Surgery Program (A.C.), Clinica de Epilepsia de Sao Paulo, Brazil; Department of Epidemiology (D.K.H.), Columbia University Medical Center, New York, NY; RTI International (B.L.K.), Rockville, MD; Department of Neurology (C.B.S.), Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and Epilepsy Center (A.S.-B.), Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Germany
| | - Dale K Hesdorffer
- From the Center for Neuroscience Research (N.T.C., W.D.G.), Children's National Hospital, The George Washington University School of Medicine, Washington, DC; UCL NIHR BRC Great Ormond Street Institute of Child Health (J.H.C.), Member of ERN-EpiCARE, London; Great Ormond Street Hospital for Children (J.H.C.), NHS Trust, London; Young Epilepsy (J.H.C.), Lingfield, Surrey, UK; Department of Pediatric Clinical Epileptology (A.A.), Sleep Disorders and Functional Neurology, Member of ERN-EpiCARE; HFME (A.A.), Hospices Civils de Lyon, France; Epilepsy Research Unit (A.A.), Barcelona's Children Hospital San Juan de Dios, Member of the ERN EpiCARE, Spain; Epilepsy Research Centre (S.F.B.), University of Melbourne, Australia; Division of Pediatric Neurology (J.F.K.), Barrow Neurological Institute at Phoenix Children's Hospital; Hope for Hypothalamic Hamartomas (I.P.M., E.W., L.S.), Phoenix, AZ; Epilepsy Surgery Program (A.C.), Clinica de Epilepsia de Sao Paulo, Brazil; Department of Epidemiology (D.K.H.), Columbia University Medical Center, New York, NY; RTI International (B.L.K.), Rockville, MD; Department of Neurology (C.B.S.), Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and Epilepsy Center (A.S.-B.), Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Germany
| | - Barbara L Kroner
- From the Center for Neuroscience Research (N.T.C., W.D.G.), Children's National Hospital, The George Washington University School of Medicine, Washington, DC; UCL NIHR BRC Great Ormond Street Institute of Child Health (J.H.C.), Member of ERN-EpiCARE, London; Great Ormond Street Hospital for Children (J.H.C.), NHS Trust, London; Young Epilepsy (J.H.C.), Lingfield, Surrey, UK; Department of Pediatric Clinical Epileptology (A.A.), Sleep Disorders and Functional Neurology, Member of ERN-EpiCARE; HFME (A.A.), Hospices Civils de Lyon, France; Epilepsy Research Unit (A.A.), Barcelona's Children Hospital San Juan de Dios, Member of the ERN EpiCARE, Spain; Epilepsy Research Centre (S.F.B.), University of Melbourne, Australia; Division of Pediatric Neurology (J.F.K.), Barrow Neurological Institute at Phoenix Children's Hospital; Hope for Hypothalamic Hamartomas (I.P.M., E.W., L.S.), Phoenix, AZ; Epilepsy Surgery Program (A.C.), Clinica de Epilepsia de Sao Paulo, Brazil; Department of Epidemiology (D.K.H.), Columbia University Medical Center, New York, NY; RTI International (B.L.K.), Rockville, MD; Department of Neurology (C.B.S.), Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and Epilepsy Center (A.S.-B.), Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Germany
| | - Clifford B Saper
- From the Center for Neuroscience Research (N.T.C., W.D.G.), Children's National Hospital, The George Washington University School of Medicine, Washington, DC; UCL NIHR BRC Great Ormond Street Institute of Child Health (J.H.C.), Member of ERN-EpiCARE, London; Great Ormond Street Hospital for Children (J.H.C.), NHS Trust, London; Young Epilepsy (J.H.C.), Lingfield, Surrey, UK; Department of Pediatric Clinical Epileptology (A.A.), Sleep Disorders and Functional Neurology, Member of ERN-EpiCARE; HFME (A.A.), Hospices Civils de Lyon, France; Epilepsy Research Unit (A.A.), Barcelona's Children Hospital San Juan de Dios, Member of the ERN EpiCARE, Spain; Epilepsy Research Centre (S.F.B.), University of Melbourne, Australia; Division of Pediatric Neurology (J.F.K.), Barrow Neurological Institute at Phoenix Children's Hospital; Hope for Hypothalamic Hamartomas (I.P.M., E.W., L.S.), Phoenix, AZ; Epilepsy Surgery Program (A.C.), Clinica de Epilepsia de Sao Paulo, Brazil; Department of Epidemiology (D.K.H.), Columbia University Medical Center, New York, NY; RTI International (B.L.K.), Rockville, MD; Department of Neurology (C.B.S.), Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and Epilepsy Center (A.S.-B.), Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Germany
| | - Andreas Schulze-Bonhage
- From the Center for Neuroscience Research (N.T.C., W.D.G.), Children's National Hospital, The George Washington University School of Medicine, Washington, DC; UCL NIHR BRC Great Ormond Street Institute of Child Health (J.H.C.), Member of ERN-EpiCARE, London; Great Ormond Street Hospital for Children (J.H.C.), NHS Trust, London; Young Epilepsy (J.H.C.), Lingfield, Surrey, UK; Department of Pediatric Clinical Epileptology (A.A.), Sleep Disorders and Functional Neurology, Member of ERN-EpiCARE; HFME (A.A.), Hospices Civils de Lyon, France; Epilepsy Research Unit (A.A.), Barcelona's Children Hospital San Juan de Dios, Member of the ERN EpiCARE, Spain; Epilepsy Research Centre (S.F.B.), University of Melbourne, Australia; Division of Pediatric Neurology (J.F.K.), Barrow Neurological Institute at Phoenix Children's Hospital; Hope for Hypothalamic Hamartomas (I.P.M., E.W., L.S.), Phoenix, AZ; Epilepsy Surgery Program (A.C.), Clinica de Epilepsia de Sao Paulo, Brazil; Department of Epidemiology (D.K.H.), Columbia University Medical Center, New York, NY; RTI International (B.L.K.), Rockville, MD; Department of Neurology (C.B.S.), Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and Epilepsy Center (A.S.-B.), Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Germany
| | - William D Gaillard
- From the Center for Neuroscience Research (N.T.C., W.D.G.), Children's National Hospital, The George Washington University School of Medicine, Washington, DC; UCL NIHR BRC Great Ormond Street Institute of Child Health (J.H.C.), Member of ERN-EpiCARE, London; Great Ormond Street Hospital for Children (J.H.C.), NHS Trust, London; Young Epilepsy (J.H.C.), Lingfield, Surrey, UK; Department of Pediatric Clinical Epileptology (A.A.), Sleep Disorders and Functional Neurology, Member of ERN-EpiCARE; HFME (A.A.), Hospices Civils de Lyon, France; Epilepsy Research Unit (A.A.), Barcelona's Children Hospital San Juan de Dios, Member of the ERN EpiCARE, Spain; Epilepsy Research Centre (S.F.B.), University of Melbourne, Australia; Division of Pediatric Neurology (J.F.K.), Barrow Neurological Institute at Phoenix Children's Hospital; Hope for Hypothalamic Hamartomas (I.P.M., E.W., L.S.), Phoenix, AZ; Epilepsy Surgery Program (A.C.), Clinica de Epilepsia de Sao Paulo, Brazil; Department of Epidemiology (D.K.H.), Columbia University Medical Center, New York, NY; RTI International (B.L.K.), Rockville, MD; Department of Neurology (C.B.S.), Beth-Israel Deaconess Medical Center, Harvard Medical School, Boston, MA; and Epilepsy Center (A.S.-B.), Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Germany
| | | |
Collapse
|
7
|
Marczenke M, Sunaga-Franze DY, Popp O, Althaus IW, Sauer S, Mertins P, Christ A, Allen BL, Willnow TE. GAS1 is required for NOTCH-dependent facilitation of SHH signaling in the ventral forebrain neuroepithelium. Development 2021; 148:272617. [PMID: 34698766 PMCID: PMC8627604 DOI: 10.1242/dev.200080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022]
Abstract
Growth arrest-specific 1 (GAS1) acts as a co-receptor to patched 1, promoting sonic hedgehog (SHH) signaling in the developing nervous system. GAS1 mutations in humans and animal models result in forebrain and craniofacial malformations, defects ascribed to a function for GAS1 in SHH signaling during early neurulation. Here, we confirm loss of SHH activity in the forebrain neuroepithelium in GAS1-deficient mice and in induced pluripotent stem cell-derived cell models of human neuroepithelial differentiation. However, our studies document that this defect can be attributed, at least in part, to a novel role for GAS1 in facilitating NOTCH signaling, which is essential to sustain a persistent SHH activity domain in the forebrain neuroepithelium. GAS1 directly binds NOTCH1, enhancing ligand-induced processing of the NOTCH1 intracellular domain, which drives NOTCH pathway activity in the developing forebrain. Our findings identify a unique role for GAS1 in integrating NOTCH and SHH signal reception in neuroepithelial cells, and they suggest that loss of GAS1-dependent NOTCH1 activation contributes to forebrain malformations in individuals carrying GAS1 mutations.
Collapse
Affiliation(s)
- Maike Marczenke
- Molecular Physiology, Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany.,Department of Biology, Chemistry and Pharmacy, Freie Universitaet Berlin, 12169 Berlin, Germany
| | | | - Oliver Popp
- Proteomics Platform, Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Irene W Althaus
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sascha Sauer
- Genomics Platform, Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Philipp Mertins
- Proteomics Platform, Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Annabel Christ
- Molecular Physiology, Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany
| | - Benjamin L Allen
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Thomas E Willnow
- Molecular Physiology, Max-Delbrueck-Center for Molecular Medicine, 13125 Berlin, Germany.,Department of Biomedicine, Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
8
|
Huang WK, Wong SZH, Pather SR, Nguyen PTT, Zhang F, Zhang DY, Zhang Z, Lu L, Fang W, Chen L, Fernandes A, Su Y, Song H, Ming GL. Generation of hypothalamic arcuate organoids from human induced pluripotent stem cells. Cell Stem Cell 2021; 28:1657-1670.e10. [PMID: 33961804 PMCID: PMC8419002 DOI: 10.1016/j.stem.2021.04.006] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/21/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022]
Abstract
Human brain organoids represent remarkable platforms for recapitulating features of human brain development and diseases. Existing organoid models do not resolve fine brain subregions, such as different nuclei in the hypothalamus. We report the generation of arcuate organoids (ARCOs) from human induced pluripotent stem cells (iPSCs) to model the development of the human hypothalamic arcuate nucleus. Single-cell RNA sequencing of ARCOs revealed significant molecular heterogeneity underlying different arcuate cell types, and machine learning-aided analysis based on the neonatal human hypothalamus single-nucleus transcriptome further showed a human arcuate nucleus molecular signature. We also explored ARCOs generated from Prader-Willi syndrome (PWS) patient iPSCs. These organoids exhibit aberrant differentiation and transcriptomic dysregulation similar to postnatal hypothalamus of PWS patients, indicative of cellular differentiation deficits and exacerbated inflammatory responses. Thus, patient iPSC-derived ARCOs represent a promising experimental model for investigating nucleus-specific features and disease-relevant mechanisms during early human arcuate development.
Collapse
Affiliation(s)
- Wei-Kai Huang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Program in Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Samuel Zheng Hao Wong
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Sarshan R Pather
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Phuong T T Nguyen
- Neuroscience Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Feng Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel Y Zhang
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhijian Zhang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lu Lu
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wanqi Fang
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Luyun Chen
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Analiese Fernandes
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yijing Su
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
9
|
Yoo S, Kim J, Lyu P, Hoang TV, Ma A, Trinh V, Dai W, Jiang L, Leavey P, Duncan L, Won JK, Park SH, Qian J, Brown SP, Blackshaw S. Control of neurogenic competence in mammalian hypothalamic tanycytes. SCIENCE ADVANCES 2021; 7:eabg3777. [PMID: 34049878 PMCID: PMC8163082 DOI: 10.1126/sciadv.abg3777] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/09/2021] [Indexed: 05/07/2023]
Abstract
Hypothalamic tanycytes, radial glial cells that share many features with neuronal progenitors, can generate small numbers of neurons in the postnatal hypothalamus, but the identity of these neurons and the molecular mechanisms that control tanycyte-derived neurogenesis are unknown. In this study, we show that tanycyte-specific disruption of the NFI family of transcription factors (Nfia/b/x) robustly stimulates tanycyte proliferation and tanycyte-derived neurogenesis. Single-cell RNA sequencing (scRNA-seq) and single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) analysis reveals that NFI (nuclear factor I) factors repress Sonic hedgehog (Shh) and Wnt signaling in tanycytes and modulation of these pathways blocks proliferation and tanycyte-derived neurogenesis in Nfia/b/x-deficient mice. Nfia/b/x-deficient tanycytes give rise to multiple mediobasal hypothalamic neuronal subtypes that can mature, fire action potentials, receive synaptic inputs, and selectively respond to changes in internal states. These findings identify molecular mechanisms that control tanycyte-derived neurogenesis, which can potentially be targeted to selectively remodel the hypothalamic neural circuitry that controls homeostatic physiological processes.
Collapse
Affiliation(s)
- Sooyeon Yoo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Pathology, Seoul National University Hospital, 71 Daehak-ro, Jongno-gu 03082, Republic of Korea
| | - Juhyun Kim
- Department of Psychiatry and Behavioral Science, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Pin Lyu
- Department of Ophthalmology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Thanh V Hoang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Alex Ma
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Vickie Trinh
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Weina Dai
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Lizhi Jiang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Patrick Leavey
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Leighton Duncan
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jae-Kyung Won
- Department of Pathology, Seoul National University Hospital, 71 Daehak-ro, Jongno-gu 03082, Republic of Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University Hospital, 71 Daehak-ro, Jongno-gu 03082, Republic of Korea
| | - Jiang Qian
- Department of Ophthalmology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Solange P Brown
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA.
- Department of Ophthalmology, Johns Hopkins University, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
- Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
10
|
Crane-Smith Z, Schoenebeck J, Graham KA, Devenney PS, Rose L, Ditzell M, Anderson E, Thomson JI, Klenin N, Kurrasch DM, Lettice LA, Hill RE. A Highly Conserved Shh Enhancer Coordinates Hypothalamic and Craniofacial Development. Front Cell Dev Biol 2021; 9:595744. [PMID: 33869166 PMCID: PMC8047142 DOI: 10.3389/fcell.2021.595744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 02/28/2021] [Indexed: 11/24/2022] Open
Abstract
Enhancers that are conserved deep in evolutionary time regulate characteristics held in common across taxonomic classes. Here, deletion of the highly conserved Shh enhancer SBE2 (Shh brain enhancer 2) in mouse markedly reduced Shh expression within the embryonic brain specifically in the rostral diencephalon; however, no abnormal anatomical phenotype was observed. Secondary enhancer activity was subsequently identified which likely mediates low levels of expression. In contrast, when crossing the SBE2 deletion with the Shh null allele, brain and craniofacial development were disrupted; thus, linking SBE2 regulated Shh expression to multiple defects and further enabling the study of the effects of differing levels of Shh on embryogenesis. Development of the hypothalamus, derived from the rostral diencephalon, was disrupted along both the anterior-posterior (AP) and the dorsal-ventral (DV) axes. Expression of DV patterning genes and subsequent neuronal population induction were particularly sensitive to Shh expression levels, demonstrating a novel morphogenic context for Shh. The role of SBE2, which is highlighted by DV gene expression, is to step-up expression of Shh above the minimal activity of the second enhancer, ensuring the necessary levels of Shh in a regional-specific manner. We also show that low Shh levels in the diencephalon disrupted neighbouring craniofacial development, including mediolateral patterning of the bones along the cranial floor and viscerocranium. Thus, SBE2 contributes to hypothalamic morphogenesis and ensures there is coordination with the formation of the adjacent midline cranial bones that subsequently protect the neural tissue.
Collapse
Affiliation(s)
- Zoe Crane-Smith
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Jeffrey Schoenebeck
- The Roslin Institute and Royal (Dick) School for Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Katy A Graham
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Paul S Devenney
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Lorraine Rose
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark Ditzell
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Eve Anderson
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Joseph I Thomson
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Natasha Klenin
- Department of Medical Genetics, Alberta Children's Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, Alberta Children's Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Laura A Lettice
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Robert E Hill
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
11
|
Diaz C, Puelles L. Developmental Genes and Malformations in the Hypothalamus. Front Neuroanat 2020; 14:607111. [PMID: 33324176 PMCID: PMC7726113 DOI: 10.3389/fnana.2020.607111] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022] Open
Abstract
The hypothalamus is a heterogeneous rostral forebrain region that regulates physiological processes essential for survival, energy metabolism, and reproduction, mainly mediated by the pituitary gland. In the updated prosomeric model, the hypothalamus represents the rostralmost forebrain, composed of two segmental regions (terminal and peduncular hypothalamus), which extend respectively into the non-evaginated preoptic telencephalon and the evaginated pallio-subpallial telencephalon. Complex genetic cascades of transcription factors and signaling molecules rule their development. Alterations of some of these molecular mechanisms acting during forebrain development are associated with more or less severe hypothalamic and pituitary dysfunctions, which may be associated with brain malformations such as holoprosencephaly or septo-optic dysplasia. Studies on transgenic mice with mutated genes encoding critical transcription factors implicated in hypothalamic-pituitary development are contributing to understanding the high clinical complexity of these pathologies. In this review article, we will analyze first the complex molecular genoarchitecture of the hypothalamus resulting from the activity of previous morphogenetic signaling centers and secondly some malformations related to alterations in genes implicated in the development of the hypothalamus.
Collapse
Affiliation(s)
- Carmen Diaz
- Department of Medical Sciences, School of Medicine and Institute for Research in Neurological Disabilities, University of Castilla-La Mancha, Albacete, Spain
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, University of Murcia, Murcia, Spain
| |
Collapse
|
12
|
Hedgehog Signaling Regulates Neurogenesis in the Larval and Adult Zebrafish Hypothalamus. eNeuro 2020; 7:ENEURO.0226-20.2020. [PMID: 33106384 PMCID: PMC7769882 DOI: 10.1523/eneuro.0226-20.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 12/15/2022] Open
Abstract
Neurogenesis is now known to play a role in adult hypothalamic function, yet the cell-cell mechanisms regulating this neurogenesis remain poorly understood. Here, we show that Hedgehog (Hh)/Gli signaling positively regulates hypothalamic neurogenesis in both larval and adult zebrafish and is necessary and sufficient for normal hypothalamic proliferation rates. Hh-responsive radial glia represent a relatively highly proliferative precursor population that gives rise to dopaminergic, serotonergic, and GABAergic neurons. In situ and transgenic reporter analyses revealed substantial heterogeneity in cell-cell signaling within the hypothalamic niche, with slow cycling Nestin-expressing cells residing among distinct and overlapping populations of Sonic Hh (Shh)-expressing, Hh-responsive, Notch-responsive, and Wnt-responsive radial glia. This work shows for the first time that Hh/Gli signaling is a key component of the complex cell-cell signaling environment that regulates hypothalamic neurogenesis throughout life.
Collapse
|
13
|
Lin YC, Wu CY, Hu CH, Pai TW, Chen YR, Wang WD. Integrated Hypoxia Signaling and Oxidative Stress in Developmental Neurotoxicity of Benzo[a]Pyrene in Zebrafish Embryos. Antioxidants (Basel) 2020; 9:antiox9080731. [PMID: 32796530 PMCID: PMC7464806 DOI: 10.3390/antiox9080731] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/08/2020] [Accepted: 08/09/2020] [Indexed: 12/14/2022] Open
Abstract
Benzo[a]pyrene (B[a]P) is a polycyclic aromatic hydrocarbon formed by the incomplete combustion of organic matter. Environmental B[a]P contamination poses a serious health risk to many organisms because the pollutant may negatively affect many physiological systems. As such, chronic exposure to B[a]P is known to lead to locomotor dysfunction and neurodegeneration in several organisms. In this study, we used the zebrafish model to delineate the acute toxic effects of B[a]P on the developing nervous system. We found that embryonic exposure of B[a]P downregulates shh and isl1, causing morphological hypoplasia in the telencephalon, ventral thalamus, hypothalamus, epiphysis and posterior commissure. Moreover, hypoxia-inducible factors (hif1a and hif2a) are repressed upon embryonic exposure of B[a]P, leading to reduced expression of the Hif-target genes, epo and survivin, which are associated with neural differentiation and maintenance. During normal embryogenesis, low-level oxidative stress regulates neuronal development and function. However, our experiments revealed that embryonic oxidative stress is greatly increased in B[a]P-treated embryos. The expression of catalase was decreased and sod1 expression increased in B[a]P-treated embryos. These transcriptional changes were coincident with increased embryonic levels of H2O2 and malondialdehyde, with the levels in B[a]P-treated fish similar to those in embryos treated with 120-μM H2O2. Together, our data suggest that reduced Hif signaling and increased oxidative stress are involved in B[a]P-induced acute neurotoxicity during embryogenesis.
Collapse
Affiliation(s)
- Yi-Chen Lin
- Department of Bioagricultural Sciences, National Chiayi University, Chiayi City 60004, Taiwan;
| | - Chang-Yi Wu
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan;
| | - Chin-Hwa Hu
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung 20224, Taiwan;
| | - Tun-Wen Pai
- Department of Computer Science and Engineering, National Taiwan Ocean University, Keelung 20224, Taiwan;
- Department of Computer Science and Information Engineering, National Taipei University of Technology, Taipei 10608, Taiwan
| | - Yet-Ran Chen
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 11529, Taiwan;
| | - Wen-Der Wang
- Department of Bioagricultural Sciences, National Chiayi University, Chiayi City 60004, Taiwan;
- Correspondence:
| |
Collapse
|
14
|
Chin JSR, Loomis CL, Albert LT, Medina-Trenche S, Kowalko J, Keene AC, Duboué ER. Analysis of stress responses in Astyanax larvae reveals heterogeneity among different populations. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2020; 334:486-496. [PMID: 32767504 DOI: 10.1002/jez.b.22987] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 06/15/2020] [Accepted: 06/18/2020] [Indexed: 11/07/2022]
Abstract
Stress responses are conserved physiological and behavioral outcomes as a result of facing potentially harmful stimuli, yet in pathological states, stress becomes debilitating. Stress responses vary considerably throughout the animal kingdom, but how these responses are shaped evolutionarily is unknown. The Mexican cavefish has emerged as a powerful system for examining genetic principles underlying behavioral evolution. Here, we demonstrate that cave Astyanax have reduced behavioral and physiological measures of stress when examined at larval stages. We also find increased expression of the glucocorticoid receptor, a repressible element of the neuroendocrine stress pathway. Additionally, we examine stress in three different cave populations, and find that some, but not all, show reduced stress measures. Together, these results reveal a mechanistic system by which cave-dwelling fish reduced stress, presumably to compensate for a predator poor environment.
Collapse
Affiliation(s)
- Jacqueline S R Chin
- Program in Neurogenetics, Florida Atlantic University, Jupiter, Florida.,Department of Biological Science, Florida Atlantic University, Jupiter, Florida
| | - Cody L Loomis
- Program in Neurogenetics, Florida Atlantic University, Jupiter, Florida.,Department of Biological Science, Florida Atlantic University, Jupiter, Florida
| | - Lydia T Albert
- Program in Neurogenetics, Florida Atlantic University, Jupiter, Florida.,Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, Florida
| | - Shirley Medina-Trenche
- Program in Neurogenetics, Florida Atlantic University, Jupiter, Florida.,Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, Florida
| | - Johanna Kowalko
- Program in Neurogenetics, Florida Atlantic University, Jupiter, Florida.,Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, Florida
| | - Alex C Keene
- Program in Neurogenetics, Florida Atlantic University, Jupiter, Florida.,Department of Biological Science, Florida Atlantic University, Jupiter, Florida
| | - Erik R Duboué
- Program in Neurogenetics, Florida Atlantic University, Jupiter, Florida.,Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, Florida
| |
Collapse
|
15
|
Le TL, Sribudiani Y, Dong X, Huber C, Kois C, Baujat G, Gordon CT, Mayne V, Galmiche L, Serre V, Goudin N, Zarhrate M, Bole-Feysot C, Masson C, Nitschké P, Verheijen FW, Pais L, Pelet A, Sadedin S, Pugh JA, Shur N, White SM, El Chehadeh S, Christodoulou J, Cormier-Daire V, Hofstra RMW, Lyonnet S, Tan TY, Attié-Bitach T, Kerstjens-Frederikse WS, Amiel J, Thomas S. Bi-allelic Variations of SMO in Humans Cause a Broad Spectrum of Developmental Anomalies Due to Abnormal Hedgehog Signaling. Am J Hum Genet 2020; 106:779-792. [PMID: 32413283 DOI: 10.1016/j.ajhg.2020.04.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 04/08/2020] [Indexed: 12/12/2022] Open
Abstract
The evolutionarily conserved hedgehog (Hh) pathway is essential for organogenesis and plays critical roles in postnatal tissue maintenance and renewal. A unique feature of the vertebrate Hh pathway is that signal transduction requires the primary cilium (PC) where major pathway components are dynamically enriched. These factors include smoothened (SMO) and patched, which constitute the core reception system for sonic hedgehog (SHH) as well as GLI transcription factors, the key mediators of the pathway. Here, we report bi-allelic loss-of-function variations in SMO in seven individuals from five independent families; these variations cause a wide phenotypic spectrum of developmental anomalies affecting the brain (hypothalamic hamartoma and microcephaly), heart (atrioventricular septal defect), skeleton (postaxial polydactyly, narrow chest, and shortening of long bones), and enteric nervous system (aganglionosis). Cells derived from affected individuals showed normal ciliogenesis but severely altered Hh-signal transduction as a result of either altered PC trafficking or abnormal activation of the pathway downstream of SMO. In addition, Hh-independent GLI2 accumulation at the PC tip in cells from the affected individuals suggests a potential function of SMO in regulating basal ciliary trafficking of GLI2 when the pathway is off. Thus, loss of SMO function results in abnormal PC dynamics of key components of the Hh signaling pathway and leads to a large continuum of malformations in humans.
Collapse
Affiliation(s)
- Thuy-Linh Le
- Université de Paris, Imagine Institute, Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, 75015 Paris, France
| | - Yunia Sribudiani
- Department of Clinical Genetics, Erasmus Medical Center, 3015 GD Rotterdam, the Netherlands; Department of Biomedical Sciences, Division of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Padjadjaran, Bandung 40132, Indonesia
| | - Xiaomin Dong
- Brain and Mitochondrial Research Group, Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville VIC 3052, Australia; Department of Paediatrics, University of Melbourne, Melbourne, 3010 Victoria, Australia
| | - Céline Huber
- Université de Paris, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, 75015 Paris, France
| | - Chelsea Kois
- Albany Medical Center, 43 New Scotland Ave, Albany, NY 12208, USA
| | - Geneviève Baujat
- Université de Paris, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, 75015 Paris, France; Fédération de Génétique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Christopher T Gordon
- Université de Paris, Imagine Institute, Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, 75015 Paris, France
| | - Valerie Mayne
- Department of Medical Imaging, Royal Children's Hospital, Melbourne, Australia 3052
| | - Louise Galmiche
- Department of Pathology, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Valérie Serre
- Université de Paris, Institut Jacques Monod, UMR7592 CNRS, 15 Rue Hélène Brion, 75013 Paris, France
| | - Nicolas Goudin
- Université de Paris, Imagine Institute, Cell Imaging, INSERM UMR 1163, 75015 Paris, France
| | - Mohammed Zarhrate
- Université de Paris, Imagine Institute, Structure Fédérative de Recherche Necker, Genomic Platform, INSERM UMR 1163 and INSERM US24, Centre National de la Recherche Scientifique UMS3633, 75015 Paris, France
| | - Christine Bole-Feysot
- Université de Paris, Imagine Institute, Structure Fédérative de Recherche Necker, Genomic Platform, INSERM UMR 1163 and INSERM US24, Centre National de la Recherche Scientifique UMS3633, 75015 Paris, France
| | - Cécile Masson
- Université de Paris, Imagine Institute, Bioinformatics Platform, INSERM UMR 1163, 75015 Paris, France
| | - Patrick Nitschké
- Université de Paris, Imagine Institute, Bioinformatics Platform, INSERM UMR 1163, 75015 Paris, France
| | - Frans W Verheijen
- Department of Clinical Genetics, Erasmus Medical Center, 3015 GD Rotterdam, the Netherlands
| | - Lynn Pais
- Center for Mendelian Genomics, Broad Institute of MIT and Harvard, 415 Main St, Cambridge, MA 02142, USA
| | - Anna Pelet
- Université de Paris, Imagine Institute, Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, 75015 Paris, France
| | - Simon Sadedin
- Brain and Mitochondrial Research Group, Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville VIC 3052, Australia; Department of Paediatrics, University of Melbourne, Melbourne, 3010 Victoria, Australia
| | - John A Pugh
- Albany Medical Center, 43 New Scotland Ave, Albany, NY 12208, USA
| | - Natasha Shur
- Children's National, 111 Michigan Ave NW, Washington, D.C. 20010, USA
| | - Susan M White
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute and Department of Pediatrics, University of Melbourne, Melbourne, Australia 3052
| | - Salima El Chehadeh
- Service de Génétique Médicale, Hôpital de Hautepierre, 67098 Strasbourg, France
| | - John Christodoulou
- Brain and Mitochondrial Research Group, Murdoch Children's Research Institute, Royal Children's Hospital, 50 Flemington Rd, Parkville VIC 3052, Australia; Department of Paediatrics, University of Melbourne, Melbourne, 3010 Victoria, Australia
| | - Valérie Cormier-Daire
- Université de Paris, Imagine Institute, Laboratory of Molecular and Physiopathological Bases of Osteochondrodysplasia, INSERM UMR 1163, 75015 Paris, France; Fédération de Génétique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - R M W Hofstra
- Department of Clinical Genetics, Erasmus Medical Center, 3015 GD Rotterdam, the Netherlands
| | - Stanislas Lyonnet
- Université de Paris, Imagine Institute, Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, 75015 Paris, France; Fédération de Génétique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Tiong Yang Tan
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute and Department of Pediatrics, University of Melbourne, Melbourne, Australia 3052
| | - Tania Attié-Bitach
- Fédération de Génétique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, 75015 Paris, France; Université de Paris, Imagine Institute, Laboratory of Genetics and Development of the Cerebral Cortex, INSERM UMR 1163, 75015 Paris, France
| | | | - Jeanne Amiel
- Université de Paris, Imagine Institute, Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, 75015 Paris, France; Fédération de Génétique, Hôpital Necker-Enfants Malades, Assistance Publique Hôpitaux de Paris, 75015 Paris, France
| | - Sophie Thomas
- Université de Paris, Imagine Institute, Laboratory of Embryology and Genetics of Malformations, INSERM UMR 1163, 75015 Paris, France.
| |
Collapse
|
16
|
Fu T, Pearson C, Towers M, Placzek M. Development of the basal hypothalamus through anisotropic growth. J Neuroendocrinol 2019; 31:e12727. [PMID: 31050853 PMCID: PMC6563594 DOI: 10.1111/jne.12727] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/26/2019] [Accepted: 04/28/2019] [Indexed: 02/06/2023]
Abstract
The adult hypothalamus is subdivided into distinct domains: pre-optic, anterior, tuberal and mammillary. Each domain harbours an array of neurones that act together to regulate homeostasis. The embryonic origins and the development of hypothalamic neurones, however, remain enigmatic. Here, we summarise recent studies in model organisms that challenge current views of hypothalamic development, which traditionally have attempted to map adult domains to correspondingly located embryonic domains. Instead, new studies indicate that hypothalamic neurones arise from progenitor cells that undergo anisotropic growth, expanding to a greater extent than other progenitors, and grow in different dimensions. We describe in particular how a multipotent Shh/ Fgf10-expressing progenitor population gives rise to progenitors throughout the basal hypothalamus that grow anisotropically and sequentially: first, a subset displaced rostrally give rise to anterior-ventral/tuberal neuronal progenitors; then a subset displaced caudally give rise to mammillary neuronal progenitors; and, finally, a subset(s) displaced ventrally give rise to tuberal infundibular glial progenitors. As this occurs, stable populations of Shh+ive and Fgf10+ive progenitors form. We describe current understanding of the mechanisms that induce Shh+ive /Fgf10+ive progenitors and begin to direct their differentiation to anterior-ventral/tuberal neuronal progenitors, mammillary neuronal progenitors and tuberal infundibular progenitors. Taken together, these studies suggest a new model for hypothalamic development that we term the "anisotropic growth model". We discuss the implications of the model for understanding the origins of adult hypothalamic neurones.
Collapse
Affiliation(s)
- Travis Fu
- Department of Biomedical ScienceBateson CentreUniversity of SheffieldSheffieldUK
| | - Caroline Pearson
- Department of Biomedical ScienceBateson CentreUniversity of SheffieldSheffieldUK
| | - Matthew Towers
- Department of Biomedical ScienceBateson CentreUniversity of SheffieldSheffieldUK
| | - Marysia Placzek
- Department of Biomedical ScienceBateson CentreUniversity of SheffieldSheffieldUK
| |
Collapse
|
17
|
Zhu J, Wang C, Gao X, Zhu J, Wang L, Cao S, Wu Q, Qiao S, Zhang Z, Li L. Comparative effects of mercury chloride and methylmercury exposure on early neurodevelopment in zebrafish larvae. RSC Adv 2019; 9:10766-10775. [PMID: 35515286 PMCID: PMC9062475 DOI: 10.1039/c9ra00770a] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/31/2019] [Indexed: 12/21/2022] Open
Abstract
Mercury (Hg) is a ubiquitous environmental toxicant with important public health implications. Hg causes neurotoxicity through astrocytes, Ca2+, neurotransmitters, mitochondrial damage, elevations of reactive oxygen species and post-translational modifications. However, the similarities and differences between the neurotoxic mechanisms caused by different chemical forms of Hg remain unclear. Zebrafish embryos were exposed to methylmercury (MeHgCl) or mercury chloride (HgCl2) (0, 4, 40, 400 nM) up for 96 h. HgCl2 exposure could significantly decrease survival rate, body length and eye size, delay the hatching period, induce tail bending and reduce the locomotor activity, and these effects were aggravated in the MeHgCl group. The compounds could increase the number of apoptotic cells in the brain and downregulate the expression of Shha, Ngn1 and Nrd, which contribute to early nervous development. The underlying mechanisms were investigated by metabolomics data. Galactose metabolism, tyrosine metabolism and starch and sucrose metabolism pathways were disturbed after HgCl2 or MeHgCl exposure. In addition, the levels of three neurotransmitters including tyrosine, dopamine and tryptophan were reduced after HgCl2 or MeHgCl exposure. Oxidative stress is related to metabolite changes, such as changes in the putrescine, niacinamide and uric acid contents in the HgCl2 group, and squalene in the MeHgCl group. These data indicated that downregulation of these genes and abnormal metabolic profile and pathways contribute to the neurotoxicity of HgCl2 and MeHgCl.
Collapse
Affiliation(s)
- Jun Zhu
- Center for Global Health, School of Public Health, Nanjing Medical University 101 Longmian Avenue Nanjing Jiangsu 211166 P. R. China +86-25-8686-8499 +86-25-8686-8402 +86-25-8686-8404
| | - Chundan Wang
- Center for Global Health, School of Public Health, Nanjing Medical University 101 Longmian Avenue Nanjing Jiangsu 211166 P. R. China +86-25-8686-8499 +86-25-8686-8402 +86-25-8686-8404
| | - Xingsu Gao
- Center for Global Health, School of Public Health, Nanjing Medical University 101 Longmian Avenue Nanjing Jiangsu 211166 P. R. China +86-25-8686-8499 +86-25-8686-8402 +86-25-8686-8404
| | - Jiansheng Zhu
- Key Lab of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University 101 Longmian Avenue Nanjing Jiangsu 211166 P. R. China
| | - Li Wang
- Center for Global Health, School of Public Health, Nanjing Medical University 101 Longmian Avenue Nanjing Jiangsu 211166 P. R. China +86-25-8686-8499 +86-25-8686-8402 +86-25-8686-8404
| | - Shuyuan Cao
- Center for Global Health, School of Public Health, Nanjing Medical University 101 Longmian Avenue Nanjing Jiangsu 211166 P. R. China +86-25-8686-8499 +86-25-8686-8402 +86-25-8686-8404
| | - Qian Wu
- Center for Global Health, School of Public Health, Nanjing Medical University 101 Longmian Avenue Nanjing Jiangsu 211166 P. R. China +86-25-8686-8499 +86-25-8686-8402 +86-25-8686-8404
| | - Shanlei Qiao
- Center for Global Health, School of Public Health, Nanjing Medical University 101 Longmian Avenue Nanjing Jiangsu 211166 P. R. China +86-25-8686-8499 +86-25-8686-8402 +86-25-8686-8404
| | - Zhan Zhang
- Center for Global Health, School of Public Health, Nanjing Medical University 101 Longmian Avenue Nanjing Jiangsu 211166 P. R. China +86-25-8686-8499 +86-25-8686-8402 +86-25-8686-8404
| | - Lei Li
- Center for Global Health, School of Public Health, Nanjing Medical University 101 Longmian Avenue Nanjing Jiangsu 211166 P. R. China +86-25-8686-8499 +86-25-8686-8402 +86-25-8686-8404
| |
Collapse
|