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
|
Buck SA, Rubin SA, Kunkhyen T, Treiber CD, Xue X, Fenno LE, Mabry SJ, Sundar VR, Yang Z, Shah D, Ketchesin KD, Becker-Krail DD, Vasylieva I, Smith MC, Weisel FJ, Wang W, Erickson-Oberg MQ, O’Leary EI, Aravind E, Ramakrishnan C, Kim YS, Wu Y, Quick M, Coleman JA, MacDonald WA, Elbakri R, De Miranda BR, Palladino MJ, McCabe BD, Fish KN, Seney ML, Rayport S, Mingote S, Deisseroth K, Hnasko TS, Awatramani R, Watson AM, Waddell S, Cheetham CEJ, Logan RW, Freyberg Z. Sexually dimorphic mechanisms of VGLUT-mediated protection from dopaminergic neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.02.560584. [PMID: 37873436 PMCID: PMC10592912 DOI: 10.1101/2023.10.02.560584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Parkinson's disease (PD) targets some dopamine (DA) neurons more than others. Sex differences offer insights, with females more protected from DA neurodegeneration. The mammalian vesicular glutamate transporter VGLUT2 and Drosophila ortholog dVGLUT have been implicated as modulators of DA neuron resilience. However, the mechanisms by which VGLUT2/dVGLUT protects DA neurons remain unknown. We discovered DA neuron dVGLUT knockdown increased mitochondrial reactive oxygen species in a sexually dimorphic manner in response to depolarization or paraquat-induced stress, males being especially affected. DA neuron dVGLUT also reduced ATP biosynthetic burden during depolarization. RNA sequencing of VGLUT+ DA neurons in mice and flies identified candidate genes that we functionally screened to further dissect VGLUT-mediated DA neuron resilience across PD models. We discovered transcription factors modulating dVGLUT-dependent DA neuroprotection and identified dj-1β as a regulator of sex-specific DA neuron dVGLUT expression. Overall, VGLUT protects DA neurons from PD-associated degeneration by maintaining mitochondrial health.
Collapse
Affiliation(s)
- Silas A. Buck
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sophie A. Rubin
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tenzin Kunkhyen
- Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Christoph D. Treiber
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - Xiangning Xue
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Lief E. Fenno
- Departments of Psychiatry and Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Samuel J. Mabry
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Varun R. Sundar
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Zilu Yang
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Divia Shah
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kyle D. Ketchesin
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Darius D. Becker-Krail
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Iaroslavna Vasylieva
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Megan C. Smith
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Florian J. Weisel
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Wenjia Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - M. Quincy Erickson-Oberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Emma I. O’Leary
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Eshan Aravind
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Charu Ramakrishnan
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Yoon Seok Kim
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Yanying Wu
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | - Matthias Quick
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Jonathan A. Coleman
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | | | - Rania Elbakri
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Briana R. De Miranda
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michael J. Palladino
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
- Pittsburgh Institute of Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Brian D. McCabe
- Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Kenneth N. Fish
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Marianne L. Seney
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Stephen Rayport
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
| | - Susana Mingote
- Department of Psychiatry, Columbia University, New York, NY 10032, USA
- Department of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA
- Neuroscience Initiative, Advanced Science Research Center, Graduate Center of The City University of New York, New York, NY 10031, USA
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA 94305, USA
| | - Thomas S. Hnasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
- Research Service, VA San Diego Healthcare System, San Diego, CA 92161, USA
| | | | - Alan M. Watson
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Scott Waddell
- Centre for Neural Circuits & Behaviour, University of Oxford, Oxford OX1 3TA, UK
| | | | - Ryan W. Logan
- Department of Psychiatry, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Zachary Freyberg
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
3
|
MacKay H, Gunasekara CJ, Yam KY, Srisai D, Yalamanchili HK, Li Y, Chen R, Coarfa C, Waterland RA. Sex-specific epigenetic development in the mouse hypothalamic arcuate nucleus pinpoints human genomic regions associated with body mass index. SCIENCE ADVANCES 2022; 8:eabo3991. [PMID: 36170368 PMCID: PMC9519050 DOI: 10.1126/sciadv.abo3991] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 08/10/2022] [Indexed: 06/16/2023]
Abstract
Recent genome-wide association studies corroborate classical research on developmental programming indicating that obesity is primarily a neurodevelopmental disease strongly influenced by nutrition during critical ontogenic windows. Epigenetic mechanisms regulate neurodevelopment; however, little is known about their role in establishing and maintaining the brain's energy balance circuitry. We generated neuron and glia methylomes and transcriptomes from male and female mouse hypothalamic arcuate nucleus, a key site for energy balance regulation, at time points spanning the closure of an established critical window for developmental programming of obesity risk. We find that postnatal epigenetic maturation is markedly cell type and sex specific and occurs in genomic regions enriched for heritability of body mass index in humans. Our results offer a potential explanation for both the limited ontogenic windows for and sex differences in sensitivity to developmental programming of obesity and provide a rich resource for epigenetic analyses of developmental programming of energy balance.
Collapse
Affiliation(s)
- Harry MacKay
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Chathura J. Gunasekara
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Kit-Yi Yam
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Dollada Srisai
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Hari Krishna Yalamanchili
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s Hospital, Houston, TX, USA
| | - Yumei Li
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Rui Chen
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Cristian Coarfa
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, USA
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Robert A. Waterland
- USDA/ARS Children’s Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
4
|
Benevento M, Hökfelt T, Harkany T. Ontogenetic rules for the molecular diversification of hypothalamic neurons. Nat Rev Neurosci 2022; 23:611-627. [PMID: 35906427 DOI: 10.1038/s41583-022-00615-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2022] [Indexed: 11/09/2022]
Abstract
The hypothalamus is an evolutionarily conserved endocrine interface that, among other roles, links central homeostatic control to adaptive bodily responses by releasing hormones and neuropeptides from its many neuronal subtypes. In its preoptic, anterior, tuberal and mammillary subdivisions, a kaleidoscope of magnocellular and parvocellular neuroendocrine command neurons, local-circuit neurons, and neurons that project to extrahypothalamic areas are intermingled in partially overlapping patches of nuclei. Molecular fingerprinting has produced data of unprecedented mass and depth to distinguish and even to predict the synaptic and endocrine competences, connectivity and stimulus selectivity of many neuronal modalities. These new insights support eminent studies from the past century but challenge others on the molecular rules that shape the developmental segregation of hypothalamic neuronal subtypes and their use of morphogenic cues for terminal differentiation. Here, we integrate single-cell RNA sequencing studies with those of mouse genetics and endocrinology to describe key stages of hypothalamus development, including local neurogenesis, the direct terminal differentiation of glutamatergic neurons, transition cascades for GABAergic and GABAergic cell-derived dopamine cells, waves of local neuronal migration, and sequential enrichment in neuropeptides and hormones. We particularly emphasize how transcription factors determine neuronal identity and, consequently, circuit architecture, and whether their deviations triggered by environmental factors and hormones provoke neuroendocrine illnesses.
Collapse
Affiliation(s)
- Marco Benevento
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Tomas Hökfelt
- Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria. .,Department of Neuroscience, Biomedicum 7D, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
5
|
Croizier S, Bouret SG. Molecular Control of the Development of Hypothalamic Neurons Involved in Metabolic Regulation. J Chem Neuroanat 2022; 123:102117. [DOI: 10.1016/j.jchemneu.2022.102117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 03/03/2022] [Accepted: 06/03/2022] [Indexed: 10/18/2022]
|
6
|
Bouret SG. Developmental programming of hypothalamic melanocortin circuits. Exp Mol Med 2022; 54:403-413. [PMID: 35474338 PMCID: PMC9076880 DOI: 10.1038/s12276-021-00625-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 01/14/2023] Open
Abstract
The melanocortin system plays a critical role in the central regulation of food intake and energy balance. This system consists of neurons producing pro-opiomelanocortin (POMC), melanocortin receptors (MC4Rs), and the endogenous antagonist agouti-related peptide (AgRP). Pomc and Mc4r deficiency in rodents and humans causes early onset of obesity, whereas a loss of Agrp function is associated with leanness. Accumulating evidence shows that many chronic diseases, including obesity, might originate during early life. The melanocortin system develops during a relatively long period beginning during embryonic life with the birth of POMC and AgRP neurons and continuing postnatally with the assembly of their neuronal circuitry. The development of the melanocortin system requires the tight temporal regulation of molecular factors, such as transcription factors and axon guidance molecules, and cellular mechanisms, such as autophagy. It also involves a complex interplay of endocrine and nutritional factors. The disruption of one or more of these developmental factors can lead to abnormal maturation and function of the melanocortin system and has profound metabolic consequences later in life.
Collapse
Affiliation(s)
- Sebastien G Bouret
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition Research Center, UMR-S 1172, Lille, 59000, France.
- University of Lille, FHU 1,000 Days for Health, Lille, 59000, France.
| |
Collapse
|
7
|
Yaghmaeian Salmani B, Balderson B, Bauer S, Ekman H, Starkenberg A, Perlmann T, Piper M, Bodén M, Thor S. Selective requirement for polycomb repressor complex 2 in the generation of specific hypothalamic neuronal subtypes. Development 2022; 149:274592. [PMID: 35245348 PMCID: PMC8959139 DOI: 10.1242/dev.200076] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 01/18/2022] [Indexed: 11/20/2022]
Abstract
The hypothalamus displays staggering cellular diversity, chiefly established during embryogenesis by the interplay of several signalling pathways and a battery of transcription factors. However, the contribution of epigenetic cues to hypothalamus development remains unclear. We mutated the polycomb repressor complex 2 gene Eed in the developing mouse hypothalamus, which resulted in the loss of H3K27me3, a fundamental epigenetic repressor mark. This triggered ectopic expression of posteriorly expressed regulators (e.g. Hox homeotic genes), upregulation of cell cycle inhibitors and reduced proliferation. Surprisingly, despite these effects, single cell transcriptomic analysis revealed that most neuronal subtypes were still generated in Eed mutants. However, we observed an increase in glutamatergic/GABAergic double-positive cells, as well as loss/reduction of dopamine, hypocretin and Tac2-Pax6 neurons. These findings indicate that many aspects of the hypothalamic gene regulatory flow can proceed without the key H3K27me3 epigenetic repressor mark, but points to a unique sensitivity of particular neuronal subtypes to a disrupted epigenomic landscape. Summary: Polycomb repressor complex 2 inactivation results in selective effects on mouse hypothalamic development, increasing glutamatergic/GABA cells, while reducing dopamine, Hcrt and Tac2-Pax6 cells.
Collapse
Affiliation(s)
- Behzad Yaghmaeian Salmani
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
- Department of Cell and Molecular Biology, Karolinska Institute, SE-17177 Stockholm, Sweden
| | - Brad Balderson
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Susanne Bauer
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
| | - Helen Ekman
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
| | - Annika Starkenberg
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institute, SE-17177 Stockholm, Sweden
| | - Michael Piper
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Mikael Bodén
- School of Chemistry and Molecular Biosciences, University of Queensland, St Lucia, QLD 4072, Australia
| | - Stefan Thor
- Department of Clinical and Experimental Medicine, Linkoping University, SE-58185 Linkoping, Sweden
- School of Biomedical Sciences, University of Queensland, St Lucia, QLD 4072, Australia
| |
Collapse
|
8
|
Pahl MC, Doege CA, Hodge KM, Littleton SH, Leonard ME, Lu S, Rausch R, Pippin JA, De Rosa MC, Basak A, Bradfield JP, Hammond RK, Boehm K, Berkowitz RI, Lasconi C, Su C, Chesi A, Johnson ME, Wells AD, Voight BF, Leibel RL, Cousminer DL, Grant SFA. Cis-regulatory architecture of human ESC-derived hypothalamic neuron differentiation aids in variant-to-gene mapping of relevant complex traits. Nat Commun 2021; 12:6749. [PMID: 34799566 PMCID: PMC8604959 DOI: 10.1038/s41467-021-27001-4] [Citation(s) in RCA: 6] [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: 10/02/2020] [Accepted: 10/27/2021] [Indexed: 11/09/2022] Open
Abstract
The hypothalamus regulates metabolic homeostasis by influencing behavior and endocrine systems. Given its role governing key traits, such as body weight and reproductive timing, understanding the genetic regulation of hypothalamic development and function could yield insights into disease pathogenesis. However, given its inaccessibility, studying human hypothalamic gene regulation has proven challenging. To address this gap, we generate a high-resolution chromatin architecture atlas of an established embryonic stem cell derived hypothalamic-like neuron model across three stages of in vitro differentiation. We profile accessible chromatin and identify physical contacts between gene promoters and putative cis-regulatory elements to characterize global regulatory landscape changes during hypothalamic differentiation. Next, we integrate these data with GWAS loci for various complex traits, identifying multiple candidate effector genes. Our results reveal common target genes for these traits, potentially affecting core developmental pathways. Our atlas will enable future efforts to determine hypothalamic mechanisms influencing disease susceptibility.
Collapse
Affiliation(s)
- Matthew C Pahl
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Claudia A Doege
- Department of Pathology, Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Kenyaita M Hodge
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Sheridan H Littleton
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Michelle E Leonard
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Sumei Lu
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Rick Rausch
- Department of Pediatrics, Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - James A Pippin
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Maria Caterina De Rosa
- Department of Pediatrics, Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Alisha Basak
- Department of Pediatrics, Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Columbia University, Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Jonathan P Bradfield
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Reza K Hammond
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Keith Boehm
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Robert I Berkowitz
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Chiara Lasconi
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Chun Su
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Matthew E Johnson
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Andrew D Wells
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Benjamin F Voight
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Rudolph L Leibel
- Division of Molecular Genetics (Pediatrics) and the Naomi Berrie Diabetes Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Diana L Cousminer
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA
- GSK, Human Genetics and Computational Biology, 1250 South Collegeville Road, Collegeville, PA, 19426, USA
| | - Struan F A Grant
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA.
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
9
|
Zhang Q, Zhang L, Huang Y, Ma P, Mao B, Ding YQ, Song NN. Satb2 regulates the development of dopaminergic neurons in the arcuate nucleus by Dlx1. Cell Death Dis 2021; 12:879. [PMID: 34564702 PMCID: PMC8464595 DOI: 10.1038/s41419-021-04175-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 09/02/2021] [Accepted: 09/15/2021] [Indexed: 12/21/2022]
Abstract
Dopaminergic (DA) neurons in the arcuate nucleus (ARC) of the hypothalamus play essential roles in the secretion of prolactin and the regulation of energy homeostasis. However, the gene regulatory network responsible for the development of the DA neurons remains poorly understood. Here we report that the transcription factor special AT-rich binding protein 2 (Satb2) is required for the development of ARC DA neurons. Satb2 is expressed in a large proportion of DA neurons without colocalization with proopiomelanocortin (POMC), orexigenic agouti-related peptide (AgRP), neuropeptide-Y (NPY), somatostatin (Sst), growth hormone-releasing hormone (GHRH), or galanin in the ARC. Nestin-Cre;Satb2flox/flox (Satb2 CKO) mice show a reduced number of ARC DA neurons with unchanged numbers of the other types of ARC neurons, and exhibit an increase of serum prolactin level and an elevated metabolic rate. The reduction of ARC DA neurons in the CKO mice is observed at an embryonic stage and Dlx1 is identified as a potential downstream gene of Satb2 in regulating the development of ARC DA neurons. Together, our study demonstrates that Satb2 plays a critical role in the gene regulatory network directing the development of DA neurons in ARC.
Collapse
Affiliation(s)
- Qiong Zhang
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Lei Zhang
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ying Huang
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
| | - Pengcheng Ma
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Bingyu Mao
- State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Yu-Qiang Ding
- Department of Laboratory Animal Science, Fudan University, Shanghai, China
- Key Laboratory of Arrhythmias, Ministry of Education of China, East Hospital, and Department of Anatomy and Neurobiology, Tongji University School of Medicine, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China
| | - Ning-Ning Song
- Department of Laboratory Animal Science, Fudan University, Shanghai, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Ma T, Wong SZH, Lee B, Ming GL, Song H. Decoding neuronal composition and ontogeny of individual hypothalamic nuclei. Neuron 2021; 109:1150-1167.e6. [PMID: 33600763 DOI: 10.1016/j.neuron.2021.01.026] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 10/10/2020] [Accepted: 01/26/2021] [Indexed: 01/30/2023]
Abstract
The hypothalamus plays crucial roles in regulating endocrine, autonomic, and behavioral functions via its diverse nuclei and neuronal subtypes. The developmental mechanisms underlying ontogenetic establishment of different hypothalamic nuclei and generation of neuronal diversity remain largely unknown. Here, we show that combinatorial T-box 3 (TBX3), orthopedia homeobox (OTP), and distal-less homeobox (DLX) expression delineates all arcuate nucleus (Arc) neurons and defines four distinct subpopulations, whereas combinatorial NKX2.1/SF1 and OTP/DLX expression identifies ventromedial hypothalamus (VMH) and tuberal nucleus (TuN) neuronal subpopulations, respectively. Developmental analysis indicates that all four Arc subpopulations are mosaically and simultaneously generated from embryonic Arc progenitors, whereas glutamatergic VMH neurons and GABAergic TuN neurons are sequentially generated from common embryonic VMH progenitors. Moreover, clonal lineage-tracing analysis reveals that diverse lineages from multipotent radial glia progenitors orchestrate Arc and VMH-TuN establishment. Together, our study reveals cellular mechanisms underlying generation and organization of diverse neuronal subtypes and ontogenetic establishment of individual nuclei in the mammalian hypothalamus.
Collapse
Affiliation(s)
- Tong Ma
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Samuel Zheng Hao Wong
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; The Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Bora Lee
- Center for Neuroscience, Korea Institute of Science and Technology, Seoul 02792, Korea
| | - 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
| | - 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 Epigenetic Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
11
|
Ammari R, Broberger C. Pre- and post-synaptic modulation by GABA B receptors of rat neuroendocrine dopamine neurones. J Neuroendocrinol 2020; 32:e12881. [PMID: 32803906 DOI: 10.1111/jne.12881] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 05/22/2020] [Accepted: 06/05/2020] [Indexed: 12/17/2022]
Abstract
The secretion of prolactin from the pituitary is negatively controlled by tuberoinfundibular dopamine (TIDA) neurones. The electrical properties of TIDA cells have recently been identified as a modulatory target of neurotransmitters and hormones in the lactotrophic axis. The role of the GABAB receptor in this control has received little attention, yet is of particular interest because it may act as a TIDA neurone autoreceptor. Here, this issue was explored in a spontaneously active rat TIDA in vitro slice preparation using whole-cell recordings. Application of the GABAB receptor agonist, baclofen, dose-dependently slowed down or abolished the network oscillations typical of this preparation. Pharmacological manipulations identify the underlying mechanism as an outward current mediated by G-protein-coupled inwardly rectifying K+ -like channels. In addition to this postsynaptic modulation, we describe a presynaptic modulation where GABAB receptors restrain the release of glutamate and GABA onto TIDA neurones. Our data identify both pre- and postsynaptic modulation of TIDA neurones by GABAB receptors that may play a role in the neuronal network control of pituitary prolactin secretion and lactation.
Collapse
Affiliation(s)
- Rachida Ammari
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Christian Broberger
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| |
Collapse
|
12
|
Marshall CJ, Prescott M, Campbell RE. Investigating the NPY/AgRP/GABA to GnRH Neuron Circuit in Prenatally Androgenized PCOS-Like Mice. J Endocr Soc 2020; 4:bvaa129. [PMID: 33094210 PMCID: PMC7566551 DOI: 10.1210/jendso/bvaa129] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 08/24/2020] [Indexed: 12/14/2022] Open
Abstract
Polycystic ovary syndrome (PCOS), the most common form of anovulatory infertility, is associated with altered signaling within the hormone-sensitive neuronal network that regulates gonadotropin-releasing hormone (GnRH) neurons, leading to a pathological increase in GnRH secretion. Circuit remodeling is evident between GABAergic neurons in the arcuate nucleus (ARN) and GnRH neurons in a murine model of PCOS. One-third of ARN GABA neurons co-express neuropeptide Y (NPY), which has a known yet complex role in regulating GnRH neurons and reproductive function. Here, we investigated whether the NPY-expressing subpopulation (NPYARN) of ARN GABA neurons (GABAARN) is also affected in prenatally androgenized (PNA) PCOS-like NPYARN reporter mice [Agouti-related protein (AgRP)-Cre;τGFP]. PCOS-like mice and controls were generated by exposure to di-hydrotestosterone or vehicle (VEH) in late gestation. τGFP-expressing NPYARN neuron fiber appositions with GnRH neurons and gonadal steroid hormone receptor expression in τGFP-expressing NPYARN neurons were assessed using confocal microscopy. Although GnRH neurons received abundant close contacts from τGFP-expressing NPYARN neuron fibers, the number and density of putative inputs was not affected by prenatal androgen excess. NPYARN neurons did not co-express progesterone receptor or estrogen receptor α in either PNA or VEH mice. However, the proportion of NPYARN neurons co-expressing the androgen receptor was significantly elevated in PNA mice. Therefore, NPYARN neurons are not remodeled by prenatal androgen excess like the wider GABAARN population, indicating GABA-to-GnRH neuron circuit remodeling occurs in a presently unidentified non-NPY/AgRP population of GABAARN neurons. NPYARN neurons do, however, show independent changes in the form of elevated androgen sensitivity.
Collapse
Affiliation(s)
- Christopher J Marshall
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Melanie Prescott
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Rebecca E Campbell
- Centre for Neuroendocrinology and Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
13
|
Ding W, Zhang C, Wang B, Zhou X, Sun L, Zhong S, Liu J, Zhang J, Wang X, Wu Q. Loss of the centrosomal protein Cenpj leads to dysfunction of the hypothalamus and obesity in mice. SCIENCE CHINA-LIFE SCIENCES 2020; 64:419-433. [PMID: 32803714 DOI: 10.1007/s11427-020-1767-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 06/23/2020] [Indexed: 10/23/2022]
Abstract
Cenpj is a centrosomal protein located at the centrosomes and the base of cilia, it plays essential roles in regulating neurogenesis and cerebral cortex development. Although centrosomal and cilium dysfunction are one of the causes of obesity, insulin resistance, and type 2 diabetes, the role that Cenpj plays in the regulation of body weight remains unclear. Here, we deleted Cenpj by crossing Cenpjflox/flox mice with Nkx2.1-Cre mice. Loss of the centrosomal protein Cenpj in Nkx2.1-expressing cells causes morbid obesity in mice at approximately 4 months of age with expended brain ventricles but no change of brain size. We found that hypothalamic cells exhibited reduced proliferation and increased apoptosis upon Cenpj depletion at the embryonic stages, resulting in a dramatic decrease in the number of Proopiomelanocortin (POMC) neurons and electrophysiological dysfunction of NPY neurons in the arcuate nucleus (ARC) in adults. Furthermore, depletion of Cenpj also reduced the neuronal projection from the ARC to the paraventricular nucleus (PVN), with decreased melanocortin-4 receptors (MC4R) expression in PVN neurons. The study defines the roles that Cenpj plays in regulating hypothalamus development and body weight, providing a foundation for further understanding of the pathological mechanisms of related diseases.
Collapse
Affiliation(s)
- Wenyu Ding
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Changjiang Zhang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Brain-Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Baisong Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China
| | - Xin Zhou
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Brain-Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Le Sun
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Brain-Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Suijuan Zhong
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Jing Liu
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Brain-Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Junjing Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China.,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China
| | - Xiaoqun Wang
- State Key Laboratory of Brain and Cognitive Science, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Brain-Intelligence Technology (Shanghai), Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China. .,Advanced Innovation Center for Human Brain Protection, Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| | - Qian Wu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, 100875, China. .,IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, 100875, China.
| |
Collapse
|
14
|
Piórkowska K, Żukowski K, Połtowicz K, Nowak J, Ropka-Molik K, Derebecka N, Wesoły J, Wojtysiak D. Identification of candidate genes and regulatory factors related to growth rate through hypothalamus transcriptome analyses in broiler chickens. BMC Genomics 2020; 21:509. [PMID: 32703165 PMCID: PMC7376931 DOI: 10.1186/s12864-020-06884-5] [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: 12/18/2019] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Background Intensive selection for growth rate (GR) in broiler chickens carries negative after-effects, such as aberrations in skeletal development and the immune system, heart failure, and deterioration of meat quality. In Poland, fast-growing chicken populations are highly non-uniform in term of growth rate, which is highly unprofitable for poultry producers. Therefore, the identification of genetic markers for boiler GR that could support the selection process is needed. The hypothalamus is strongly associated with growth regulation by inducing important pituitary hormones. Therefore, the present study used this tissue to pinpoint genes involved in chicken growth control. Results The experiment included male broilers of Ross 308 strain in two developmental stages, after 3rd and 6th week of age, which were maintained in the same housing and feeding conditions. The obtained results show for the overexpression of genes related to orexigenic molecules, such as neuropeptide Y (NPY), aldehyde dehydrogenase 1 family, member A1 (ALDH1A1), galanin (GAL), and pro-melanin concentrating hormone (PMCH) in low GR cockerels. Conclusion The results reveal strong associations between satiety centre and the growth process. The present study delivers new insights into hypothalamic regulation in broiler chickens and narrows the area for the searching of genetic markers for GR. Graphical abstract ![]()
Collapse
Affiliation(s)
- Katarzyna Piórkowska
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Balice, Poland
| | - Kacper Żukowski
- Department of Cattle Breeding, National Research Institute of Animal Production, Balice, Poland
| | - Katarzyna Połtowicz
- Department of Poultry Breeding, National Research Institute of Animal Production, Balice, Poland.
| | - Joanna Nowak
- Department of Poultry Breeding, National Research Institute of Animal Production, Balice, Poland
| | - Katarzyna Ropka-Molik
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Balice, Poland
| | - Natalia Derebecka
- Adam Mickiewicz University, Faculty of Biology, Laboratory of High Throughput Technologies Institute of Molecular Biology and Biotechnology, Poznań, Poland
| | - Joanna Wesoły
- Adam Mickiewicz University, Faculty of Biology, Laboratory of High Throughput Technologies Institute of Molecular Biology and Biotechnology, Poznań, Poland
| | - Dorota Wojtysiak
- Department of Animal Genetics, Breeding and Ethology, University of Agriculture in Krakow, Cracow, Poland
| |
Collapse
|
15
|
Transcriptional control of lung alveolar type 1 cell development and maintenance by NK homeobox 2-1. Proc Natl Acad Sci U S A 2019; 116:20545-20555. [PMID: 31548395 DOI: 10.1073/pnas.1906663116] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The extraordinarily thin alveolar type 1 (AT1) cell constitutes nearly the entire gas exchange surface and allows passive diffusion of oxygen into the blood stream. Despite such an essential role, the transcriptional network controlling AT1 cells remains unclear. Using cell-specific knockout mouse models, genomic profiling, and 3D imaging, we found that NK homeobox 2-1 (Nkx2-1) is expressed in AT1 cells and is required for the development and maintenance of AT1 cells. Without Nkx2-1, developing AT1 cells lose 3 defining features-molecular markers, expansive morphology, and cellular quiescence-leading to alveolar simplification and lethality. NKX2-1 is also cell-autonomously required for the same 3 defining features in mature AT1 cells. Intriguingly, Nkx2-1 mutant AT1 cells activate gastrointestinal (GI) genes and form dense microvilli-like structures apically. Single-cell RNA-seq supports a linear transformation of Nkx2-1 mutant AT1 cells toward a GI fate. Whole lung ChIP-seq shows NKX2-1 binding to 68% of genes that are down-regulated upon Nkx2-1 deletion, including 93% of known AT1 genes, but near-background binding to up-regulated genes. Our results place NKX2-1 at the top of the AT1 cell transcriptional hierarchy and demonstrate remarkable plasticity of an otherwise terminally differentiated cell type.
Collapse
|
16
|
Kano M, Suga H, Ishihara T, Sakakibara M, Soen M, Yamada T, Ozaki H, Mitsumoto K, Kasai T, Sugiyama M, Onoue T, Tsunekawa T, Takagi H, Hagiwara D, Ito Y, Iwama S, Goto M, Banno R, Arima H. Tanycyte-Like Cells Derived From Mouse Embryonic Stem Culture Show Hypothalamic Neural Stem/Progenitor Cell Functions. Endocrinology 2019; 160:1701-1718. [PMID: 31135891 DOI: 10.1210/en.2019-00105] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/22/2019] [Indexed: 01/01/2023]
Abstract
Tanycytes have recently been accepted as neural stem/progenitor cells in the postnatal hypothalamus. Persistent retina and anterior neural fold homeobox (Rax) expression is characteristic of tanycytes in contrast to its transient expression of whole hypothalamic precursors. In this study, we found that Rax+ residual cells in the maturation phase of hypothalamic differentiation in mouse embryonic stem cell (mESC) cultures had similar characteristics to ventral tanycytes. They expressed typical neural stem/progenitor cell markers, including Sox2, vimentin, and nestin, and differentiated into mature neurons and glial cells. Quantitative RT-PCR analysis showed that Rax+ residual cells expressed Fgf-10, Fgf-18, and Lhx2, which are expressed by ventral tanycytes. They highly expressed tanycyte-specific genes Dio2 and Gpr50 compared with Rax+ early hypothalamic progenitor cells. Therefore, Rax+ residual cells in the maturation phase of hypothalamic differentiation were considered to be more differentiated and similar to late progenitor cells and tanycytes. They self-renewed and formed neurospheres when cultured with exogenous FGF-2. Additionally, these Rax+ neurospheres differentiated into three neuronal lineages (neurons, astrocytes, and oligodendrocytes), including neuropeptide Y+ neuron, that are reported to be differentiated from ventral tanycytes toward the arcuate nuclei. Thus, Rax+ residual cells were multipotent neural stem/progenitor cells. Rax+ neurospheres were stably passaged and retained high Sox2 expression even after multiple passages. These results suggest the successful induction of Rax+ tanycyte-like cells from mESCs [induced tanycyte-like (iTan) cells]. These hypothalamic neural stem/progenitor cells may have potential in regenerative medicine and as a research tool.
Collapse
Affiliation(s)
- Mayuko Kano
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidetaka Suga
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takeshi Ishihara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Drug Discovery Technologies, Drug Discovery and Disease Research Laboratory, Shionogi and Co., Ltd., Osaka, Japan
| | - Mayu Sakakibara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mika Soen
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomiko Yamada
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hajime Ozaki
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuki Mitsumoto
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takatoshi Kasai
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mariko Sugiyama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takeshi Onoue
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Taku Tsunekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Takagi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Daisuke Hagiwara
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshihiro Ito
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Motomitsu Goto
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryoichi Banno
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
17
|
Bouyakdan K, Martin H, Liénard F, Budry L, Taib B, Rodaros D, Chrétien C, Biron É, Husson Z, Cota D, Pénicaud L, Fulton S, Fioramonti X, Alquier T. The gliotransmitter ACBP controls feeding and energy homeostasis via the melanocortin system. J Clin Invest 2019; 129:2417-2430. [PMID: 30938715 PMCID: PMC6546475 DOI: 10.1172/jci123454] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Glial cells have emerged as key players in the central control of energy balance and etiology of obesity. Astrocytes play a central role in neural communication via the release of gliotransmitters. Acyl-CoA binding protein (ACBP)-derived endozepines are secreted peptides that modulate the GABAA receptor. In the hypothalamus, ACBP is enriched in arcuate nucleus (ARC) astrocytes, ependymocytes and tanycytes. Central administration of the endozepine octadecaneuropeptide (ODN) reduces feeding and improves glucose tolerance, yet the contribution of endogenous ACBP in energy homeostasis is unknown. We demonstrated that ACBP deletion in GFAP+ astrocytes, but not in Nkx2.1-lineage neural cells, promoted diet-induced hyperphagia and obesity in both male and female mice, an effect prevented by viral rescue of ACBP in ARC astrocytes. ACBP-astrocytes were observed in apposition with proopiomelanocortin (POMC) neurons and ODN selectively activated POMC neurons through the ODN-GPCR but not GABAA, and supressed feeding while increasing carbohydrate utilization via the melanocortin system. Similarly, ACBP overexpression in ARC astrocytes reduced feeding and weight gain. Finally, the ODN-GPCR agonist decreased feeding and promoted weight loss in ob/ob mice. These findings uncover ACBP as an ARC gliopeptide playing a key role in energy balance control and exerting strong anorectic effects via the central melanocortin system.
Collapse
Affiliation(s)
- Khalil Bouyakdan
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal Diabetes Research Center, and Departments of Medicine, Pathology and Cell Biology, Biochemistry, Neurosciences, and Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Hugo Martin
- Université de Bordeaux, INRA, NutriNeuro, Bordeaux, France
- Bordeaux INP, NutriNeuro, Talence, France
| | - Fabienne Liénard
- Centre des Sciences du Goût et de l’Alimentation, UMR 6265 CNRS, 1324 INRA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Lionel Budry
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal Diabetes Research Center, and Departments of Medicine, Pathology and Cell Biology, Biochemistry, Neurosciences, and Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Bouchra Taib
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal Diabetes Research Center, and Departments of Medicine, Pathology and Cell Biology, Biochemistry, Neurosciences, and Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Demetra Rodaros
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal Diabetes Research Center, and Departments of Medicine, Pathology and Cell Biology, Biochemistry, Neurosciences, and Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Chloé Chrétien
- Centre des Sciences du Goût et de l’Alimentation, UMR 6265 CNRS, 1324 INRA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Éric Biron
- Faculty of Pharmacy, Université Laval and Laboratory of Medicinal Chemistry, Centre de Recherche du Centre Hospitalier Universitaire de Québec (CRCHUQ), Quebec, Quebec, Canada
| | - Zoé Husson
- Université de Bordeaux, INRA, NutriNeuro, Bordeaux, France
- Bordeaux INP, NutriNeuro, Talence, France
- INSERM, Université de Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
| | - Daniela Cota
- INSERM, Université de Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
| | - Luc Pénicaud
- Centre des Sciences du Goût et de l’Alimentation, UMR 6265 CNRS, 1324 INRA, Université de Bourgogne Franche-Comté, Dijon, France
- Stromalab, CNRS ERL 5311, Université de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Stephanie Fulton
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal Diabetes Research Center, and Departments of Medicine, Pathology and Cell Biology, Biochemistry, Neurosciences, and Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Xavier Fioramonti
- Université de Bordeaux, INRA, NutriNeuro, Bordeaux, France
- Bordeaux INP, NutriNeuro, Talence, France
- Centre des Sciences du Goût et de l’Alimentation, UMR 6265 CNRS, 1324 INRA, Université de Bourgogne Franche-Comté, Dijon, France
| | - Thierry Alquier
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montreal Diabetes Research Center, and Departments of Medicine, Pathology and Cell Biology, Biochemistry, Neurosciences, and Nutrition, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
18
|
The Homeodomain Transcription Factor NKX2.1 Is Essential for the Early Specification of Melanocortin Neuron Identity and Activates Pomc Expression in the Developing Hypothalamus. J Neurosci 2019; 39:4023-4035. [PMID: 30886014 DOI: 10.1523/jneurosci.2924-18.2019] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 03/03/2019] [Accepted: 03/08/2019] [Indexed: 12/14/2022] Open
Abstract
Food intake is tightly regulated by a group of neurons present in the arcuate nucleus of the hypothalamus, which release Pomc-encoded melanocortins, the absence of which induces marked hyperphagia and early-onset obesity. Although the relevance of hypothalamic POMC neurons in the regulation of body weight and energy balance is well appreciated, little is known about the transcription factors that establish the melanocortin neuron identity during brain development and its phenotypic maintenance in postnatal life. Here, we report that the transcription factor NKX2.1 is present in mouse hypothalamic POMC neurons from early development to adulthood. Electromobility shift assays showed that NKX2.1 binds in vitro to NKX binding motifs present in the neuronal Pomc enhancers nPE1 and nPE2 and chromatin immunoprecipitation assays detected in vivo binding of NKX2.1 to nPE1 and nPE2 in mouse hypothalamic extracts. Transgenic and mutant studies performed in mouse embryos of either sex and adult males showed that the NKX motifs present in nPE1 and nPE2 are essential for their transcriptional enhancer activity. The conditional early inactivation of Nkx2.1 in the ventral hypothalamus prevented the onset of Pomc expression. Selective Nkx2.1 ablation from POMC neurons decreased Pomc expression in adult males and mildly increased their body weight and adiposity. Our results demonstrate that NKX2.1 is necessary to activate Pomc expression by binding to conserved canonical NKX motifs present in nPE1 and nPE2. Therefore, NKX2.1 plays a critical role in the early establishment of hypothalamic melanocortin neuron identity and participates in the maintenance of Pomc expression levels during adulthood.SIGNIFICANCE STATEMENT Food intake and body weight regulation depend on hypothalamic neurons that release satiety-inducing neuropeptides, known as melanocortins. Central melanocortins are encoded byPomc, and Pomc mutations may lead to hyperphagia and severe obesity. Although the importance of central melanocortins is well appreciated, the genetic program that establishes and maintains fully functional POMC neurons remains to be explored. Here, we combined molecular, genetic, developmental, and functional studies that led to the discovery of NKX2.1, a transcription factor that participates in the early morphogenesis of the developing hypothalamus, as a key player in establishing the early identity of melanocortin neurons by activating Pomc expression. Thus, Nkx2.1 adds to the growing list of genes that participate in body weight regulation and adiposity.
Collapse
|
19
|
Wang L, De Solis AJ, Goffer Y, Birkenbach KE, Engle SE, Tanis R, Levenson JM, Li X, Rausch R, Purohit M, Lee JY, Tan J, De Rosa MC, Doege CA, Aaron HL, Martins GJ, Brüning JC, Egli D, Costa R, Berbari N, Leibel RL, Stratigopoulos G. Ciliary gene RPGRIP1L is required for hypothalamic arcuate neuron development. JCI Insight 2019; 4:e123337. [PMID: 30728336 PMCID: PMC6413800 DOI: 10.1172/jci.insight.123337] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 01/03/2019] [Indexed: 12/16/2022] Open
Abstract
Intronic polymorphisms in the α-ketoglutarate-dependent dioxygenase gene (FTO) that are highly associated with increased body weight have been implicated in the transcriptional control of a nearby ciliary gene, retinitis pigmentosa GTPase regulator-interacting protein-1 like (RPGRIP1L). Previous studies have shown that congenital Rpgrip1l hypomorphism in murine proopiomelanocortin (Pomc) neurons causes obesity by increasing food intake. Here, we show by congenital and adult-onset Rpgrip1l deletion in Pomc-expressing neurons that the hyperphagia and obesity are likely due to neurodevelopmental effects that are characterized by a reduction in the Pomc/Neuropeptide Y (Npy) neuronal number ratio and marked increases in arcuate hypothalamic-paraventricular hypothalamic (ARH-PVH) axonal projections. Biallelic RPGRIP1L mutations result in fewer cilia-positive human induced pluripotent stem cell-derived (iPSC-derived) neurons and blunted responses to Sonic Hedgehog (SHH). Isogenic human ARH-like embryonic stem cell-derived (ESc-derived) neurons homozygous for the obesity-risk alleles at rs8050136 or rs1421085 have decreased RPGRIP1L expression and have lower numbers of POMC neurons. RPGRIP1L overexpression increases POMC cell number. These findings suggest that apparently functional intronic polymorphisms affect hypothalamic RPGRIP1L expression and impact development of POMC neurons and their derivatives, leading to hyperphagia and increased adiposity.
Collapse
Affiliation(s)
- Liheng Wang
- Naomi Berrie Diabetes Center and Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Alain J. De Solis
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Yossef Goffer
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Kathryn E. Birkenbach
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Staci E. Engle
- Department of Biology, Indiana University-Purdue University, Indianapolis, Indiana, USA
| | - Ross Tanis
- University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Jacob M. Levenson
- University of Central Florida College of Medicine, Orlando, Florida, USA
| | - Xueting Li
- Institute of Human Nutrition graduate program, Columbia University, New York, New York, USA
| | - Richard Rausch
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Manika Purohit
- Zuckerman Institute, Columbia University, New York, New York, USA
| | - Jen-Yi Lee
- Cancer Research Laboratory Molecular Imaging Center, University of California, Berkeley, 94720, USA
| | - Jerica Tan
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Maria Caterina De Rosa
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Claudia A. Doege
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Holly L. Aaron
- Cancer Research Laboratory Molecular Imaging Center, University of California, Berkeley, 94720, USA
| | | | - Jens C. Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
- National Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Dieter Egli
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - Rui Costa
- Zuckerman Institute, Columbia University, New York, New York, USA
| | - Nicolas Berbari
- Department of Biology, Indiana University-Purdue University, Indianapolis, Indiana, USA
| | - Rudolph L. Leibel
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| | - George Stratigopoulos
- Naomi Berrie Diabetes Center & Division of Molecular Genetics, Department of Pediatrics, College of Physicians and Surgeons of Columbia University, New York, New York, USA
| |
Collapse
|
20
|
Corman TS, Bergendahl SE, Epstein DJ. Distinct temporal requirements for Sonic hedgehog signaling in development of the tuberal hypothalamus. Development 2018; 145:dev167379. [PMID: 30291164 PMCID: PMC6240319 DOI: 10.1242/dev.167379] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 09/20/2018] [Indexed: 12/14/2022]
Abstract
Sonic hedgehog (Shh) plays well characterized roles in brain and spinal cord development, but its functions in the hypothalamus have been more difficult to elucidate owing to the complex neuroanatomy of this brain area. Here, we use fate mapping and conditional deletion models in mice to define requirements for dynamic Shh activity at distinct developmental stages in the tuberal hypothalamus, a brain region with important homeostatic functions. At early time points, Shh signaling regulates dorsoventral patterning, neurogenesis and the size of the ventral midline. Fate-mapping experiments demonstrate that Shh-expressing and -responsive progenitors contribute to distinct neuronal subtypes, accounting for some of the cellular heterogeneity in tuberal hypothalamic nuclei. Conditional deletion of the hedgehog transducer smoothened (Smo), after dorsoventral patterning has been established, reveals that Shh signaling is necessary to maintain proliferation and progenitor identity during peak periods of hypothalamic neurogenesis. We also find that mosaic disruption of Smo causes a non-cell autonomous gain in Shh signaling activity in neighboring wild-type cells, suggesting a mechanism for the pathogenesis of hypothalamic hamartomas, benign tumors that form during hypothalamic development.
Collapse
Affiliation(s)
- Tanya S Corman
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6145, USA
| | - Solsire E Bergendahl
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6145, USA
| | - Douglas J Epstein
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6145, USA
| |
Collapse
|
21
|
Cellular fate decisions in the developing female anteroventral periventricular nucleus are regulated by canonical Notch signaling. Dev Biol 2018; 442:87-100. [PMID: 29885287 DOI: 10.1016/j.ydbio.2018.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/05/2018] [Indexed: 01/20/2023]
Abstract
The hypothalamic anteroventral periventricular nucleus (AVPV) is the major regulator of reproductive function within the hypothalamic-pituitary-gonadal (HPG) axis. Despite an understanding of the function of neuronal subtypes within the AVPV, little is known about the molecular mechanisms regulating their development. Previous work from our laboratory has demonstrated that Notch signaling is required in progenitor cell maintenance and formation of kisspeptin neurons of the arcuate nucleus (ARC) while simultaneously restraining POMC neuron number. Based on these findings, we hypothesized that the Notch signaling pathway may act similarly in the AVPV by promoting development of kisspeptin neurons at the expense of other neuronal subtypes. To address this hypothesis, we utilized a genetic mouse model with a conditional loss of Rbpj in Nkx2.1 expressing cells (Rbpj cKO). We noted an increase in cellular proliferation, as marked by Ki-67, in the hypothalamic ventricular zone (HVZ) in Rbpj cKO mice at E13.5. This corresponded to an increase in general neurogenesis and more TH-positive neurons. Additionally, an increase in OLIG2-positive early oligodendrocytic precursor cells was observed at postnatal day 0 in Rbpj cKO mice. By 5 weeks of age in Rbpj cKO mice, TH-positive cells were readily detected in the AVPV but few kisspeptin neurons were present. To elucidate the direct effects of Notch signaling on neuron and glia differentiation, an in vitro primary hypothalamic neurosphere assay was employed. We demonstrated that treatment with the chemical Notch inhibitor DAPT increased mKi67 and Olig2 mRNA expression while decreasing astroglial Gfap expression, suggesting Notch signaling regulates both proliferation and early glial fate decisions. A modest increase in expression of TH in both the cell soma and neurite extensions was observed after extended culture, suggesting that inhibition of Notch signaling alone is enough to bias progenitors towards a dopaminergic fate. Together, these data suggest that Notch signaling restricts early cellular proliferation and differentiation of neurons and oligodendrocytes both in vivo and in vitro and acts as a fate selector of kisspeptin neurons.
Collapse
|
22
|
Development of neuroendocrine neurons in the mammalian hypothalamus. Cell Tissue Res 2018; 375:23-39. [PMID: 29869716 DOI: 10.1007/s00441-018-2859-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 05/11/2018] [Indexed: 12/21/2022]
Abstract
The neuroendocrine system consists of a heterogeneous collection of (mostly) neuropeptidergic neurons found in four hypothalamic nuclei and sharing the ability to secrete neurohormones (all of them neuropeptides except dopamine) into the bloodstream. There are, however, abundant hypothalamic non-neuroendocrine neuropeptidergic neurons developing in parallel with the neuroendocrine system, so that both cannot be entirely disentangled. This heterogeneity results from the workings of a network of transcription factors many of which are already known. Olig2 and Fezf2 expressed in the progenitors, acting through mantle-expressed Otp and Sim1, Sim2 and Pou3f2 (Brn2), regulate production of magnocellular and anterior parvocellular neurons. Nkx2-1, Rax, Ascl1, Neurog3 and Dbx1 expressed in the progenitors, acting through mantle-expressed Isl1, Dlx1, Gsx1, Bsx, Hmx2/3, Ikzf1, Nr5a2 (LH-1) and Nr5a1 (SF-1) are responsible for tuberal parvocellular (arcuate nucleus) and other neuropeptidergic neurons. The existence of multiple progenitor domains whose progeny undergoes intricate tangential migrations as one source of complexity in the neuropeptidergic hypothalamus is the focus of much attention. How neurosecretory cells target axons to the medial eminence and posterior hypophysis is gradually becoming clear and exciting progress has been made on the mechanisms underlying neurovascular interface formation. While rat neuroanatomy and targeted mutations in mice have yielded fundamental knowledge about the neuroendocrine system in mammals, experiments on chick and zebrafish are providing key information about cellular and molecular mechanisms. Looking forward, data from every source will be necessary to unravel the ways in which the environment affects neuroendocrine development with consequences for adult health and disease.
Collapse
|
23
|
Lee B, Kim J, An T, Kim S, Patel EM, Raber J, Lee SK, Lee S, Lee JW. Dlx1/2 and Otp coordinate the production of hypothalamic GHRH- and AgRP-neurons. Nat Commun 2018; 9:2026. [PMID: 29795232 PMCID: PMC5966420 DOI: 10.1038/s41467-018-04377-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 04/23/2018] [Indexed: 01/07/2023] Open
Abstract
Despite critical roles of the hypothalamic arcuate neurons in controlling the growth and energy homeostasis, the gene regulatory network directing their development remains unclear. Here we report that the transcription factors Dlx1/2 and Otp coordinate the balanced generation of the two functionally related neurons in the hypothalamic arcuate nucleus, GHRH-neurons promoting the growth and AgRP-neurons controlling the feeding and energy expenditure. Dlx1/2-deficient mice show a loss-of-GHRH-neurons and an increase of AgRP-neurons, and consistently develop dwarfism and consume less energy. These results indicate that Dlx1/2 are crucial for specifying the GHRH-neuronal identity and, simultaneously, for suppressing AgRP-neuronal fate. We further show that Otp is required for the generation of AgRP-neurons and that Dlx1/2 repress the expression of Otp by directly binding the Otp gene. Together, our study demonstrates that the identity of GHRH- and AgRP-neurons is synchronously specified and segregated by the Dlx1/2-Otp gene regulatory axis.
Collapse
Affiliation(s)
- Bora Lee
- Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul, 02792, Korea
| | - Janghyun Kim
- Neuroscience Section, Papé Family Pediatrics Research Center, Department of Pediatrics, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Taekyeong An
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, 06978, Korea
| | - Sangsoo Kim
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, 06978, Korea
| | - Esha M Patel
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, OR, 97239, USA
- Departments of Neurology and Radiation Medicine, and Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Soo-Kyung Lee
- Neuroscience Section, Papé Family Pediatrics Research Center, Department of Pediatrics, Oregon Health and Science University, Portland, OR, 97239, USA
- Vollum Institute, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Seunghee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, 08826, Korea.
| | - Jae W Lee
- Neuroscience Section, Papé Family Pediatrics Research Center, Department of Pediatrics, Oregon Health and Science University, Portland, OR, 97239, USA.
| |
Collapse
|
24
|
Gao Y, Yao T, Deng Z, Sohn JW, Sun J, Huang Y, Kong X, Yu KJ, Wang RT, Chen H, Guo H, Yan J, Cunningham KA, Chang Y, Liu T, Williams KW. TrpC5 Mediates Acute Leptin and Serotonin Effects via Pomc Neurons. Cell Rep 2017; 18:583-592. [PMID: 28099839 DOI: 10.1016/j.celrep.2016.12.072] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/12/2016] [Accepted: 12/21/2016] [Indexed: 01/28/2023] Open
Abstract
The molecular mechanisms underlying acute leptin and serotonin 2C receptor-induced hypophagia remain unclear. Here, we show that neuronal and pro-opiomelanocortin (Pomc)-specific loss of transient receptor potential cation 5 (TrpC5) subunits is sufficient to decrease energy expenditure and increase food intake resulting in elevated body weight. Deficiency of Trpc5 subunits in Pomc neurons is also sufficient to block the anorexigenic effects of leptin and serotonin 2C receptor (Ht2Cr) agonists. The loss of acute anorexigenic effects of these receptors is concomitant with a blunted electrophysiological response to both leptin and Ht2Cr agonists in arcuate Pomc neurons. We also demonstrate that the Ht2Cr agonist lorcaserin-induced improvements in glucose and insulin tolerance are blocked by TrpC5 deficiency in Pomc neurons. Together, our results link TrpC5 subunits in the brain with leptin- and serotonin 2C receptor-dependent changes in neuronal activity, as well as energy balance, feeding behavior, and glucose metabolism.
Collapse
Affiliation(s)
- Yong Gao
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China; Division of Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9077, USA
| | - Ting Yao
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, China; Division of Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9077, USA
| | - Zhuo Deng
- Department of Gynecology, Shaanxi Provincial People's Hospital, Shaanxi 710000, China; Division of Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9077, USA
| | - Jong-Woo Sohn
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea; Division of Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9077, USA
| | - Jia Sun
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China; Division of Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9077, USA
| | - Yiru Huang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China; Division of Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9077, USA
| | - Xingxing Kong
- Division of Endocrinology, Beth Israel Deaconess Medical Center and Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Kai-Jiang Yu
- Department of Intensive Care Unit, Third Affiliated Hospital, Harbin Medical University, Nangang District, Harbin, 150081, China
| | - Rui-Tao Wang
- Department of Intensive Care Unit, Third Affiliated Hospital, Harbin Medical University, Nangang District, Harbin, 150081, China
| | - Hong Chen
- Department of Endocrinology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Hongbo Guo
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jianqun Yan
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, China
| | - Kathryn A Cunningham
- Center for Addiction Research and the Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Yongsheng Chang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China.
| | - Tiemin Liu
- Department of Intensive Care Unit, Third Affiliated Hospital, Harbin Medical University, Nangang District, Harbin, 150081, China; Division of Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9077, USA.
| | - Kevin W Williams
- Division of Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390-9077, USA.
| |
Collapse
|
25
|
Abstract
The hypothalamus is an evolutionarily conserved brain structure that regulates an organism's basic functions, such as homeostasis and reproduction. Several hypothalamic nuclei and neuronal circuits have been the focus of many studies seeking to understand their role in regulating these basic functions. Within the hypothalamic neuronal populations, the arcuate melanocortin system plays a major role in controlling homeostatic functions. The arcuate pro-opiomelanocortin (POMC) neurons in particular have been shown to be critical regulators of metabolism and reproduction because of their projections to several brain areas both in and outside of the hypothalamus, such as autonomic regions of the brain stem and spinal cord. Here, we review and discuss the current understanding of POMC neurons from their development and intracellular regulators to their physiological functions and pathological dysregulation.
Collapse
Affiliation(s)
- Chitoku Toda
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Anna Santoro
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Jung Dae Kim
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Sabrina Diano
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520.,Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520.,Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
26
|
Xie Y, Dorsky RI. Development of the hypothalamus: conservation, modification and innovation. Development 2017; 144:1588-1599. [PMID: 28465334 DOI: 10.1242/dev.139055] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The hypothalamus, which regulates fundamental aspects of physiological homeostasis and behavior, is a brain region that exhibits highly conserved anatomy across vertebrate species. Its development involves conserved basic mechanisms of induction and patterning, combined with a more plastic process of neuronal fate specification, to produce brain circuits that mediate physiology and behavior according to the needs of each species. Here, we review the factors involved in the induction, patterning and neuronal differentiation of the hypothalamus, highlighting recent evidence that illustrates how changes in Wnt/β-catenin signaling during development may lead to species-specific form and function of this important brain structure.
Collapse
Affiliation(s)
- Yuanyuan Xie
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Richard I Dorsky
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
27
|
Solek CM, Feng S, Perin S, Weinschutz Mendes H, Ekker M. Lineage tracing of dlx1a/2a and dlx5a/6a expressing cells in the developing zebrafish brain. Dev Biol 2017; 427:131-147. [PMID: 28479339 DOI: 10.1016/j.ydbio.2017.04.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 04/28/2017] [Accepted: 04/29/2017] [Indexed: 02/06/2023]
Abstract
Lineage tracing of specific populations of progenitor cells provides crucial information about developmental programs. Four members of the Dlx homeobox gene family, Dlx1,2, 5 and 6, are involved in the specification of γ-aminobutyric acid (GABA)ergic neurons in the vertebrate forebrain. Orthologous genes in mammals and teleost show similarities in expression patterns and transcriptional regulation mechanisms. We have used lineage tracing to permanently label dlx-expressing cells in the zebrafish and have characterized the progeny of these cells in the larva and in the juvenile and adult brain. We have found that dlx1a/2a and dlx5a/6a expressing progenitors give rise, for the most part, to small populations of cells which constitute only a small proportion of GABAergic cells in the adult brain tissue. Moreover, some of the cells do not acquire a neuronal phenotype suggesting that, regardless of the time a cell expresses dlx genes in the brain, it can potentially give rise to cells other than neurons. In some instances, labeling larval dlx5a/6a-expressing cells, but not dlx1a/2a-expressing cells, results in massively expanding, widespread clonal expansion throughout the adult brain. Our data provide a detailed lineage analysis of the dlx1a/2a and dlx5a/6a expressing progenitors in the zebrafish brain and lays the foundation for further characterization of the role of these transcription factors beyond the specification of GABAergic neurons.
Collapse
Affiliation(s)
- Cynthia M Solek
- CAREG, Department of Biology, University of Ottawa, 20 Marie-Curie Private, Ottawa, ON, Canada K1N 6N5; Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC, Canada H3A 2B4
| | - Shengrui Feng
- CAREG, Department of Biology, University of Ottawa, 20 Marie-Curie Private, Ottawa, ON, Canada K1N 6N5; Department of Medical Biophysics, University of Toronto, 610 University Avenue, Toronto, ON, Canada M5G 2M9
| | - Sofia Perin
- CAREG, Department of Biology, University of Ottawa, 20 Marie-Curie Private, Ottawa, ON, Canada K1N 6N5
| | - Hellen Weinschutz Mendes
- CAREG, Department of Biology, University of Ottawa, 20 Marie-Curie Private, Ottawa, ON, Canada K1N 6N5
| | - Marc Ekker
- CAREG, Department of Biology, University of Ottawa, 20 Marie-Curie Private, Ottawa, ON, Canada K1N 6N5.
| |
Collapse
|
28
|
Melanocortin-3 receptors expressed in Nkx2.1(+ve) neurons are sufficient for controlling appetitive responses to hypocaloric conditioning. Sci Rep 2017; 7:44444. [PMID: 28294152 PMCID: PMC5353610 DOI: 10.1038/srep44444] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 02/08/2017] [Indexed: 01/12/2023] Open
Abstract
Melanocortin-3 receptors (MC3R) have a contextual role in appetite control that is amplified with hypocaloric conditioning. C57BL/6J (B6) mice subjected to hypocaloric feeding schedules (HFS) exhibit compulsive behavioral responses involving food anticipatory activity (FAA) and caloric loading following food access. These homeostatic responses to calorie-poor environs are attenuated in B6 mice in which Mc3r transcription is suppressed by a lox-stop-lox sequence in the 5'UTR (Mc3rTB/TB). Here, we report that optimization of caloric loading in B6 mice subject to HFS, characterized by increased meal size and duration, is not observed in Mc3rTB/TB mice. Analysis of hypothalamic and neuroendocrine responses to HFS throughout the light-dark cycle suggests uncoupling of hypothalamic responses involving appetite-stimulating fasting-responsive hypothalamic neurons expressing agouti-related peptide (AgRP) and neuropeptide Y (Npy). Rescuing Mc3rs expression in Nkx2.1(+ve) neurons is sufficient to restore normal hypothalamic responses to negative energy balance. In addition, Mc3rs expressed in Nkx2.1(+ve) neurons are also sufficient to restore FAA and caloric loading of B6 mice subjected to HFS. In summary, MC3Rs expressed in Nkx2.1(+ve) neurons are sufficient to coordinate hypothalamic response and expression of compulsive behavioral responses involving meal anticipation and consumption of large meals during situations of prolonged negative energy balance.
Collapse
|
29
|
Origins and Functions of the Ventrolateral VMH: A Complex Neuronal Cluster Orchestrating Sex Differences in Metabolism and Behavior. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1043:199-213. [PMID: 29224096 DOI: 10.1007/978-3-319-70178-3_10] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The neuroendocrine brain or hypothalamus has emerged as one of the most highly sexually dimorphic brain regions in mammals, and specifically in rodents. It is not surprising that hypothalamic nuclei play a pivotal role in controlling sex-dependent physiology. This brain region functions as a chief executive officer or master regulator of homeostatic physiological systems to integrate both external and internal signals. In this review, we describe sex differences in energy homeostasis that arise in one area of the hypothalamus, the ventrolateral subregion of the ventromedial hypothalamus (VMHvl) with a focus on how male and female neurons function in metabolic and behavioral aspects. Because other chapters within this book provide details on signaling pathways in the VMH that contribute to sex differences in metabolism, our discussion will be limited to how the sexually dimorphic VMHvl develops and what key regulators are thought to control the many functional and physiological endpoints attributed to this region. In the last decade, several exciting new studies using state-of-the-art genetic and molecular tools are beginning to provide some understanding as to how specific neurons contribute to the coordinated physiological responses needed by male and females. New technology that combines intersectional spatial and genetic approaches is now allowing further refinement in how we describe, probe, and manipulate critical male and female neurocircuits involved in metabolism.
Collapse
|
30
|
Nesan D, Kurrasch DM. Genetic programs of the developing tuberal hypothalamus and potential mechanisms of their disruption by environmental factors. Mol Cell Endocrinol 2016; 438:3-17. [PMID: 27720896 DOI: 10.1016/j.mce.2016.09.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/22/2016] [Accepted: 09/29/2016] [Indexed: 12/15/2022]
Abstract
The hypothalamus is a critical regulator of body homeostasis, influencing the autonomic nervous system and releasing trophic hormones to modulate the endocrine system. The developmental mechanisms that govern formation of the mature hypothalamus are becoming increasingly understood as research in this area grows, leading us to gain appreciation for how these developmental programs are susceptible to disruption by maternal exposure to endocrine disrupting chemicals or other environmental factors in utero. These vulnerabilities, combined with the prominent roles of the various hypothalamic nuclei in regulating appetite, reproductive behaviour, mood, and other physiologies, create a window whereby early developmental disruption can have potent long-term effects. Here we broadly outline our current understanding of hypothalamic development, with a particular focus on the tuberal hypothalamus, including what is know about nuclear coalescing and maturation. We finish by discussing how exposure to environmental or maternally-derived factors can perhaps disrupt these hypothalamic developmental programs, and potentially lead to neuroendocrine disease states.
Collapse
Affiliation(s)
- Dinushan Nesan
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, University of Calgary, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
31
|
Lee B, Lee S, Lee SK, Lee JW. The LIM-homeobox transcription factor Isl1 plays crucial roles in the development of multiple arcuate nucleus neurons. Development 2016; 143:3763-3773. [PMID: 27578785 DOI: 10.1242/dev.133967] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 08/22/2016] [Indexed: 12/24/2022]
Abstract
Neurons in the hypothalamic arcuate nucleus relay and translate important cues from the periphery into the central nervous system. However, the gene regulatory program directing their development remains poorly understood. Here, we report that the LIM-homeodomain transcription factor Isl1 is expressed in several subpopulations of developing arcuate neurons and plays crucial roles in their fate specification. Mice with conditional deletion of the Isl1 gene in developing hypothalamus display severe deficits in both feeding and linear growth. Consistent with these results, their arcuate nucleus fails to express key fate markers of Isl1-expressing neurons that regulate feeding and growth. These include the orexigenic neuropeptides AgRP and NPY for specifying AgRP-neurons, the anorexigenic neuropeptide αMSH for POMC-neurons, and two growth-stimulatory peptides, growth hormone-releasing hormone (GHRH) for GHRH-neurons and somatostatin (Sst) for Sst-neurons. Finally, we show that Isl1 directly enhances the expression of AgRP by cooperating with the key orexigenic transcription factors glucocorticoid receptor and brain-specific homeobox factor. Our results identify Isl1 as a crucial transcription factor that plays essential roles in the gene regulatory program directing development of multiple arcuate neuronal subpopulations.
Collapse
Affiliation(s)
- Bora Lee
- Neuroscience Section, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA.,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| | - Seunghee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Soo-Kyung Lee
- Neuroscience Section, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA.,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Jae W Lee
- Neuroscience Section, Papé Family Pediatric Research Institute, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA .,Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
32
|
Muthu V, Eachus H, Ellis P, Brown S, Placzek M. Rx3 and Shh direct anisotropic growth and specification in the zebrafish tuberal/anterior hypothalamus. Development 2016; 143:2651-63. [PMID: 27317806 PMCID: PMC4958342 DOI: 10.1242/dev.138305] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/03/2016] [Indexed: 12/11/2022]
Abstract
In the developing brain, growth and differentiation are intimately linked. Here, we show that in the zebrafish embryo, the homeodomain transcription factor Rx3 coordinates these processes to build the tuberal/anterior hypothalamus. Analysis of rx3 chk mutant/rx3 morphant fish and EdU pulse-chase studies reveal that rx3 is required to select tuberal/anterior hypothalamic progenitors and to orchestrate their anisotropic growth. In the absence of Rx3 function, progenitors accumulate in the third ventricular wall, die or are inappropriately specified, the shh(+) anterior recess does not form, and its resident pomc(+), ff1b(+) and otpb(+) Th1(+) cells fail to differentiate. Manipulation of Shh signalling shows that Shh coordinates progenitor cell selection and behaviour by acting as an on-off switch for rx3 Together, our studies show that Shh and Rx3 govern formation of a distinct progenitor domain that elaborates patterning through its anisotropic growth and differentiation.
Collapse
Affiliation(s)
- Victor Muthu
- The Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Helen Eachus
- The Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Pam Ellis
- The Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Sarah Brown
- The Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| | - Marysia Placzek
- The Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield S10 2TN, UK
| |
Collapse
|
33
|
Sokolowski K, Tran T, Esumi S, Kamal Y, Oboti L, Lischinsky J, Goodrich M, Lam A, Carter M, Nakagawa Y, Corbin JG. Molecular and behavioral profiling of Dbx1-derived neurons in the arcuate, lateral and ventromedial hypothalamic nuclei. Neural Dev 2016; 11:12. [PMID: 27209204 PMCID: PMC4875659 DOI: 10.1186/s13064-016-0067-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 05/04/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Neurons in the hypothalamus function to regulate the state of the animal during both learned and innate behaviors, and alterations in hypothalamic development may contribute to pathological conditions such as anxiety, depression or obesity. Despite many studies of hypothalamic development and function, the link between embryonic development and innate behaviors remains unexplored. Here, focusing on the embryonically expressed homeodomain-containing gene Developing Brain Homeobox 1 (Dbx1), we explored the relationship between embryonic lineage, post-natal neuronal identity and lineage-specific responses to innate cues. We found that Dbx1 is widely expressed across multiple developing hypothalamic subdomains. Using standard and inducible fate-mapping to trace the Dbx1-derived neurons, we identified their contribution to specific neuronal subtypes across hypothalamic nuclei and further mapped their activation patterns in response to a series of well-defined innate behaviors. RESULTS Dbx1-derived neurons occupy multiple postnatal hypothalamic nuclei including the lateral hypothalamus (LH), arcuate nucleus (Arc) and the ventral medial hypothalamus (VMH). Within these nuclei, Dbx1 (+) progenitors generate a large proportion of the Pmch-, Nesfatin-, Cart-, Hcrt-, Agrp- and ERα-expressing neuronal populations, and to a lesser extent the Pomc-, TH- and Aromatase-expressing populations. Inducible fate-mapping reveals distinct temporal windows for development of the Dbx1-derived LH and Arc populations, with Agrp(+) and Cart(+) populations in the Arc arising early (E7.5-E9.5), while Pmch(+) and Hcrt(+) populations in the LH derived from progenitors expressing Dbx1 later (E9.5-E11.5). Moreover, as revealed by c-Fos labeling, Dbx1-derived cells in male and female LH, Arc and VMH are responsive during mating and aggression. In contrast, Dbx1-lineage cells in the Arc and LH have a broader behavioral tuning, which includes responding to fasting and predator odor cues. CONCLUSION We define a novel fate map of the hypothalamus with respect to Dbx1 expression in hypothalamic progenitor zones. We demonstrate that in a temporally regulated manner, Dbx1-derived neurons contribute to molecularly distinct neuronal populations in the LH, Arc and VMH that have been implicated in a variety of hypothalamic-driven behaviors. Consistent with this, Dbx1-derived neurons in the LH, Arc and VMH are activated during stress and other innate behavioral responses, implicating their involvement in these diverse behaviors.
Collapse
Affiliation(s)
- Katie Sokolowski
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue, NW, Washington, 20010, DC, USA
| | - Tuyen Tran
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue, NW, Washington, 20010, DC, USA
| | - Shigeyuki Esumi
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue, NW, Washington, 20010, DC, USA
- Department of Morphological Neural Science, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Yasmin Kamal
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue, NW, Washington, 20010, DC, USA
| | - Livio Oboti
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue, NW, Washington, 20010, DC, USA
| | - Julieta Lischinsky
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue, NW, Washington, 20010, DC, USA
- Institute for Biomedical Sciences, The George Washington University, Washington, 20037, DC, USA
| | - Meredith Goodrich
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue, NW, Washington, 20010, DC, USA
| | - Andrew Lam
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue, NW, Washington, 20010, DC, USA
| | - Margaret Carter
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue, NW, Washington, 20010, DC, USA
| | - Yasushi Nakagawa
- Department of Neuroscience, University of Minnesota Medical School, Minneapolis, 55455, MN, USA
| | - Joshua G Corbin
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue, NW, Washington, 20010, DC, USA.
| |
Collapse
|
34
|
Malt EA, Juhasz K, Malt UF, Naumann T. A Role for the Transcription Factor Nk2 Homeobox 1 in Schizophrenia: Convergent Evidence from Animal and Human Studies. Front Behav Neurosci 2016; 10:59. [PMID: 27064909 PMCID: PMC4811959 DOI: 10.3389/fnbeh.2016.00059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/11/2016] [Indexed: 12/22/2022] Open
Abstract
Schizophrenia is a highly heritable disorder with diverse mental and somatic symptoms. The molecular mechanisms leading from genes to disease pathology in schizophrenia remain largely unknown. Genome-wide association studies (GWASs) have shown that common single-nucleotide polymorphisms associated with specific diseases are enriched in the recognition sequences of transcription factors that regulate physiological processes relevant to the disease. We have used a “bottom-up” approach and tracked a developmental trajectory from embryology to physiological processes and behavior and recognized that the transcription factor NK2 homeobox 1 (NKX2-1) possesses properties of particular interest for schizophrenia. NKX2-1 is selectively expressed from prenatal development to adulthood in the brain, thyroid gland, parathyroid gland, lungs, skin, and enteric ganglia, and has key functions at the interface of the brain, the endocrine-, and the immune system. In the developing brain, NKX2-1-expressing progenitor cells differentiate into distinct subclasses of forebrain GABAergic and cholinergic neurons, astrocytes, and oligodendrocytes. The transcription factor is highly expressed in mature limbic circuits related to context-dependent goal-directed patterns of behavior, social interaction and reproduction, fear responses, responses to light, and other homeostatic processes. It is essential for development and mature function of the thyroid gland and the respiratory system, and is involved in calcium metabolism and immune responses. NKX2-1 interacts with a number of genes identified as susceptibility genes for schizophrenia. We suggest that NKX2-1 may lie at the core of several dose dependent pathways that are dysregulated in schizophrenia. We correlate the symptoms seen in schizophrenia with the temporal and spatial activities of NKX2-1 in order to highlight promising future research areas.
Collapse
Affiliation(s)
- Eva A Malt
- Department of Adult Habilitation, Akershus University HospitalLørenskog, Norway; Institute of Clinical Medicine, Ahus Campus University of OsloOslo, Norway
| | - Katalin Juhasz
- Department of Adult Habilitation, Akershus University Hospital Lørenskog, Norway
| | - Ulrik F Malt
- Institute of Clinical Medicine, University of OsloOslo, Norway; Department of Research and Education, Institution of Oslo University HospitalOslo, Norway
| | - Thomas Naumann
- Centre of Anatomy, Institute of Cell Biology and Neurobiology, Charite Universitätsmedizin Berlin Berlin, Germany
| |
Collapse
|
35
|
Abstract
The neuroendocrine hypothalamus is composed of the tuberal and anterodorsal hypothalamus, together with the median eminence/neurohypophysis. It centrally governs wide-ranging physiological processes, including homeostasis of energy balance, circadian rhythms and stress responses, as well as growth and reproductive behaviours. Homeostasis is maintained by integrating sensory inputs and effecting responses via autonomic, endocrine and behavioural outputs, over diverse time-scales and throughout the lifecourse of an individual. Here, we summarize studies that begin to reveal how different territories and cell types within the neuroendocrine hypothalamus are assembled in an integrated manner to enable function, thus supporting the organism's ability to survive and thrive. We discuss how signaling pathways and transcription factors dictate the appearance and regionalization of the hypothalamic primordium, the maintenance of progenitor cells, and their specification and differentiation into neurons. We comment on recent studies that harness such programmes for the directed differentiation of human ES/iPS cells. We summarize how developmental plasticity is maintained even into adulthood and how integration between the hypothalamus and peripheral body is established in the median eminence and neurohypophysis. Analysis of model organisms, including mouse, chick and zebrafish, provides a picture of how complex, yet elegantly coordinated, developmental programmes build glial and neuronal cells around the third ventricle of the brain. Such conserved processes enable the hypothalamus to mediate its function as a central integrating and response-control mediator for the homeostatic processes that are critical to life. Early indications suggest that deregulation of these events may underlie multifaceted pathological conditions and dysfunctional physiology in humans, such as obesity.
Collapse
Affiliation(s)
- Sarah Burbridge
- Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Iain Stewart
- Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Marysia Placzek
- Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
36
|
McPherson AD, Barrios JP, Luks-Morgan SJ, Manfredi JP, Bonkowsky JL, Douglass AD, Dorsky RI. Motor Behavior Mediated by Continuously Generated Dopaminergic Neurons in the Zebrafish Hypothalamus Recovers after Cell Ablation. Curr Biol 2016; 26:263-269. [PMID: 26774784 DOI: 10.1016/j.cub.2015.11.064] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 10/14/2015] [Accepted: 11/24/2015] [Indexed: 12/21/2022]
Abstract
Postembryonic neurogenesis has been observed in several regions of the vertebrate brain, including the dentate gyrus and rostral migratory stream in mammals, and is required for normal behavior [1-3]. Recently, the hypothalamus has also been shown to undergo continuous neurogenesis as a way to mediate energy balance [4-10]. As the hypothalamus regulates multiple functional outputs, it is likely that additional behaviors may be affected by postembryonic neurogenesis in this brain structure. Here, we have identified a progenitor population in the zebrafish hypothalamus that continuously generates neurons that express tyrosine hydroxylase 2 (th2). We develop and use novel transgenic tools to characterize the lineage of th2(+) cells and demonstrate that they are dopaminergic. Through genetic ablation and optogenetic activation, we then show that th2(+) neurons modulate the initiation of swimming behavior in zebrafish larvae. Finally, we find that the generation of new th2(+) neurons following ablation correlates with restoration of normal behavior. This work thus identifies for the first time a population of dopaminergic neurons that regulates motor behavior capable of functional recovery.
Collapse
Affiliation(s)
- Adam D McPherson
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Joshua P Barrios
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Sasha J Luks-Morgan
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - John P Manfredi
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA
| | - Joshua L Bonkowsky
- Department of Pediatrics, University of Utah, Salt Lake City, UT 84112, USA
| | - Adam D Douglass
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA.
| | - Richard I Dorsky
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
37
|
Rbpj-κ mediated Notch signaling plays a critical role in development of hypothalamic Kisspeptin neurons. Dev Biol 2015; 406:235-46. [PMID: 26318021 DOI: 10.1016/j.ydbio.2015.08.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 08/21/2015] [Accepted: 08/24/2015] [Indexed: 02/06/2023]
Abstract
The mammalian arcuate nucleus (ARC) houses neurons critical for energy homeostasis and sexual maturation. Proopiomelanocortin (POMC) and Neuropeptide Y (NPY) neurons function to balance energy intake and Kisspeptin neurons are critical for the onset of puberty and reproductive function. While the physiological roles of these neurons have been well established, their development remains unclear. We have previously shown that Notch signaling plays an important role in cell fate within the ARC of mice. Active Notch signaling prevented neural progenitors from differentiating into feeding circuit neurons, whereas conditional loss of Notch signaling lead to a premature differentiation of these neurons. Presently, we hypothesized that Kisspeptin neurons would similarly be affected by Notch manipulation. To address this, we utilized mice with a conditional deletion of the Notch signaling co-factor Rbpj-κ (Rbpj cKO), or mice persistently expressing the Notch1 intracellular domain (NICD tg) within Nkx2.1 expressing cells of the developing hypothalamus. Interestingly, we found that in both models, a lack of Kisspeptin neurons are observed. This suggests that Notch signaling must be properly titrated for formation of Kisspeptin neurons. These results led us to hypothesize that Kisspeptin neurons of the ARC may arise from a different lineage of intermediate progenitors than NPY neurons and that Notch was responsible for the fate choice between these neurons. To determine if Kisspeptin neurons of the ARC differentiate similarly through a Pomc intermediate, we utilized a genetic model expressing the tdTomato fluorescent protein in all cells that have ever expressed Pomc. We observed some Kisspeptin expressing neurons labeled with the Pomc reporter similar to NPY neurons, suggesting that these distinct neurons can arise from a common progenitor. Finally, we hypothesized that temporal differences leading to premature depletion of progenitors in cKO mice lead to our observed phenotype. Using a BrdU birthdating paradigm, we determined the percentage of NPY and Kisspeptin neurons born on embryonic days 11.5, 12.5, and 13.5. We found no difference in the timing of differentiation of either neuronal subtype, with a majority occurring at e11.5. Taken together, our findings suggest that active Notch signaling is an important molecular switch involved in instructing subpopulations of progenitor cells to differentiate into Kisspeptin neurons.
Collapse
|
38
|
Ozek C, Zimmer DJ, De Jonghe BC, Kalb RG, Bence KK. Ablation of intact hypothalamic and/or hindbrain TrkB signaling leads to perturbations in energy balance. Mol Metab 2015; 4:867-80. [PMID: 26629410 PMCID: PMC4632115 DOI: 10.1016/j.molmet.2015.08.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 08/10/2015] [Accepted: 08/11/2015] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Brain-derived neurotrophic factor (BDNF) and its receptor, tropomyosin receptor kinase B (TrkB), play a paramount role in the central regulation of energy balance. Despite the substantial body of genetic evidence implicating BDNF- or TrkB-deficiency in human obesity, the critical brain region(s) contributing to the endogenous role of BDNF/TrkB signaling in metabolic control remain unknown. METHODS We assessed the importance of intact hypothalamic or hindbrain TrkB signaling in central regulation of energy balance by generating Nkx2.1-Ntrk2-/- and Phox2b-Ntrk2+/- mice, respectively, and comparing metabolic parameters (body weight, adiposity, food intake, energy expenditure and glucose homeostasis) under high-fat diet or chow fed conditions. RESULTS Our data show that when fed a high-fat diet, male and female Nkx2.1-Ntrk2-/- mice have significantly increased body weight and adiposity that is likely driven by reduced locomotor activity and core body temperature. When maintained on a chow diet, female Nkx2.1-Ntrk2-/- mice exhibit an increased body weight and adiposity phenotype more robust than in males, which is accompanied by hyperphagia that precedes the onset of a body weight difference. In addition, under both diet conditions, Nkx2.1-Ntrk2-/- mice show increased blood glucose, serum insulin and leptin levels. Mice with complete hindbrain TrkB-deficiency (Phox2b-Ntrk2-/-) are perinatal lethal, potentially indicating a vital role for TrkB in visceral motor neurons that control cardiovascular, respiratory, and digestive functions during development. Phox2b-Ntrk2+/- heterozygous mice are similar in body weight, adiposity and glucose homeostasis parameters compared to wild type littermate controls when maintained on a high-fat or chow diet. Interestingly, despite the absence of a body weight difference, Phox2b-Ntrk2+/- heterozygous mice exhibit pronounced hyperphagia. CONCLUSION Taken together, our findings suggest that the hypothalamus is a key brain region involved in endogenous BDNF/TrkB signaling and central metabolic control and that endogenous hindbrain TrkB likely plays a role in modulating food intake and survival of mice. Our findings also show that female mice lacking TrkB in the hypothalamus have a more robust metabolic phenotype.
Collapse
Key Words
- Agrp, agouti-related peptide
- BAT, brown adipose tissue
- BDNF
- BDNF, brain-derived neurotrophic factor
- Cidea, cell death-inducing DFFA-like effector a
- Cre, Cre recombinase
- DVC, dorsal vagal complex
- Elovl3, elongation of very long fatty acids-like 3
- GTT, glucose tolerance test
- HFD, high-fat diet
- HPA axis, hypothalamic-pituitary-adrenal axis
- Hindbrain
- Hypothalamus
- LepR, leptin receptor
- Mc4R, melanocortin 4 receptor
- NTS, nucleus of the solitary tract
- Nkx2.1, Nk2 homeobox 1 protein
- Npy, neuropeptide Y
- Obesity
- PVH, paraventricular nucleus of the hypothalamus
- Pgc1α, peroxisome proliferator-activated receptor gamma coactivator 1 alpha
- Phox2b, paired-like homeobox 2b protein
- Pomc, pro-opiomelanocortin
- Pparγ, peroxisome proliferator-activated receptor gamma
- Prdm16, PR domain containing 16
- TrkB
- TrkB, tropomyosin receptor kinase B
- Ucp1, uncoupling protein 1
- VMH, ventromedial nucleus of the hypothalamus
- eWAT, epididymal white adipose tissue
Collapse
Affiliation(s)
- Ceren Ozek
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Derek J Zimmer
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Bart C De Jonghe
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Robert G Kalb
- Department of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Kendra K Bence
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
39
|
Sokolowski K, Esumi S, Hirata T, Kamal Y, Tran T, Lam A, Oboti L, Brighthaupt SC, Zaghlula M, Martinez J, Ghimbovschi S, Knoblach S, Pierani A, Tamamaki N, Shah NM, Jones KS, Corbin JG. Specification of select hypothalamic circuits and innate behaviors by the embryonic patterning gene dbx1. Neuron 2015; 86:403-16. [PMID: 25864637 DOI: 10.1016/j.neuron.2015.03.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 12/29/2014] [Accepted: 03/03/2015] [Indexed: 12/30/2022]
Abstract
The hypothalamus integrates information required for the production of a variety of innate behaviors such as feeding, mating, aggression, and predator avoidance. Despite an extensive knowledge of hypothalamic function, how embryonic genetic programs specify circuits that regulate these behaviors remains unknown. Here, we find that in the hypothalamus the developmentally regulated homeodomain-containing transcription factor Dbx1 is required for the generation of specific subclasses of neurons within the lateral hypothalamic area/zona incerta (LH) and the arcuate (Arc) nucleus. Consistent with this specific developmental role, Dbx1 hypothalamic-specific conditional-knockout mice display attenuated responses to predator odor and feeding stressors but do not display deficits in other innate behaviors such as mating or conspecific aggression. Thus, activity of a single developmentally regulated gene, Dbx1, is a shared requirement for the specification of hypothalamic nuclei governing a subset of innate behaviors. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Katie Sokolowski
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Shigeyuki Esumi
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA; Graduate School of Medical Sciences, Kumamoto University, 2-39-1 Kurokami, Chuo Ward, Kumamoto, Kumamoto Prefecture 860-0862, Japan
| | - Tsutomu Hirata
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Yasman Kamal
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Tuyen Tran
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Andrew Lam
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Livio Oboti
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Sherri-Chanelle Brighthaupt
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Manar Zaghlula
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Jennifer Martinez
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Svetlana Ghimbovschi
- Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Susan Knoblach
- Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA
| | - Alessandra Pierani
- Institut Jacques Monod, Universite Paris Diderot, 15 rue Hélène Brion, 75013 Paris, France
| | - Nobuaki Tamamaki
- Graduate School of Medical Sciences, Kumamoto University, 2-39-1 Kurokami, Chuo Ward, Kumamoto, Kumamoto Prefecture 860-0862, Japan
| | - Nirao M Shah
- Department of Anatomy, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Kevin S Jones
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA; Department of Biology, Howard University, 415 College Street NW, Washington, DC 20059, USA
| | - Joshua G Corbin
- Center for Neuroscience Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010, USA.
| |
Collapse
|
40
|
Bedont JL, Newman EA, Blackshaw S. Patterning, specification, and differentiation in the developing hypothalamus. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2015; 4:445-68. [PMID: 25820448 DOI: 10.1002/wdev.187] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 02/10/2015] [Accepted: 02/12/2015] [Indexed: 12/21/2022]
Abstract
Owing to its complex structure and highly diverse cell populations, the study of hypothalamic development has historically lagged behind that of other brain regions. However, in recent years, a greatly expanded understanding of hypothalamic gene expression during development has opened up new avenues of investigation. In this review, we synthesize existing work to present a holistic picture of hypothalamic development from early induction and patterning through nuclear specification and differentiation, with a particular emphasis on determination of cell fate. We will also touch on special topics in the field including the prosomere model, adult neurogenesis, and integration of migratory cells originating outside the hypothalamic neuroepithelium, and how these topics relate to our broader theme.
Collapse
Affiliation(s)
- Joseph L Bedont
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth A Newman
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Seth Blackshaw
- 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 Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,High-Throughput Biology Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
41
|
Lee DK, Jeong JH, Chun SK, Chua S, Jo YH. Interplay between glucose and leptin signalling determines the strength of GABAergic synapses at POMC neurons. Nat Commun 2015; 6:6618. [PMID: 25808323 PMCID: PMC4375782 DOI: 10.1038/ncomms7618] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 02/12/2015] [Indexed: 01/31/2023] Open
Abstract
Regulation of GABAergic inhibitory inputs and alterations in POMC neuron activity by nutrients and adiposity signals regulate energy and glucose homeostasis. Thus, understanding how POMC neurons integrate these two signal molecules at the synaptic level is important. Here we show that leptin’s action on GABA release to POMC neurons is influenced by glucose levels. Leptin stimulates the JAK2-PI3K pathway in both presynaptic GABAergic terminals and postsynaptic POMC neurons. Inhibition of AMPK activity in presynaptic terminals decreases GABA release at 10 mM glucose. However, postsynaptic TRPC channel opening by the PI3K-PLC signaling pathway in POMC neurons enhances spontaneous GABA release via activation of presynaptic MC3/4 and mGlu receptors at 2.5 mM glucose. High-fat feeding blunts AMPK-dependent presynaptic inhibition, whereas PLC-mediated GABAergic feedback inhibition remains responsive to leptin. Our data indicate that the interplay between glucose and leptin signaling in glutamatergic POMC neurons is critical for determining the strength of inhibitory tone towards POMC neurons.
Collapse
Affiliation(s)
- Dong Kun Lee
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, New York 10461, USA
| | - Jae Hoon Jeong
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, New York 10461, USA
| | - Sung-Kun Chun
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, New York 10461, USA
| | - Streamson Chua
- Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, New York 10461, USA
| | - Young-Hwan Jo
- 1] Division of Endocrinology, Department of Medicine, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, New York 10461, USA [2] Department of Molecular Pharmacology, Albert Einstein College of Medicine of Yeshiva University, 1300 Morris Park Avenue, Bronx, New York 10461, USA
| |
Collapse
|
42
|
A novel anxiogenic role for the delta opioid receptor expressed in GABAergic forebrain neurons. Biol Psychiatry 2015; 77:404-15. [PMID: 25444168 PMCID: PMC4297504 DOI: 10.1016/j.biopsych.2014.07.033] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 07/15/2014] [Accepted: 07/24/2014] [Indexed: 01/10/2023]
Abstract
BACKGROUND The delta opioid receptor (DOR) is broadly expressed throughout the nervous system; it regulates chronic pain, emotional responses, motivation, and memory. Neural circuits underlying DOR activities have been poorly explored by genetic approaches. We used conditional mouse mutagenesis to elucidate receptor function in GABAergic neurons of the forebrain. METHODS We characterized DOR distribution in the brain of Dlx5/6-CreXOprd1(fl/fl) (Dlx-DOR) mice and tested main central DOR functions through behavioral testing. RESULTS The DOR proteins were strongly deleted in olfactory bulb and striatum and remained intact in cortex and basolateral amygdala. Olfactory perception, circadian activity, and despair-like behaviors were unchanged. In contrast, locomotor stimulant effects of SNC80 (DOR agonist) and SKF81297 (D1 agonist) were abolished and increased, respectively. The Dlx-DOR mice showed lower levels of anxiety in the elevated plus maze, opposing the known high anxiety in constitutive DOR knockout animals. Also, Dlx-DOR mice reached the food more rapidly in a novelty suppressed feeding task, despite their lower motivation for food reward observed in an operant paradigm. Finally, c-fos protein staining after novelty suppressed feeding was strongly reduced in amygdala, concordant with the low anxiety phenotype of Dlx-DOR mice. CONCLUSIONS We demonstrate that DORs expressed in the forebrain mediate the described locomotor effect of SNC80 and inhibit D1-stimulated hyperactivity. Our data also reveal an unanticipated anxiogenic role for this particular DOR subpopulation, with a potential novel adaptive role. In emotional responses, DORs exert dual anxiolytic and anxiogenic roles, both of which may have implications in the area of anxiety disorders.
Collapse
|
43
|
Domínguez L, González A, Moreno N. Patterns of hypothalamic regionalization in amphibians and reptiles: common traits revealed by a genoarchitectonic approach. Front Neuroanat 2015; 9:3. [PMID: 25691860 PMCID: PMC4315040 DOI: 10.3389/fnana.2015.00003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 01/09/2015] [Indexed: 01/05/2023] Open
Abstract
Most studies in mammals and birds have demonstrated common patterns of hypothalamic development highlighted by the combination of developmental regulatory genes (genoarchitecture), supporting the notion of the hypothalamus as a component of the secondary prosencephalon, topologically rostral to the diencephalon. In our comparative analysis we have summarized the data on the expression patterns of different transcription factors and neuroactive substances, used as anatomical markers, in the developing hypothalamus of the amphibian Xenopus laevis and the juvenile turtle Pseudemys scripta. This analysis served to highlight the organization of the hypothalamus in the anamniote/amniotic transition. We have identified supraoptoparaventricular and the suprachiasmatic regions (SCs) in the alar part of the hypothalamus, and tuberal and mammillary regions in the basal hypothalamus. Shared features in the two species are: (1) The supraoptoparaventricular region (SPV) is defined by the expression of Otp and the lack of Nkx2.1/Isl1. It is subdivided into rostral, rich in Otp and Nkx2.2, and caudal, only Otp-positive, portions. (2) The suprachiasmatic area contains catecholaminergic cell groups and lacks Otp, and can be further divided into rostral (rich in Nkx2.1 and Nkx2.2) and a caudal (rich in Isl1 and devoid of Nkx2.1) portions. (3) Expression of Nkx2.1 and Isl1 define the tuberal hypothalamus and only the rostral portion expresses Otp. (4) Its caudal boundary is evident by the lack of Isl1 in the adjacent mammillary region, which expresses Nkx2.1 and Otp. Differences in the anamnio-amniote transition were noted since in the turtle, like in other amniotes, the boundary between the alar hypothalamus and the telencephalic preoptic area shows distinct Nkx2.2 and Otp expressions but not in the amphibian (anamniote), and the alar SPV is defined by the expression of Otp/Pax6, whereas in Xenopus only Otp is expressed.
Collapse
Affiliation(s)
- Laura Domínguez
- Faculty of Biology, Department of Cell Biology, University Complutense of Madrid Madrid, Spain
| | - Agustín González
- Faculty of Biology, Department of Cell Biology, University Complutense of Madrid Madrid, Spain
| | - Nerea Moreno
- Faculty of Biology, Department of Cell Biology, University Complutense of Madrid Madrid, Spain
| |
Collapse
|
44
|
Díaz C, Morales-Delgado N, Puelles L. Ontogenesis of peptidergic neurons within the genoarchitectonic map of the mouse hypothalamus. Front Neuroanat 2015; 8:162. [PMID: 25628541 PMCID: PMC4290630 DOI: 10.3389/fnana.2014.00162] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 12/12/2014] [Indexed: 11/13/2022] Open
Abstract
During early development, the hypothalamic primordium undergoes anteroposterior and dorsoventral regionalization into diverse progenitor domains, each characterized by a differential gene expression code. The types of neurons produced selectively in each of these distinct progenitor domains are still poorly understood. Recent analysis of the ontogeny of peptidergic neuronal populations expressing Sst, Ghrh, Crh and Trh mRNAs in the mouse hypothalamus showed that these cell types originate from particular dorsoventral domains, characterized by specific combinations of gene markers. Such analysis implies that the differentiation of diverse peptidergic cell populations depends on the molecular environment where they are born. Moreover, a number of these peptidergic neurons were observed to migrate radially and/or tangentially, invading different adult locations, often intermingled with other cell types. This suggests that a developmental approach is absolutely necessary for the understanding of their adult distribution. In this essay, we examine comparatively the ontogenetic hypothalamic topography of twelve additional peptidergic populations documented in the Allen Developmental Mouse Brain Atlas, and discuss shared vs. variant aspects in their apparent origins, migrations and final distribution, in the context of the respective genoarchitectonic backgrounds. This analysis should aid ulterior attempts to explain causally the development of neuronal diversity in the hypothalamus, and contribute to our understanding of its topographic complexity in the adult.
Collapse
Affiliation(s)
- Carmen Díaz
- Department of Medical Sciences, School of Medicine and Institute for Research in Neurological Disabilities, University of Castilla-La Mancha Albacete, Spain
| | - Nicanor Morales-Delgado
- Department of Human Anatomy and Psychobiology, University of Murcia, School of Medicine and IMIB (Instituto Murciano de Investigación Biosanitaria) Murcia, Spain
| | - Luis Puelles
- Department of Human Anatomy and Psychobiology, University of Murcia, School of Medicine and IMIB (Instituto Murciano de Investigación Biosanitaria) Murcia, Spain
| |
Collapse
|
45
|
Neuromolecular responses to social challenge: common mechanisms across mouse, stickleback fish, and honey bee. Proc Natl Acad Sci U S A 2014; 111:17929-34. [PMID: 25453090 DOI: 10.1073/pnas.1420369111] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Certain complex phenotypes appear repeatedly across diverse species due to processes of evolutionary conservation and convergence. In some contexts like developmental body patterning, there is increased appreciation that common molecular mechanisms underlie common phenotypes; these molecular mechanisms include highly conserved genes and networks that may be modified by lineage-specific mutations. However, the existence of deeply conserved mechanisms for social behaviors has not yet been demonstrated. We used a comparative genomics approach to determine whether shared neuromolecular mechanisms could underlie behavioral response to territory intrusion across species spanning a broad phylogenetic range: house mouse (Mus musculus), stickleback fish (Gasterosteus aculeatus), and honey bee (Apis mellifera). Territory intrusion modulated similar brain functional processes in each species, including those associated with hormone-mediated signal transduction and neurodevelopment. Changes in chromosome organization and energy metabolism appear to be core, conserved processes involved in the response to territory intrusion. We also found that several homologous transcription factors that are typically associated with neural development were modulated across all three species, suggesting that shared neuronal effects may involve transcriptional cascades of evolutionarily conserved genes. Furthermore, immunohistochemical analyses of a subset of these transcription factors in mouse again implicated modulation of energy metabolism in the behavioral response. These results provide support for conserved genetic "toolkits" that are used in independent evolutions of the response to social challenge in diverse taxa.
Collapse
|
46
|
Domínguez L, González A, Moreno N. Characterization of the hypothalamus of Xenopus laevis during development. II. The basal regions. J Comp Neurol 2014; 522:1102-31. [PMID: 24122702 DOI: 10.1002/cne.23471] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 07/25/2013] [Accepted: 09/13/2013] [Indexed: 01/10/2023]
Abstract
The expression patterns of conserved developmental regulatory transcription factors and neuronal markers were analyzed in the basal hypothalamus of Xenopus laevis throughout development by means of combined immunohistochemical and in situ hybridization techniques. The connectivity of the main subdivisions was investigated by in vitro tracing techniques with dextran amines. The basal hypothalamic region is topologically rostral to the basal diencephalon and is composed of the tuberal (rostral) and mammillary (caudal) subdivisions, according to the prosomeric model. It is dorsally bounded by the optic chiasm and the alar hypothalamus, and caudally by the diencephalic prosomere p3. The tuberal hypothalamus is defined by the expression of Nkx2.1, xShh, and Isl1, and rostral and caudal portions can be distinguished by the distinct expression of Otp rostrally and Nkx2.2 caudally. In the mammillary region the xShh/Nkx2.1 combination defined the rostral mammillary area, expressing Nkx2.1, and the caudal retromammillary area, expressing xShh. The expression of xLhx1, xDll4, and Otp in the mammillary area and Isl1 in the tuberal region highlights the boundary between the two basal hypothalamic territories. Both regions are strongly connected with subpallial regions, especially those conveying olfactory/vomeronasal information, and also possess abundant intrahypothalamic connections. They show reciprocal connections with the diencephalon (mainly the thalamus), project to the midbrain tectum, and are bidirectionally related to the rhombencephalon. These results illustrate that the basal hypothalamus of anurans shares many features of specification, regionalization, and hodology with amniotes, reinforcing the idea of a basic bauplan in the organization of this prosencephalic region in all tetrapods.
Collapse
Affiliation(s)
- Laura Domínguez
- Faculty of Biology, Department of Cell Biology, University Complutense of Madrid, Madrid, Spain
| | | | | |
Collapse
|
47
|
Heinrich G, Meece K, Wardlaw SL, Accili D. Preserved energy balance in mice lacking FoxO1 in neurons of Nkx2.1 lineage reveals functional heterogeneity of FoxO1 signaling within the hypothalamus. Diabetes 2014; 63:1572-82. [PMID: 24487022 PMCID: PMC3994959 DOI: 10.2337/db13-0651] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Transcription factor forkhead box O1 (FoxO1) regulates energy expenditure (EE), food intake, and hepatic glucose production. These activities have been mapped to specific hypothalamic neuronal populations using cell type-specific knockout experiments in mice. To parse out the integrated output of FoxO1-dependent transcription from different neuronal populations and multiple hypothalamic regions, we used transgenic mice expressing Cre recombinase from the Nkx2.1 promoter to ablate loxP-flanked Foxo1 alleles from a majority of hypothalamic neurons (Foxo1KO(Nkx2.1) mice). This strategy resulted in the expected inhibition of FoxO1 expression, but only produced a transient reduction of body weight as well as a decreased body length. The transient decrease of body weight in male mice was accompanied by decreased fat mass. Male Foxo1KO(Nkx2.1) mice show food intake similar to that in wild-type controls, and, although female knockout mice eat less, they do so in proportion to a reduced body size. EE is unaffected in Foxo1KO(Nkx2.1) mice, although small increases in body temperature are present. Unlike other neuron-specific Foxo1 knockout mice, Foxo1KO(Nkx2.1) mice are not protected from diet-induced obesity. These studies indicate that, unlike the metabolic effects of highly restricted neuronal subsets (proopiomelanocortin, neuropeptide Y/agouti-related peptide, and steroidogenic factor 1), those of neurons derived from the Nkx2.1 lineage either occur in a FoxO1-independent fashion or are compensated for through developmental plasticity.
Collapse
Affiliation(s)
- Garrett Heinrich
- Berrie Diabetes Center, Department of Medicine, College of Physicians & Surgeons, Columbia University, New York, NY
| | | | | | | |
Collapse
|
48
|
Directed differentiation and functional maturation of cortical interneurons from human embryonic stem cells. Cell Stem Cell 2014; 12:559-72. [PMID: 23642365 DOI: 10.1016/j.stem.2013.04.008] [Citation(s) in RCA: 431] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 03/02/2013] [Accepted: 04/12/2013] [Indexed: 12/16/2022]
Abstract
Human pluripotent stem cells are a powerful tool for modeling brain development and disease. The human cortex is composed of two major neuronal populations: projection neurons and local interneurons. Cortical interneurons comprise a diverse class of cell types expressing the neurotransmitter GABA. Dysfunction of cortical interneurons has been implicated in neuropsychiatric diseases, including schizophrenia, autism, and epilepsy. Here, we demonstrate the highly efficient derivation of human cortical interneurons in an NKX2.1::GFP human embryonic stem cell reporter line. Manipulating the timing of SHH activation yields three distinct GFP+ populations with specific transcriptional profiles, neurotransmitter phenotypes, and migratory behaviors. Further differentiation in a murine cortical environment yields parvalbumin- and somatostatin-expressing neurons that exhibit synaptic inputs and electrophysiological properties of cortical interneurons. Our study defines the signals sufficient for modeling human ventral forebrain development in vitro and lays the foundation for studying cortical interneuron involvement in human disease pathology.
Collapse
|
49
|
Delaere F, Akaoka H, De Vadder F, Duchampt A, Mithieux G. Portal glucose influences the sensory, cortical and reward systems in rats. Eur J Neurosci 2013; 38:3476-86. [DOI: 10.1111/ejn.12354] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 08/06/2013] [Accepted: 08/08/2013] [Indexed: 12/01/2022]
Affiliation(s)
- Fabien Delaere
- Institut National de la Santé et de la Recherche Médicale; U855; Lyon France
- Université Lyon 1; Villeurbanne France
- Université de Lyon; Lyon France
- AgroParisTech; ENGREF; Paris France
| | - Hideo Akaoka
- Université Lyon 1; Villeurbanne France
- Université de Lyon; Lyon France
- Lyon Neuroscience Research Center; INSERM U1028-CNRS UMR 5292; Lyon France
| | - Filipe De Vadder
- Institut National de la Santé et de la Recherche Médicale; U855; Lyon France
- Université Lyon 1; Villeurbanne France
- Université de Lyon; Lyon France
| | - Adeline Duchampt
- Institut National de la Santé et de la Recherche Médicale; U855; Lyon France
- Université Lyon 1; Villeurbanne France
- Université de Lyon; Lyon France
| | - Gilles Mithieux
- Institut National de la Santé et de la Recherche Médicale; U855; Lyon France
- Université Lyon 1; Villeurbanne France
- Université de Lyon; Lyon France
| |
Collapse
|
50
|
Aujla PK, Naratadam GT, Xu L, Raetzman LT. Notch/Rbpjκ signaling regulates progenitor maintenance and differentiation of hypothalamic arcuate neurons. Development 2013; 140:3511-21. [PMID: 23884446 DOI: 10.1242/dev.098681] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The hypothalamic arcuate nucleus (Arc), containing pro-opoiomelanocortin (POMC), neuropeptide Y (NPY) and growth hormone releasing hormone (GHRH) neurons, regulates feeding, energy balance and body size. Dysregulation of this homeostatic mediator underlies diseases ranging from growth failure to obesity. Despite considerable investigation regarding the function of Arc neurons, mechanisms governing their development remain unclear. Notch signaling factors such as Hes1 and Mash1 are present in hypothalamic progenitors that give rise to Arc neurons. However, how Notch signaling controls these progenitor populations is unknown. To elucidate the role of Notch signaling in Arc development, we analyzed conditional loss-of-function mice lacking a necessary Notch co-factor, Rbpjκ, in Nkx2.1-cre-expressing cells (Rbpjκ cKO), as well as mice with expression of the constitutively active Notch1 intracellular domain (NICD) in Nkx2.1-cre-expressing cells (NICD Tg). We found that loss of Rbpjκ results in absence of Hes1 but not of Hes5 within the primordial Arc at E13.5. Additionally, Mash1 expression is increased, coincident with increased proliferation and accumulation of Arc neurons at E13.5. At E18.5, Rbpjκ cKO mice have few progenitors and show increased numbers of differentiated Pomc, NPY and Ghrh neurons. By contrast, NICD Tg mice have increased hypothalamic progenitors, show an absence of differentiated Arc neurons and aberrant glial differentiation at E18.5. Subsequently, both Rbpjκ cKO and NICD Tg mice have changes in growth and body size during postnatal development. Taken together, our results demonstrate that Notch/Rbpjκ signaling regulates the generation and differentiation of Arc neurons, which contribute to homeostatic regulation of body size.
Collapse
Affiliation(s)
- Paven K Aujla
- University of Illinois at Urbana-Champaign, 407 South Goodwin Avenue, Urbana, IL 61801, USA
| | | | | | | |
Collapse
|