1
|
Wiegering A, Anselme I, Brunetti L, Metayer-Derout L, Calderon D, Thomas S, Nedelec S, Eschstruth A, Serpieri V, Catala M, Antoniewski C, Schneider-Maunoury S, Stedman A. A differential requirement for ciliary transition zone proteins in human and mouse neural progenitor fate specification. Nat Commun 2025; 16:3258. [PMID: 40188187 PMCID: PMC11972330 DOI: 10.1038/s41467-025-58554-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
Studying ciliary genes in the context of the human central nervous system is crucial for understanding the underlying causes of neurodevelopmental ciliopathies. Here, we use pluripotent stem cell-derived spinal organoids to reveal distinct functions of the ciliopathy gene RPGRIP1L in humans and mice, and uncover an unexplored role for cilia in human axial patterning. Previous research has emphasized Rpgrip1l critical functions in mouse brain and spinal cord development through the regulation of SHH/GLI pathway. Here, we show that RPGRIP1L is not required for SHH activation or motoneuron lineage commitment in human spinal progenitors and that this feature is shared by another ciliopathy gene, TMEM67. Furthermore, human RPGRIP1L-mutant motoneurons adopt hindbrain and cervical identities instead of caudal brachial identity. Temporal transcriptome analysis reveals that this antero-posterior patterning defect originates in early axial progenitors and correlates with cilia loss. These findings provide important insights into the role of cilia in human neural development.
Collapse
Affiliation(s)
- Antonia Wiegering
- Sorbonne Université, CNRS, Inserm, Development, Adaptation and Aging, Dev2A, Paris, France.
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris Seine, Paris, France.
| | - Isabelle Anselme
- Sorbonne Université, CNRS, Inserm, Development, Adaptation and Aging, Dev2A, Paris, France
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris Seine, Paris, France
| | - Ludovica Brunetti
- Sorbonne Université, CNRS, Inserm, Development, Adaptation and Aging, Dev2A, Paris, France
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris Seine, Paris, France
| | - Laura Metayer-Derout
- Sorbonne Université, CNRS, Inserm, Development, Adaptation and Aging, Dev2A, Paris, France
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris Seine, Paris, France
| | - Damelys Calderon
- INSERM UMR 1163, Institut Imagine, Université Paris Cité, Paris, France
| | - Sophie Thomas
- INSERM UMR 1163, Institut Imagine, Université Paris Cité, Paris, France
| | - Stéphane Nedelec
- Sorbonne Université, Inserm, Institut du Fer à Moulin, UMR-S 1270, Paris, France
- Université Paris Cité, CNRS, Inserm U1340, Institut Jacques Monod, Paris, France
| | - Alexis Eschstruth
- Sorbonne Université, CNRS, Inserm, Development, Adaptation and Aging, Dev2A, Paris, France
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris Seine, Paris, France
| | | | - Martin Catala
- Sorbonne Université, CNRS, Inserm, Development, Adaptation and Aging, Dev2A, Paris, France
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris Seine, Paris, France
| | | | - Sylvie Schneider-Maunoury
- Sorbonne Université, CNRS, Inserm, Development, Adaptation and Aging, Dev2A, Paris, France.
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris Seine, Paris, France.
| | - Aline Stedman
- Sorbonne Université, CNRS, Inserm, Development, Adaptation and Aging, Dev2A, Paris, France.
- Sorbonne Université, CNRS, Inserm, Institut de Biologie Paris Seine, Paris, France.
| |
Collapse
|
2
|
Dudoignon B, Bokov P, Benterki F, Couque N, Rand CM, Weese‐Mayer DE, Delclaux C. Reliability of Composite Autonomic Symptom Score (COMPASS)-31 in Congenital Central Hypoventilation Syndrome. Pediatr Pulmonol 2025; 60:e71072. [PMID: 40152101 PMCID: PMC11951072 DOI: 10.1002/ppul.71072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/08/2025] [Indexed: 03/29/2025]
Abstract
RATIONALE Congenital central hypoventilation syndrome (CCHS) is a rare disorder characterized by alveolar hypoventilation and variable autonomic nervous system (ANS) dysfunction (ANSD) due to mutations in PHOX2B, a gene crucial for ANS neural crest lineage differentiation. OBJECTIVES AND METHODS Our prospective study aims were twofold: to (1) assess the relationships between the subjective Composite Autonomic Symptom Score (COMPASS)-31 and objective indices of ANSD obtained from heart rate variability analyses, ambulatory blood pressure (BP) monitoring, and CO2 chemosensitivities and (2) describe the organ system ANSD, its relationship to PHOX2B genotype, and its consequences on quality of life (PedsQL) in children with CCHS. RESULTS Thirty-two PHOX2B mutation-confirmed subjects (median [range] age 9.2 years (4.4; 18.0), 15 girls) were enrolled. COMPASS-31 was assessed in 32 matched (sex and age, range: 4.3; 18.9 years) healthy controls. As compared to healthy controls, children with CCHS had increased vasomotor (p = 0.001), secretomotor (p = 0.021), gastrointestinal (p = 0.002) and pupillomotor (p = 0.028) scores and decreased orthostatic intolerance scores (p = 0.050). There was no difference in overall COMPASS-31 score between CCHS and controls (p = 0.083). However, in CCHS, overall COMPASS-31 scores correlated with high frequencies (HF) normalized (cardiac parasympathetic modulation: R = -0.53; p = 0.002), low frequencies (LF)/HF ratio (R = 0.56; p< 0.001), and both systolic and diastolic nighttime BP dipping (R = 0.45, p = 0.012 and R = 0.40, p = 0.028, respectively). No significant relationships between COMPASS-31 scores and chemosensitivity testing, PedsQL scores, or PHOX2B genotype were identified. CONCLUSIONS COMPASS-31 identified some aspects of CCHS-related ANSD, and scores correlate with objective ANS function measures, supporting the potential utility of COMPASS-31 in CCHS.
Collapse
Affiliation(s)
- Benjamin Dudoignon
- Service de Physiologie Pédiatrique ‐Centre du Sommeil ‐ CRMR Hypoventilations alvéolaires rares, AP‐HP, Hôpital Robert Debré, INSERM NeuroDiderotUniversité de Paris CitéParisFrance
| | - Plamen Bokov
- Service de Physiologie Pédiatrique ‐Centre du Sommeil ‐ CRMR Hypoventilations alvéolaires rares, AP‐HP, Hôpital Robert Debré, INSERM NeuroDiderotUniversité de Paris CitéParisFrance
| | - Fatima Benterki
- Service de Physiologie PédiatriqueAP‐HP, Hôpital Robert DebréParisFrance
| | - Nathalie Couque
- Département de GénétiqueAP‐HP, Hôpital Robert DebréParisFrance
| | - Casey M. Rand
- Division of Autonomic Medicine, Department of PediatricsAnn & Robert H. Lurie Children's Hospital of Chicago and Stanley Manne Children's Research InstituteChicagoIllinoisUSA
| | - Debra E. Weese‐Mayer
- Division of Autonomic Medicine, Department of PediatricsAnn & Robert H. Lurie Children's Hospital of Chicago and Stanley Manne Children's Research InstituteChicagoIllinoisUSA
- Department of PediatricsNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Christophe Delclaux
- Service de Physiologie Pédiatrique ‐Centre du Sommeil ‐ CRMR Hypoventilations alvéolaires rares, AP‐HP, Hôpital Robert Debré, INSERM NeuroDiderotUniversité de Paris CitéParisFrance
| |
Collapse
|
3
|
Huisman BD, Michelson DA, Rubin SA, Kohlsaat K, Gomarga W, Fang Y, Lee JM, Del Nido P, Nathan M, Benoist C, Zon L, Mathis D. Cross-species analyses of thymic mimetic cells reveal evolutionarily ancient origins and both conserved and species-specific elements. Immunity 2025; 58:108-123.e7. [PMID: 39731911 PMCID: PMC11735279 DOI: 10.1016/j.immuni.2024.11.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/19/2024] [Accepted: 11/27/2024] [Indexed: 12/30/2024]
Abstract
Thymic mimetic cells are molecular hybrids between medullary-thymic-epithelial cells (mTECs) and diverse peripheral cell types. They are involved in eliminating autoreactive T cells and can perform supplementary functions reflective of their peripheral-cell counterparts. Current knowledge about mimetic cells derives largely from mouse models. To provide the high resolution that proved revelatory for mice, we performed single-cell RNA sequencing on purified mimetic-cell compartments from human pediatric donors. The single-cell profiles of individual donors were surprisingly similar, with diversification of neuroendocrine subtypes and expansion of the muscle subtype relative to mice. Informatic and imaging studies on the muscle-mTEC population highlighted a maturation trajectory suggestive of skeletal-muscle differentiation, some striated structures, and occasional cellular groupings reminiscent of neuromuscular junctions. We also profiled thymic mimetic cells from zebrafish. Integration of data from the three species identified species-specific adaptations but substantial interspecies conservation, highlighting the evolutionarily ancient nature of mimetic mTECs. Our findings provide a landscape view of human mimetic cells, with anticipated relevance in autoimmunity.
Collapse
Affiliation(s)
- Brooke D Huisman
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Daniel A Michelson
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA; PhD Program in Immunology, Harvard Medical School, Boston, MA, USA
| | - Sara A Rubin
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA; PhD Program in Immunology, Harvard Medical School, Boston, MA, USA; Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Katherine Kohlsaat
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wilson Gomarga
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Yuan Fang
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Ji Myung Lee
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pedro Del Nido
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Meena Nathan
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | | | - Leonard Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA; Howard Hughes Medical Institute and Boston Children's Hospital, Boston, MA, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Gasparini S, Almeida‐Pereira G, Munuzuri ASP, Resch JM, Geerling JC. Molecular Ontology of the Nucleus of Solitary Tract. J Comp Neurol 2024; 532:e70004. [PMID: 39629676 PMCID: PMC11615840 DOI: 10.1002/cne.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/03/2024] [Accepted: 11/15/2024] [Indexed: 12/08/2024]
Abstract
The nucleus of the solitary tract (NTS) receives visceral information and regulates appetitive, digestive, and cardiorespiratory systems. Within the NTS, diverse processes operate in parallel to sustain life, but our understanding of their cellular composition is incomplete. Here, we integrate histologic and transcriptomic analysis to identify and compare molecular features that distinguish neurons in this brain region. Most glutamatergic neurons in the NTS and area postrema co-express the transcription factors Lmx1b and Phox2b, except for a ventral band of neurons in the far-caudal NTS, which include the Gcg-expressing neurons that produce glucagon-like peptide 1 (GLP-1). GABAergic interneurons intermingle through the Lmx1b+Phox2b macropopulation, and dense clusters of GABAergic neurons surround the NTS. The Lmx1b+Phox2b macropopulation includes subpopulations with distinct distributions expressing Grp, Hsd11b2, Npff, Pdyn, Pou3f1, Sctr, Th, and other markers. These findings highlight Lmx1b-Phox2b co-expression as a common feature of glutamatergic neurons in the NTS and improve our understanding of the organization and distribution of neurons in this critical brain region.
Collapse
Affiliation(s)
| | | | | | - Jon M. Resch
- Department of Neuroscience and PharmacologyUniversity of IowaIowa CityIowaUSA
- Iowa Neuroscience InstituteUniversity of IowaIowa CityIowaUSA
| | - Joel C. Geerling
- Department of NeurologyUniversity of IowaIowa CityIowaUSA
- Iowa Neuroscience InstituteUniversity of IowaIowa CityIowaUSA
| |
Collapse
|
5
|
Gigante ED, Piekarz KM, Gurgis A, Cohen L, Razy-Krajka F, Popsuj S, Johnson CJ, Ali HS, Mohana Sundaram S, Stolfi A. Specification and survival of post-metamorphic branchiomeric neurons in a non-vertebrate chordate. Development 2024; 151:dev202719. [PMID: 38895900 PMCID: PMC11273300 DOI: 10.1242/dev.202719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
Tunicates are the sister group to the vertebrates, yet most species have a life cycle split between swimming larva and sedentary adult phases. During metamorphosis, larval neurons are replaced by adult-specific ones. The regulatory mechanisms underlying this replacement remain largely unknown. Using tissue-specific CRISPR/Cas9-mediated mutagenesis in the tunicate Ciona, we show that orthologs of conserved hindbrain and branchiomeric neuron regulatory factors Pax2/5/8 and Phox2 are required to specify the 'neck', a cellular compartment set aside in the larva to give rise to cranial motor neuron-like neurons post-metamorphosis. Using bulk and single-cell RNA-sequencing analyses, we characterize the transcriptome of the neck downstream of Pax2/5/8. We present evidence that neck-derived adult ciliomotor neurons begin to differentiate in the larva and persist through metamorphosis, contrary to the assumption that the adult nervous system is formed after settlement and the death of larval neurons during metamorphosis. Finally, we show that FGF signaling during the larval phase alters the patterning of the neck and its derivatives. Suppression of FGF converts neck cells into larval neurons that fail to survive metamorphosis, whereas prolonged FGF signaling promotes an adult neural stem cell-like fate.
Collapse
Affiliation(s)
- Eduardo D. Gigante
- School of Biological Sciences, College of Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Katarzyna M. Piekarz
- School of Biological Sciences, College of Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Alexandra Gurgis
- School of Biological Sciences, College of Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Leslie Cohen
- School of Biological Sciences, College of Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Florian Razy-Krajka
- School of Biological Sciences, College of Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Sydney Popsuj
- School of Biological Sciences, College of Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Christopher J. Johnson
- School of Biological Sciences, College of Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Hussan S. Ali
- School of Biological Sciences, College of Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Shruthi Mohana Sundaram
- School of Biological Sciences, College of Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Alberto Stolfi
- School of Biological Sciences, College of Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
6
|
Cui K, Xia Y, Patnaik A, Salivara A, Lowenstein ED, Isik EG, Knorz AL, Airaghi L, Crotti M, Garratt AN, Meng F, Schmitz D, Studer M, Rijli FM, Nothwang HG, Rost BR, Strauß U, Hernandez-Miranda LR. Genetic identification of medullary neurons underlying congenital hypoventilation. SCIENCE ADVANCES 2024; 10:eadj0720. [PMID: 38896627 PMCID: PMC11186509 DOI: 10.1126/sciadv.adj0720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 05/14/2024] [Indexed: 06/21/2024]
Abstract
Mutations in the transcription factors encoded by PHOX2B or LBX1 correlate with congenital central hypoventilation disorders. These conditions are typically characterized by pronounced hypoventilation, central apnea, and diminished chemoreflexes, particularly to abnormally high levels of arterial PCO2. The dysfunctional neurons causing these respiratory disorders are largely unknown. Here, we show that distinct, and previously undescribed, sets of medullary neurons coexpressing both transcription factors (dB2 neurons) account for specific respiratory functions and phenotypes seen in congenital hypoventilation. By combining intersectional chemogenetics, intersectional labeling, lineage tracing, and conditional mutagenesis, we uncovered subgroups of dB2 neurons with key functions in (i) respiratory tidal volumes, (ii) the hypercarbic reflex, (iii) neonatal respiratory stability, and (iv) neonatal survival. These data provide functional evidence for the critical role of distinct medullary dB2 neurons in neonatal respiratory physiology. In summary, our work identifies distinct subgroups of dB2 neurons regulating breathing homeostasis, dysfunction of which causes respiratory phenotypes associated with congenital hypoventilation.
Collapse
Affiliation(s)
- Ke Cui
- The Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Yiling Xia
- The Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Abhisarika Patnaik
- The Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Aikaterini Salivara
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - Eser G. Isik
- The Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Adrian L. Knorz
- The Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Laura Airaghi
- The Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Michela Crotti
- The Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alistair N. Garratt
- The Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Fanqi Meng
- The Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dietmar Schmitz
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Michèle Studer
- Université Côte d'Azur (UCA), CNRS, Inserm, Institute of Biology Valrose (iBV), Nice, France
| | - Filippo M. Rijli
- Laboratory of Developmental Neuroepigenetics, Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Hans G. Nothwang
- Division of Neurogenetics, Cluster of Excellence Hearing4all, Carl von Ossietzky University, Oldenburg, Germany
| | - Benjamin R. Rost
- German Center for Neurodegenerative Diseases (DZNE), 10117 Berlin, Germany
- Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulf Strauß
- Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Luis R. Hernandez-Miranda
- The Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
7
|
Cardani S, Janes TA, Betzner W, Pagliardini S. Knockdown of PHOX2B in the retrotrapezoid nucleus reduces the central CO 2 chemoreflex in rats. eLife 2024; 13:RP94653. [PMID: 38727716 PMCID: PMC11087052 DOI: 10.7554/elife.94653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024] Open
Abstract
PHOX2B is a transcription factor essential for the development of different classes of neurons in the central and peripheral nervous system. Heterozygous mutations in the PHOX2B coding region are responsible for the occurrence of Congenital Central Hypoventilation Syndrome (CCHS), a rare neurological disorder characterised by inadequate chemosensitivity and life-threatening sleep-related hypoventilation. Animal studies suggest that chemoreflex defects are caused in part by the improper development or function of PHOX2B expressing neurons in the retrotrapezoid nucleus (RTN), a central hub for CO2 chemosensitivity. Although the function of PHOX2B in rodents during development is well established, its role in the adult respiratory network remains unknown. In this study, we investigated whether reduction in PHOX2B expression in chemosensitive neuromedin-B (NMB) expressing neurons in the RTN altered respiratory function. Four weeks following local RTN injection of a lentiviral vector expressing the short hairpin RNA (shRNA) targeting Phox2b mRNA, a reduction of PHOX2B expression was observed in Nmb neurons compared to both naive rats and rats injected with the non-target shRNA. PHOX2B knockdown did not affect breathing in room air or under hypoxia, but ventilation was significantly impaired during hypercapnia. PHOX2B knockdown did not alter Nmb expression but it was associated with reduced expression of both Task2 and Gpr4, two CO2/pH sensors in the RTN. We conclude that PHOX2B in the adult brain has an important role in CO2 chemoreception and reduced PHOX2B expression in CCHS beyond the developmental period may contribute to the impaired central chemoreflex function.
Collapse
Affiliation(s)
- Silvia Cardani
- Department of Physiology, Faculty of Medicine and Dentistry, University of AlbertaEdmontonCanada
- Women and Children’s Health Research Institute, University of AlbertaEdmontonCanada
| | - Tara A Janes
- Department of Physiology, Faculty of Medicine and Dentistry, University of AlbertaEdmontonCanada
- Women and Children’s Health Research Institute, University of AlbertaEdmontonCanada
| | - William Betzner
- Department of Physiology, Faculty of Medicine and Dentistry, University of AlbertaEdmontonCanada
| | - Silvia Pagliardini
- Department of Physiology, Faculty of Medicine and Dentistry, University of AlbertaEdmontonCanada
- Women and Children’s Health Research Institute, University of AlbertaEdmontonCanada
- Neuroscience and Mental Health Institute, University of AlbertaEdmontonCanada
| |
Collapse
|
8
|
Wu X, Xiu W, Zhou N, Zhang J, Hao X, Dong Q. Identifying a novel PHOX2B gene variant in a neuroblastoma family: A case report. Heliyon 2024; 10:e26581. [PMID: 38420445 PMCID: PMC10900999 DOI: 10.1016/j.heliyon.2024.e26581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
Neuroblastoma is a childhood cancer characterized by the formation of tumors derived from neuroblasts. Identifying the genetic mutations underlying neuroblastoma for genetic counseling and early diagnosis is essential. Thus, this study aimed to screen for pathogenic gene variants within a neuroblastoma family, aiming to contribute to genetic counseling practices. Clinical data was collected from a family affected by neuroblastoma, and peripheral blood DNA samples were obtained from all family members. A combination of whole-exome sequencing and Sanger sequencing was utilized to detect potential gene mutations. Proband 1 and her sister (Proband 2) were diagnosed with neuroblastoma, while their parents and siblings were unaffected. The analysis revealed a novel missense mutation, c.422G > A (p.Arg141Gln), in the PHOX2B gene, which was inherited from the mother. Notably, this mutation represents a previously unreported variant within the PHOX2B gene. Detecting the missense mutation c.422G > A (p.Arg141Gln) in the PHOX2B gene implies its potential pathogenic role within this neuroblastoma family. This finding widens the range of mutations observed in the PHOX2B gene and has important implications for early neuroblastoma diagnosis within this family.
Collapse
Affiliation(s)
- Xiongwei Wu
- Department of Pediatric Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenli Xiu
- Department of Pediatric Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Na Zhou
- Department of Pediatric Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingli Zhang
- Department of Pediatric Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiwei Hao
- Department of Pediatric Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qian Dong
- Department of Pediatric Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Vermeiren S, Cabochette P, Dannawi M, Desiderio S, San José AS, Achouri Y, Kricha S, Sitte M, Salinas-Riester G, Vanhollebeke B, Brunet JF, Bellefroid EJ. Prdm12 represses the expression of the visceral neuron determinants Phox2a/b in developing somatosensory ganglia. iScience 2023; 26:108364. [PMID: 38025786 PMCID: PMC10663820 DOI: 10.1016/j.isci.2023.108364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 10/13/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Prdm12 is a transcriptional regulator essential for the emergence of the somatic nociceptive lineage during sensory neurogenesis. The exact mechanisms by which Prdm12 promotes nociceptor development remain, however, poorly understood. Here, we report that the trigeminal and dorsal root ganglia hypoplasia induced by the loss of Prdm12 involves Bax-dependent apoptosis and that it is accompanied by the ectopic expression of the visceral sensory neuron determinants Phox2a and Phox2b, which is, however, not sufficient to impose a complete fate switch in surviving somatosensory neurons. Mechanistically, our data reveal that Prdm12 is required from somatosensory neural precursors to early post-mitotic differentiating nociceptive neurons to repress Phox2a/b and that its repressive function is context dependent. Together, these findings reveal that besides its essential role in nociceptor survival during development, Prdm12 also promotes nociceptor fate via an additional mechanism, by preventing precursors from engaging into an alternate Phox2 driven visceral neuronal type differentiation program.
Collapse
Affiliation(s)
- Simon Vermeiren
- Department of Molecular Biology, ULB Neuroscience Institute (UNI), Université libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | - Pauline Cabochette
- Department of Molecular Biology, ULB Neuroscience Institute (UNI), Université libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | - Maya Dannawi
- Department of Molecular Biology, ULB Neuroscience Institute (UNI), Université libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | - Simon Desiderio
- Department of Molecular Biology, ULB Neuroscience Institute (UNI), Université libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | - Alba Sabaté San José
- Department of Molecular Biology, ULB Neuroscience Institute (UNI), Université libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | - Younes Achouri
- Transgenesis Platform, de Duve Institute, Université Catholique de Louvain, Institut de Duve, Brussels, Belgium
| | - Sadia Kricha
- Department of Molecular Biology, ULB Neuroscience Institute (UNI), Université libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | - Maren Sitte
- NGS Integrative Genomics, Department of Human Genetics at the University Medical Center Göttingen (UMG), 37075 Göttingen, Germany
| | - Gabriela Salinas-Riester
- NGS Integrative Genomics, Department of Human Genetics at the University Medical Center Göttingen (UMG), 37075 Göttingen, Germany
| | - Benoit Vanhollebeke
- Department of Molecular Biology, ULB Neuroscience Institute (UNI), Université libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| | - Jean-François Brunet
- Institut de Biologie de l’ENS (IBENS), Inserm, CNRS, École Normale Supérieure, PSL Research University, 75005 Paris, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8197, 75005 Paris, France
- Institut National de la Santé et de la Recherche Médicale U1024, 75005 Paris, France
| | - Eric J. Bellefroid
- Department of Molecular Biology, ULB Neuroscience Institute (UNI), Université libre de Bruxelles (ULB), B-6041 Gosselies, Belgium
| |
Collapse
|
10
|
Honzel E, Hernandez-Morato I, Joshi A, Pennington-Fitzgerald W, Moayedi Y, Pitman MJ. Temporal Expression of Hox Genes and Phox2b in the Rat Nucleus Ambiguus During Development: Implications on Laryngeal Innervation. Laryngoscope 2023; 133:3462-3471. [PMID: 37350386 PMCID: PMC10907063 DOI: 10.1002/lary.30826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/24/2023]
Abstract
OBJECTIVES Recurrent laryngeal nerve (RLN) injury results in synkinetic reinnervation and vocal fold paralysis. Investigation of cues expressed in the developing brainstem that influence correct selective targeting of intrinsic laryngeal muscles may elucidate post-injury abnormalities contributing to non-functional reinnervation. Primary targets of interest were Hoxb1 and Hoxb2, members of the Hox family that create overlapping gradients in the developing brain, and their target Phox2b, a transcription factor necessary for cranial nerve branchio- and visceromotoneuron survival. METHODS Rat embryos at developmental days E14, E16, E18, and E20 (4 animals/age) were sectioned for RNA in situ hybridization to detect Hoxb1, Hoxb2, and Phox2b mRNA within the brainstem. Slides were costained with Islet1 antibody for identification of the nucleus ambiguus. Results were confirmed using immunohistochemistry. Sections were imaged on a confocal microscope. RNA and protein expressions were quantified using QuPath. Statistical analyses were performed using R. RESULTS Hoxb1, Hoxb2, and Phox2b expressions varied according to embryologic age. Hoxb1 and Hoxb2 expression peaked at E16, with significant decreases at E18 and E20 (one-way ANOVA p = 0.001 for both). Phox2b expression was highest at E14 and trended downward with increased embryologic age (one-way ANOVA p = 0.005). CONCLUSION Peak expression of Hoxb1 and Hoxb2 is observed at time points when the RLN arrives at the larynx and begins to branch toward individual muscles, positioning these gene products to be involved in cueing laryngeal motoneuron identity and target identification. Higher expression of Phox2b earlier in development suggests a role in laryngeal motoneuron formation. LEVEL OF EVIDENCE NA Laryngoscope, 133:3462-3471, 2023.
Collapse
Affiliation(s)
- Emily Honzel
- Department of Otolaryngology-Head & Neck Surgery, Columbia University College of Physicians and Surgeons, New York, New York, U.S.A
| | - Ignacio Hernandez-Morato
- Department of Otolaryngology-Head & Neck Surgery, Columbia University College of Physicians and Surgeons, New York, New York, U.S.A
| | - Abhinav Joshi
- Department of Otolaryngology-Head & Neck Surgery, Columbia University College of Physicians and Surgeons, New York, New York, U.S.A
| | - William Pennington-Fitzgerald
- Department of Otolaryngology-Head & Neck Surgery, Columbia University College of Physicians and Surgeons, New York, New York, U.S.A
| | - Yalda Moayedi
- Department of Otolaryngology-Head & Neck Surgery, Columbia University College of Physicians and Surgeons, New York, New York, U.S.A
- Department of Neurology, Columbia University, New York, New York, U.S.A
| | - Michael J Pitman
- Department of Otolaryngology-Head & Neck Surgery, Columbia University College of Physicians and Surgeons, New York, New York, U.S.A
- Department of Neurology, Columbia University, New York, New York, U.S.A
| |
Collapse
|
11
|
Gigante ED, Piekarz KM, Gurgis A, Cohen L, Razy-Krajka F, Popsuj S, Ali HS, Sundaram SM, Stolfi A. Specification and survival of post-metamorphic branchiomeric neurons in the hindbrain of a non-vertebrate chordate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.16.545305. [PMID: 37645866 PMCID: PMC10461979 DOI: 10.1101/2023.06.16.545305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Tunicates are the sister group to the vertebrates, yet most species have a life cycle split between swimming larva and sedentary adult phases. During metamorphosis, larval neurons are largely replaced by adult-specific ones. Yet the regulatory mechanisms underlying this neural replacement remain largely unknown. Using tissue-specific CRISPR/Cas9-mediated mutagenesis in the tunicate Ciona, we show that orthologs of conserved hindbrain and branchiomeric neuron regulatory factors Pax2/5/8 and Phox2 are required to specify the "Neck", a compartment of cells set aside in the larva to give rise to cranial motor neuron-like neurons in the adult. Using bulk and single-cell RNAseq analyses, we also characterize the transcriptome of the Neck downstream of Pax2/5/8. Surprisingly, we find that Neck-derived adult ciliomotor neurons begin to differentiate in the larva, contrary to the long-held assumption that the adult nervous system is formed only after settlement and the death of larval neurons during metamorphosis. Finally, we show that manipulating FGF signaling during the larval phase alters the patterning of the Neck and its derivatives. Suppression of FGF converts Neck cells into larval neurons that fail to survive metamorphosis, while prolonged FGF signaling promotes an adult neural stem cell-like fate instead.
Collapse
Affiliation(s)
- Eduardo D Gigante
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332; USA
| | - Katarzyna M Piekarz
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332; USA
| | - Alexandra Gurgis
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332; USA
- Department of Biology, Case Western Reserve University, Cleveland, OH, 44106; USA
| | - Leslie Cohen
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332; USA
| | - Florian Razy-Krajka
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332; USA
| | - Sydney Popsuj
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332; USA
| | - Hussan S Ali
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332; USA
| | | | - Alberto Stolfi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332; USA
| |
Collapse
|
12
|
Lui KNC, Li Z, Lai FPL, Lau ST, Ngan ESW. Organoid models of breathing disorders reveal patterning defect of hindbrain neurons caused by PHOX2B-PARMs. Stem Cell Reports 2023:S2213-6711(23)00199-6. [PMID: 37352849 PMCID: PMC10362500 DOI: 10.1016/j.stemcr.2023.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/24/2023] [Accepted: 05/24/2023] [Indexed: 06/25/2023] Open
Abstract
Retrotrapezoid nucleus (RTN) neurons in the brainstem regulate the ventilatory response to hypercarbia. It is unclear how PHOX2B-polyalanine repeat mutations (PHOX2B-PARMs) alter the function of PHOX2B and perturb the formation of RTN neurons. Here, we generated human brainstem organoids (HBSOs) with RTN-like neurons from human pluripotent stem cells. Single-cell transcriptomics revealed that expression of PHOX2B+7Ala PARM alters the differentiation trajectories of the hindbrain neurons and hampers the formation of the RTN-like neurons in HBSOs. With the unguided cerebral organoids (HCOs), PHOX2B+7Ala PARM interrupted the patterning of PHOX2B+ neurons with dysregulation of Hedgehog pathway and HOX genes. With complementary use of HBSOs and HCOs with a patient and two mutant induced pluripotent stem cell lines carrying different polyalanine repetition in PHOX2B, we further defined the association between the length of polyalanine repetition and malformation of RTN-respiratory center and demonstrated the potential toxic gain of function of PHOX2B-PARMs, highlighting the uniqueness of these organoid models for disease modeling.
Collapse
Affiliation(s)
- Kathy Nga-Chu Lui
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Zhixin Li
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Frank Pui-Ling Lai
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Sin-Ting Lau
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | - Elly Sau-Wai Ngan
- Department of Surgery, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR.
| |
Collapse
|
13
|
Valeri A, Chiricosta L, D’Angiolini S, Pollastro F, Salamone S, Mazzon E. Cannabichromene Induces Neuronal Differentiation in NSC-34 Cells: Insights from Transcriptomic Analysis. Life (Basel) 2023; 13:life13030742. [PMID: 36983897 PMCID: PMC10051538 DOI: 10.3390/life13030742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Phytocannabinoids, with their variety of beneficial effects, represent a valid group of substances that could be employed as neurogenesis-enhancers or neuronal differentiation inducers. We focused our attention on the neuronal-related potential of cannabichromene (CBC) when administered to undifferentiated NSC-34 for 24 h. Transcriptomic analysis showed an upregulation of several neuronal markers, such as Neurod1 and Tubb3, as well as indicators of neuronal differentiation process progression, such as Pax6. An in-depth investigation of the processes involved in neuronal differentiation indicates positive cytoskeleton remodeling by upregulation of Cfl2 and Tubg1, and active differentiation-targeted transcriptional program, suggested by Phox2b and Hes1. After 48 h of treatment, the markers previously examined in the transcriptomic analysis are still overexpressed, like Ache and Hes1, indicating that the differentiation process is still in progress. The lack of GFAP protein suggests that no astroglial differentiation is taking place, and it is reasonable to indicate the neuronal one as the ongoing one. These results indicate CBC as a potential neuronal differentiation inducer for NSC-34 cells.
Collapse
Affiliation(s)
- Andrea Valeri
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Luigi Chiricosta
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Simone D’Angiolini
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Federica Pollastro
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy
- Plantachem S.r.l.s., Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Stefano Salamone
- Department of Pharmaceutical Sciences, University of Eastern Piedmont, Largo Donegani 2, 28100 Novara, Italy
- Plantachem S.r.l.s., Via Amico Canobio 4/6, 28100 Novara, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi “Bonino-Pulejo”, Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
- Correspondence:
| |
Collapse
|
14
|
Xia Y, Cui K, Alonso A, Lowenstein ED, Hernandez-Miranda LR. Transcription factors regulating the specification of brainstem respiratory neurons. Front Mol Neurosci 2022; 15:1072475. [PMID: 36523603 PMCID: PMC9745097 DOI: 10.3389/fnmol.2022.1072475] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/14/2022] [Indexed: 11/12/2023] Open
Abstract
Breathing (or respiration) is an unconscious and complex motor behavior which neuronal drive emerges from the brainstem. In simplistic terms, respiratory motor activity comprises two phases, inspiration (uptake of oxygen, O2) and expiration (release of carbon dioxide, CO2). Breathing is not rigid, but instead highly adaptable to external and internal physiological demands of the organism. The neurons that generate, monitor, and adjust breathing patterns locate to two major brainstem structures, the pons and medulla oblongata. Extensive research over the last three decades has begun to identify the developmental origins of most brainstem neurons that control different aspects of breathing. This research has also elucidated the transcriptional control that secures the specification of brainstem respiratory neurons. In this review, we aim to summarize our current knowledge on the transcriptional regulation that operates during the specification of respiratory neurons, and we will highlight the cell lineages that contribute to the central respiratory circuit. Lastly, we will discuss on genetic disturbances altering transcription factor regulation and their impact in hypoventilation disorders in humans.
Collapse
Affiliation(s)
- Yiling Xia
- The Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ke Cui
- The Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Antonia Alonso
- Functional Genoarchitecture and Neurobiology Groups, Biomedical Research Institute of Murcia (IMIB-Arrixaca), Murcia, Spain
- Department of Human Anatomy and Psychobiology, Faculty of Medicine, University of Murcia, Murcia, Spain
| | - Elijah D. Lowenstein
- Developmental Biology/Signal Transduction, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Luis R. Hernandez-Miranda
- The Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
15
|
Etonogestrel Administration Reduces the Expression of PHOX2B and Its Target Genes in the Solitary Tract Nucleus. Int J Mol Sci 2022; 23:ijms23094816. [PMID: 35563209 PMCID: PMC9101578 DOI: 10.3390/ijms23094816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/15/2022] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
Heterozygous mutations of the transcription factor PHOX2B are responsible for Congenital Central Hypoventilation Syndrome, a neurological disorder characterized by inadequate respiratory response to hypercapnia and life-threatening hypoventilation during sleep. Although no cure is currently available, it was suggested that a potent progestin drug provides partial recovery of chemoreflex response. Previous in vitro data show a direct molecular link between progestins and PHOX2B expression. However, the mechanism through which these drugs ameliorate breathing in vivo remains unknown. Here, we investigated the effects of chronic administration of the potent progestin drug Etonogestrel (ETO) on respiratory function and transcriptional activity in adult female rats. We assessed respiratory function with whole-body plethysmography and measured genomic changes in brain regions important for respiratory control. Our results show that ETO reduced metabolic activity, leading to an enhanced chemoreflex response and concurrent increased breathing cycle variability at rest. Furthermore, ETO-treated brains showed reduced mRNA and protein expression of PHOX2B and its target genes selectively in the dorsal vagal complex, while other areas were unaffected. Histological analysis suggests that changes occurred in the solitary tract nucleus (NTS). Thus, we propose that the NTS, rich in both progesterone receptors and PHOX2B, is a good candidate for ETO-induced respiratory modulation.
Collapse
|
16
|
Chen Z, Liu Y, Liang R, Cui C, Zhu Y, Zhang F, Zhang J, Chen X. Comparative transcriptome analysis provides insights into the molecular mechanisms of high-frequency hearing differences between the sexes of Odorrana tormota. BMC Genomics 2022; 23:296. [PMID: 35410120 PMCID: PMC9004125 DOI: 10.1186/s12864-022-08536-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 04/07/2022] [Indexed: 11/15/2022] Open
Abstract
Background Acoustic communication is important for the survival and reproduction of anurans and masking background noise is a critical factor for their effective acoustic communication. Males of the concave-eared frog (Odorrana tormota) have evolved an ultrasonic communication capacity to avoid masking by the widespread background noise of local fast-flowing streams, whereas females exhibit no ultrasonic sensitivity. However, the molecular mechanisms underlying the high-frequency hearing differences between the sexes of O. tormota are still poorly understood. Results In this study, we sequenced the brain transcriptomes of male and female O. tormota, and compared their differential gene expression. A total of 4,605 differentially expressed genes (DEGs) between the sexes of O. tormota were identified and eleven of them were related to auditory based on the annotation and enrichment analysis. Most of these DEGs in males showed a higher expression trend than females in both quantity and expression quantity. The highly expressed genes in males were relatively concentrated in neurogenesis, signal transduction, ion transport and energy metabolism, whereas the up-expressed genes in females were mainly related to the growth and development regulation of specific auditory cells. Conclusions The transcriptome of male and female O. tormota has been sequenced and de novo assembled, which will provide gene reference for further genomic studies. In addition, this is the first research to reveal the molecular mechanisms of sex differences in ultrasonic hearing between the sexes of O. tormota and will provide new insights into the genetic basis of the auditory adaptation in amphibians during their transition from water to land. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08536-2.
Collapse
Affiliation(s)
- Zhuo Chen
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China.,The Observation and Research Field Station of Taihang Mountain Forest Ecosystems of Henan Province, Xinxiang, 453007, China
| | - Yao Liu
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Rui Liang
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Chong Cui
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Yanjun Zhu
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China
| | - Fang Zhang
- College of Life Sciences, Anhui Normal University, Wuhu, 241000, China
| | - Jie Zhang
- College of Fisheries, Henan Normal University, Xinxiang, 453007, China.
| | - Xiaohong Chen
- College of Life Sciences, Henan Normal University, Xinxiang, 453007, China. .,The Observation and Research Field Station of Taihang Mountain Forest Ecosystems of Henan Province, Xinxiang, 453007, China.
| |
Collapse
|
17
|
Wei XP, Collie M, Dempsey B, Fortin G, Yackle K. A novel reticular node in the brainstem synchronizes neonatal mouse crying with breathing. Neuron 2022; 110:644-657.e6. [PMID: 34998469 PMCID: PMC8857054 DOI: 10.1016/j.neuron.2021.12.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/14/2021] [Accepted: 12/08/2021] [Indexed: 12/30/2022]
Abstract
Human speech can be divided into short, rhythmically timed elements, similar to syllables within words. Even our cries and laughs, as well as the vocalizations of other species, are periodic. However, the cellular and molecular mechanisms underlying the tempo of mammalian vocalizations remain unknown. Furthermore, even the core cells that produce vocalizations remain ill-defined. Here, we describe rhythmically timed neonatal mouse vocalizations that occur within single breaths and identify a brainstem node that is necessary for and sufficient to structure these cries, which we name the intermediate reticular oscillator (iRO). We show that the iRO acts autonomously and sends direct inputs to key muscles and the respiratory rhythm generator in order to coordinate neonatal vocalizations with breathing, as well as paces and patterns these cries. These results reveal that a novel mammalian brainstem oscillator embedded within the conserved breathing circuitry plays a central role in the production of neonatal vocalizations.
Collapse
Affiliation(s)
- Xin Paul Wei
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Matthew Collie
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Bowen Dempsey
- Institut de Biologie de l'École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Gilles Fortin
- Institut de Biologie de l'École Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, PSL Research University, Paris, France
| | - Kevin Yackle
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
18
|
Abstract
Breathing (or respiration) is a complex motor behavior that originates in the brainstem. In minimalistic terms, breathing can be divided into two phases: inspiration (uptake of oxygen, O2) and expiration (release of carbon dioxide, CO2). The neurons that discharge in synchrony with these phases are arranged in three major groups along the brainstem: (i) pontine, (ii) dorsal medullary, and (iii) ventral medullary. These groups are formed by diverse neuron types that coalesce into heterogeneous nuclei or complexes, among which the preBötzinger complex in the ventral medullary group contains cells that generate the respiratory rhythm (Chapter 1). The respiratory rhythm is not rigid, but instead highly adaptable to the physic demands of the organism. In order to generate the appropriate respiratory rhythm, the preBötzinger complex receives direct and indirect chemosensory information from other brainstem respiratory nuclei (Chapter 2) and peripheral organs (Chapter 3). Even though breathing is a hard-wired unconscious behavior, it can be temporarily altered at will by other higher-order brain structures (Chapter 6), and by emotional states (Chapter 7). In this chapter, we focus on the development of brainstem respiratory groups and highlight the cell lineages that contribute to central and peripheral chemoreflexes.
Collapse
Affiliation(s)
- Eser Göksu Isik
- Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Luis R Hernandez-Miranda
- Brainstem Group, Institute for Cell Biology and Neurobiology, Charité Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.
| |
Collapse
|
19
|
Lee H, Lee JJ, Park NY, Dubey SK, Kim T, Ruan K, Lim SB, Park SH, Ha S, Kovlyagina I, Kim KT, Kim S, Oh Y, Kim H, Kang SU, Song MR, Lloyd TE, Maragakis NJ, Hong YB, Eoh H, Lee G. Multi-omic analysis of selectively vulnerable motor neuron subtypes implicates altered lipid metabolism in ALS. Nat Neurosci 2021; 24:1673-1685. [PMID: 34782793 PMCID: PMC8639773 DOI: 10.1038/s41593-021-00944-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/16/2021] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating disorder in which motor neurons degenerate, the causes of which remain unclear. In particular, the basis for selective vulnerability of spinal motor neurons (sMNs) and resistance of ocular motor neurons to degeneration in ALS has yet to be elucidated. Here, we applied comparative multi-omics analysis of human induced pluripotent stem cell-derived sMNs and ocular motor neurons to identify shared metabolic perturbations in inherited and sporadic ALS sMNs, revealing dysregulation in lipid metabolism and its related genes. Targeted metabolomics studies confirmed such findings in sMNs of 17 ALS (SOD1, C9ORF72, TDP43 (TARDBP) and sporadic) human induced pluripotent stem cell lines, identifying elevated levels of arachidonic acid. Pharmacological reduction of arachidonic acid levels was sufficient to reverse ALS-related phenotypes in both human sMNs and in vivo in Drosophila and SOD1G93A mouse models. Collectively, these findings pinpoint a catalytic step of lipid metabolism as a potential therapeutic target for ALS.
Collapse
Affiliation(s)
- Hojae Lee
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Robert Packard Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jae Jin Lee
- Department of Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA, USA
| | - Na Young Park
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Korea
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Korea
| | - Sandeep Kumar Dubey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Taeyong Kim
- Department of Biology, San Diego State University, San Diego, CA, USA
| | - Kai Ruan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Su Bin Lim
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Korea
| | - Seong-Hyun Park
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shinwon Ha
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Irina Kovlyagina
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Kyung-Tai Kim
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
- Jeonbuk Branch Institute, Korea Institute of Toxicology, Jeongeup, Republic of Korea
| | - Seongjun Kim
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yohan Oh
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Hyesoo Kim
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sung-Ung Kang
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mi-Ryoung Song
- School of Life Sciences, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
| | - Thomas E Lloyd
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Cellular and Molecular Medicine Program, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Nicholas J Maragakis
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Young Bin Hong
- Department of Biochemistry, College of Medicine, Dong-A University, Busan, Korea.
- Department of Translational Biomedical Sciences, Graduate School of Dong-A University, Busan, Korea.
| | - Hyungjin Eoh
- Department of Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, CA, USA.
| | - Gabsang Lee
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Robert Packard Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
20
|
Dempsey B, Sungeelee S, Bokiniec P, Chettouh Z, Diem S, Autran S, Harrell ER, Poulet JFA, Birchmeier C, Carey H, Genovesio A, McMullan S, Goridis C, Fortin G, Brunet JF. A medullary centre for lapping in mice. Nat Commun 2021; 12:6307. [PMID: 34728601 PMCID: PMC8563905 DOI: 10.1038/s41467-021-26275-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/27/2021] [Indexed: 01/14/2023] Open
Abstract
It has long been known that orofacial movements for feeding can be triggered, coordinated, and often rhythmically organized at the level of the brainstem, without input from higher centers. We uncover two nuclei that can organize the movements for ingesting fluids in mice. These neuronal groups, IRtPhox2b and Peri5Atoh1, are marked by expression of the pan-autonomic homeobox gene Phox2b and are located, respectively, in the intermediate reticular formation of the medulla and around the motor nucleus of the trigeminal nerve. They are premotor to all jaw-opening and tongue muscles. Stimulation of either, in awake animals, opens the jaw, while IRtPhox2b alone also protracts the tongue. Moreover, stationary stimulation of IRtPhox2b entrains a rhythmic alternation of tongue protraction and retraction, synchronized with jaw opening and closing, that mimics lapping. Finally, fiber photometric recordings show that IRtPhox2b is active during volitional lapping. Our study identifies one of the subcortical nuclei underpinning a stereotyped feeding behavior.
Collapse
Affiliation(s)
- Bowen Dempsey
- Institut de Biologie de l'ENS (IBENS), Inserm, CNRS, École normale supérieure, PSL Research University, Paris, France
| | - Selvee Sungeelee
- Institut de Biologie de l'ENS (IBENS), Inserm, CNRS, École normale supérieure, PSL Research University, Paris, France
| | - Phillip Bokiniec
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), and Neuroscience Research Center, Charité-Universitätsmedizin, Berlin, Germany
| | - Zoubida Chettouh
- Institut de Biologie de l'ENS (IBENS), Inserm, CNRS, École normale supérieure, PSL Research University, Paris, France
| | - Séverine Diem
- Université Paris-Saclay, CNRS, Institut des Neurosciences NeuroPSI, Gif-sur-Yvette, France
| | - Sandra Autran
- Université Paris-Saclay, CNRS, Institut des Neurosciences NeuroPSI, Gif-sur-Yvette, France
| | - Evan R Harrell
- Institut Pasteur, INSERM, Institut de l'Audition, Paris, France
| | - James F A Poulet
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), and Neuroscience Research Center, Charité-Universitätsmedizin, Berlin, Germany
| | - Carmen Birchmeier
- Developmental Biology/Signal Transduction, Max Delbrueck Center for Molecular Medicine, and Cluster of Excellence NeuroCure, Neuroscience Research Center, Charité-Universitätsmedizin, Berlin, Germany
| | - Harry Carey
- Faculty of Medicine, Health & Human Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Auguste Genovesio
- Institut de Biologie de l'ENS (IBENS), Inserm, CNRS, École normale supérieure, PSL Research University, Paris, France
| | - Simon McMullan
- Faculty of Medicine, Health & Human Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Christo Goridis
- Institut de Biologie de l'ENS (IBENS), Inserm, CNRS, École normale supérieure, PSL Research University, Paris, France
| | - Gilles Fortin
- Institut de Biologie de l'ENS (IBENS), Inserm, CNRS, École normale supérieure, PSL Research University, Paris, France
| | - Jean-François Brunet
- Institut de Biologie de l'ENS (IBENS), Inserm, CNRS, École normale supérieure, PSL Research University, Paris, France.
| |
Collapse
|
21
|
Scott K, O'Rourke R, Winkler CC, Kearns CA, Appel B. Temporal single-cell transcriptomes of zebrafish spinal cord pMN progenitors reveal distinct neuronal and glial progenitor populations. Dev Biol 2021; 479:37-50. [PMID: 34303700 PMCID: PMC8410680 DOI: 10.1016/j.ydbio.2021.07.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 11/20/2022]
Abstract
Ventral spinal cord progenitor cells, which express the basic helix loop helix transcription factor Olig2, sequentially produce motor neurons and oligodendrocyte precursor cells (OPCs). Following specification some OPCs differentiate as myelinating oligodendrocytes while others persist as OPCs. Though a considerable amount of work has described the molecular profiles that define motor neurons, OPCs, and oligodendrocytes, less is known about the progenitors that produce them. To identify the developmental origins and transcriptional profiles of motor neurons and OPCs, we performed single-cell RNA sequencing on isolated pMN cells from embryonic zebrafish trunk tissue at stages that encompassed motor neurogenesis, OPC specification, and initiation of oligodendrocyte differentiation. Downstream analyses revealed two distinct pMN progenitor populations: one that appears to produce neurons and one that appears to produce OPCs. This latter population, called Pre-OPCs, is marked by expression of GS Homeobox 2 (gsx2), a gene that encodes a homeobox transcription factor. Using fluorescent in situ hybridizations, we identified gsx2-expressing Pre-OPCs in the spinal cord prior to expression of canonical OPC marker genes. Our data therefore reveal heterogeneous gene expression profiles among pMN progenitors, supporting prior fate mapping evidence.
Collapse
Affiliation(s)
- Kayt Scott
- Department of Pediatrics, Section of Developmental Biology, Colorado, 80045, USA; Cell Biology, Stem Cells and Development Training Program, Colorado, 80045, USA
| | - Rebecca O'Rourke
- Department of Pediatrics, Section of Developmental Biology, Colorado, 80045, USA
| | - Caitlin C Winkler
- Department of Pediatrics, Section of Developmental Biology, Colorado, 80045, USA; RNA Bioscience Initiative and Department of Biochemistry and Molecular Genetics, Colorado, 80045, USA
| | - Christina A Kearns
- Department of Pediatrics, Section of Developmental Biology, Colorado, 80045, USA
| | - Bruce Appel
- Department of Pediatrics, Section of Developmental Biology, Colorado, 80045, USA; Children's Hospital Colorado, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
22
|
Tyagi A, Goyal A, Chaware P, Rathinam BA. Mutations of PHOX2B Gene in Patients of Obesity Hypoventilation Syndrome in Central India. J Lab Physicians 2021; 14:164-168. [PMID: 35982870 PMCID: PMC9381314 DOI: 10.1055/s-0041-1735582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Abstract
Background Paired-like homeobox 2B (PHOX2B) gene on chromosome 4p12 codes for a transcription factor having a role in the formation of noradrenergic neuronal circuits. Its mutations have been linked to congenital central hypoventilation syndrome (CCHS). The clinical presentation of both, obesity hypoventilation syndrome (OHS) and CCHS in adults (named late-onset central hypoventilation syndrome), is quite similar. Because of this symptomatic similarity, multifactorial causation of OHS, the mutation of PHOX2B gene was studied in patients with OHS in this study.
Methods A hospital-based cross-sectional study was performed on patients diagnosed with OHS. The deoxyribonucleic acid was extracted from 2 mL of venous blood and was further amplified, specific to exon 3. The amplified products were cast and run in 2% agarose gel and then subjected to Sanger sequencing.
Results Thirty patients of OHS (21 male; 9 female) were enrolled in the present study, average age being 51.7 years. The Sanger sequencing of the samples revealed no apparent areas of deletions and no apparent mutations.
Conclusion Primers for exon 3 were used for amplification in thermocycler, as exon 3 is the most frequently mutated exon for PHOX2B gene, as per existing literature. The entire gene needs to be studied for mutations and the sample size needs to be increased.
Collapse
Affiliation(s)
- Ankita Tyagi
- Department of Anatomy, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, India
| | - Abhishek Goyal
- Department of Pulmonary Medicine, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, India
| | - Prashant Chaware
- Department of Anatomy, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, India
| | - Bertha A.D. Rathinam
- Department of Anatomy, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, India
| |
Collapse
|
23
|
Johannsen EB, Baughn LB, Sharma N, Zjacic N, Pirooznia M, Elhaik E. The Genetics of Sudden Infant Death Syndrome-Towards a Gene Reference Resource. Genes (Basel) 2021; 12:216. [PMID: 33540853 PMCID: PMC7913088 DOI: 10.3390/genes12020216] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/21/2021] [Accepted: 01/29/2021] [Indexed: 12/16/2022] Open
Abstract
Sudden infant death syndrome (SIDS) is the unexpected death of an infant under one year of age that remains unexplained after a thorough investigation. Despite SIDS remaining a diagnosis of exclusion with an unexplained etiology, it is widely accepted that SIDS can be caused by environmental and/or biological factors, with multiple underlying candidate genes. However, the lack of biomarkers raises questions as to why genetic studies on SIDS to date are unable to provide a clearer understanding of the disease etiology. We sought to improve the identification of SIDS-associated genes by reviewing the SIDS genetic literature and objectively categorizing and scoring the reported genes based on the strength of evidence (from C1 (high) to C5 (low)). This was followed by analyses of function, associations between genes, the enrichment of gene ontology (GO) terms, and pathways and gender difference in tissue gene expression. We constructed a curated database for SIDS gene candidates consisting of 109 genes, 14 of which received a category 4 (C4) and 95 genes received the lowest category of C5. That none of the genes was classified into the higher categories indicates the low level of supporting evidence. We found that genes of both scoring categories show distinct networks and are highly diverse in function and involved in many GO terms and pathways, in agreement with the perception of SIDS as a heterogeneous syndrome. Genes of both scoring categories are part of the cardiac system, muscle, and ion channels, whereas immune-related functions showed enrichment for C4 genes. A limited association was found with neural development. Overall, inconsistent reports and missing metadata contribute to the ambiguity of genetic studies. Considering those parameters could help improve the identification of at-risk SIDS genes. However, the field is still far from offering a full-pledged genetic test to identify at-risk infants and is still hampered with methodological challenges and misunderstandings of the vulnerabilities of vital biological mechanisms.
Collapse
Affiliation(s)
| | - Linda B. Baughn
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (L.B.B.); (N.S.)
| | - Neeraj Sharma
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA; (L.B.B.); (N.S.)
| | - Nicolina Zjacic
- Department of Animal and Plant Sciences, University of Sheffield, Sheffield S10 2TN, UK;
| | - Mehdi Pirooznia
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Eran Elhaik
- Department of Biology, Lund University, 22362 Lund, Sweden;
| |
Collapse
|
24
|
Dias JM, Alekseenko Z, Jeggari A, Boareto M, Vollmer J, Kozhevnikova M, Wang H, Matise MP, Alexeyenko A, Iber D, Ericson J. A Shh/Gli-driven three-node timer motif controls temporal identity and fate of neural stem cells. SCIENCE ADVANCES 2020; 6:6/38/eaba8196. [PMID: 32938678 PMCID: PMC7494341 DOI: 10.1126/sciadv.aba8196] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 07/28/2020] [Indexed: 05/03/2023]
Abstract
How time is measured by neural stem cells during temporal neurogenesis has remained unresolved. By combining experiments and computational modeling, we define a Shh/Gli-driven three-node timer underlying the sequential generation of motor neurons (MNs) and serotonergic neurons in the brainstem. The timer is founded on temporal decline of Gli-activator and Gli-repressor activities established through down-regulation of Gli transcription. The circuitry conforms an incoherent feed-forward loop, whereby Gli proteins not only promote expression of Phox2b and thereby MN-fate but also account for a delayed activation of a self-promoting transforming growth factor-β (Tgfβ) node triggering a fate switch by repressing Phox2b. Hysteresis and spatial averaging by diffusion of Tgfβ counteract noise and increase temporal accuracy at the population level, providing a functional rationale for the intrinsically programmed activation of extrinsic switch signals in temporal patterning. Our study defines how time is reliably encoded during the sequential specification of neurons.
Collapse
Affiliation(s)
- José M Dias
- Department of Cell and Molecular Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Zhanna Alekseenko
- Department of Cell and Molecular Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Ashwini Jeggari
- Department of Cell and Molecular Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Marcelo Boareto
- D-BSSE, ETF Zürich, Mattenstrasse 26, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics (SIB), Mattenstrasse 26, 4058 Basel, Switzerland
| | - Jannik Vollmer
- D-BSSE, ETF Zürich, Mattenstrasse 26, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics (SIB), Mattenstrasse 26, 4058 Basel, Switzerland
| | - Mariya Kozhevnikova
- Department of Cell and Molecular Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden
| | - Hui Wang
- Department of Neuroscience and Cell Biology, Rutgers-Robert Wood Johnson Medical School, 675 Hoes Lane, Piscataway, NJ, 08854, USA
| | - Michael P Matise
- Department of Neuroscience and Cell Biology, Rutgers-Robert Wood Johnson Medical School, 675 Hoes Lane, Piscataway, NJ, 08854, USA
| | - Andrey Alexeyenko
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Science for Life Laboratory, Box 1031, 17121, Solna, Sweden
| | - Dagmar Iber
- D-BSSE, ETF Zürich, Mattenstrasse 26, 4058 Basel, Switzerland
- Swiss Institute of Bioinformatics (SIB), Mattenstrasse 26, 4058 Basel, Switzerland
| | - Johan Ericson
- Department of Cell and Molecular Biology, Karolinska Institutet, S-171 77 Stockholm, Sweden.
| |
Collapse
|
25
|
Alzate-Correa D, Mei-Ling Liu J, Jones M, Silva TM, Alves MJ, Burke E, Zuñiga J, Kaya B, Zaza G, Aslan MT, Blackburn J, Shimada MY, Fernandes-Junior SA, Baer LA, Stanford KI, Kempton A, Smith S, Szujewski CC, Silbaugh A, Viemari JC, Takakura AC, Garcia AJ, Moreira TS, Czeisler CM, Otero JJ. Neonatal apneic phenotype in a murine congenital central hypoventilation syndrome model is induced through non-cell autonomous developmental mechanisms. Brain Pathol 2020; 31:84-102. [PMID: 32654284 PMCID: PMC7881415 DOI: 10.1111/bpa.12877] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 05/10/2020] [Accepted: 06/11/2020] [Indexed: 12/19/2022] Open
Abstract
Congenital central hypoventilation syndrome (CCHS) represents a rare genetic disorder usually caused by mutations in the homeodomain transcription factor PHOX2B. Some CCHS patients suffer mainly from deficiencies in CO2 and/or O2 respiratory chemoreflex, whereas other patients present with full apnea shortly after birth. Our goal was to identify the neuropathological mechanisms of apneic presentations in CCHS. In the developing murine neuroepithelium, Phox2b is expressed in three discrete progenitor domains across the dorsal-ventral axis, with different domains responsible for producing unique autonomic or visceral motor neurons. Restricting the expression of mutant Phox2b to the ventral visceral motor neuron domain induces marked newborn apnea together with a significant loss of visceral motor neurons, RTN ablation, and preBötzinger complex dysfunction. This finding suggests that the observed apnea develops through non-cell autonomous developmental mechanisms. Mutant Phox2b expression in dorsal rhombencephalic neurons did not generate significant respiratory dysfunction, but did result in subtle metabolic thermoregulatory deficiencies. We confirm the expression of a novel murine Phox2b splice variant which shares exons 1 and 2 with the more widely studied Phox2b splice variant, but which differs in exon 3 where most CCHS mutations occur. We also show that mutant Phox2b expression in the visceral motor neuron progenitor domain increases cell proliferation at the expense of visceral motor neuron development. We propose that visceral motor neurons may function as organizers of brainstem respiratory neuron development, and that disruptions in their development result in secondary/non-cell autonomous maldevelopment of key brainstem respiratory neurons.
Collapse
Affiliation(s)
- Diego Alzate-Correa
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Jillian Mei-Ling Liu
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Mikayla Jones
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Talita M Silva
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Michele Joana Alves
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Elizabeth Burke
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Jessica Zuñiga
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Behiye Kaya
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Giuliana Zaza
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Mehmet Tahir Aslan
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Jessica Blackburn
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Marina Y Shimada
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Silvio A Fernandes-Junior
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Lisa A Baer
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Kristin I Stanford
- Department of Physiology and Cell Biology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Amber Kempton
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Sakima Smith
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Caroline C Szujewski
- Institute for Integrative Physiology, Grossman Institute for Neuroscience Quantitative Biology and Human Behavior, The Committee on Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Abby Silbaugh
- Institute for Integrative Physiology, Grossman Institute for Neuroscience Quantitative Biology and Human Behavior, The Committee on Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Jean-Charles Viemari
- P3M Team, Institut de Neurosciences de la Timone, UMR 7289 AMU-CNRS, Marseille, France
| | - Ana C Takakura
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Alfredo J Garcia
- Institute for Integrative Physiology, Grossman Institute for Neuroscience Quantitative Biology and Human Behavior, The Committee on Neurobiology, The University of Chicago, Chicago, IL, USA
| | - Thiago S Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
| | - Catherine M Czeisler
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| | - José J Otero
- Division of Neuropathology, Department of Pathology, The Ohio State University College of Medicine, Columbus, OH, USA
| |
Collapse
|
26
|
van der Heijden ME, Zoghbi HY. Development of the brainstem respiratory circuit. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2019; 9:e366. [PMID: 31816185 DOI: 10.1002/wdev.366] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 02/01/2023]
Abstract
The respiratory circuit is comprised of over a dozen functionally and anatomically segregated brainstem nuclei that work together to control respiratory rhythms. These respiratory rhythms emerge prenatally but only acquire vital importance at birth, which is the first time the respiratory circuit faces the sole responsibility for O2 /CO2 homeostasis. Hence, the respiratory circuit has little room for trial-and-error-dependent fine tuning and relies on a detailed genetic blueprint for development. This blueprint is provided by transcription factors that have specific spatiotemporal expression patterns along the rostral-caudal or dorsal-ventral axis of the developing brainstem, in proliferating precursor cells and postmitotic neurons. Studying these transcription factors in mice has provided key insights into the functional segregation of respiratory control and the vital importance of specific respiratory nuclei. Many studies converge on just two respiratory nuclei that each have rhythmogenic properties during the prenatal period: the preBötzinger complex (preBötC) and retrotrapezoid nucleus/parafacial nucleus (RTN/pF). Here, we discuss the transcriptional regulation that guides the development of these nuclei. We also summarize evidence showing that normal preBötC development is necessary for neonatal survival, and that neither the preBötC nor the RTN/pF alone is sufficient to sustain normal postnatal respiratory rhythms. Last, we highlight several studies that use intersectional genetics to assess the necessity of transcription factors only in subregions of their expression domain. These studies independently demonstrate that lack of RTN/pF neurons weakens the respiratory circuit, yet these neurons are not necessary for neonatal survival because developmentally related populations can compensate for abnormal RTN/pF function at birth. This article is categorized under: Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Meike E van der Heijden
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas
| | - Huda Y Zoghbi
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, Texas.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Department of Pediatrics, Baylor College of Medicine, Houston, Texas.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
27
|
Cummings KJ, Hodges MR. The serotonergic system and the control of breathing during development. Respir Physiol Neurobiol 2019; 270:103255. [PMID: 31362064 DOI: 10.1016/j.resp.2019.103255] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/28/2019] [Accepted: 07/12/2019] [Indexed: 01/26/2023]
Abstract
Serotonin (5-hydroxytryptamine 5-HT) was first discovered in the late 1940's as an endogenous bioactive amine capable of inducing vasoconstriction, and in the mid-1950's was found in the brain. It was in these early years that some of the first demonstrations were made regarding a role for brain 5-HT in neurological function and behavior, including data implicating reduced brain levels of 5-HT in clinical depression. Since that time, advances in molecular biology and physiological approaches in basic science research have intensely focused on 5-HT in the brain, and the many facets of its role during embryonic development, post-natal maturation, and neural function in adulthood continues to be established. This review focuses on what is known about the developmental roles for the 5-HT system, which we define as the neurons producing 5-HT along with pre-and post-synaptic receptors, in a vital homeostatic motor behavior - the control of breathing. We will cover what is known about the embryonic origins and fate specification of 5-HT neurons, and how the 5-HT system influences pre- and post-natal maturation of the ventilatory control system. In addition, we will focus on the role of the 5-HT system in specific respiratory behaviors during fetal, neonatal and postnatal development, and the relevance of dysfunction in this system in respiratory-related human pathologies including Sudden Infant Death Syndrome (SIDS).
Collapse
Affiliation(s)
- Kevin J Cummings
- Department of Biomedical Sciences, University of Missouri, Columbia, MO 65211, USA; Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA.
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
28
|
Czeisler CM, Silva TM, Fair SR, Liu J, Tupal S, Kaya B, Cowgill A, Mahajan S, Silva PE, Wang Y, Blissett AR, Göksel M, Borniger JC, Zhang N, Fernandes‐Junior SA, Catacutan F, Alves MJ, Nelson RJ, Sundaresean V, Rekling J, Takakura AC, Moreira TS, Otero JJ. The role of PHOX2B-derived astrocytes in chemosensory control of breathing and sleep homeostasis. J Physiol 2019; 597:2225-2251. [PMID: 30707772 PMCID: PMC6462490 DOI: 10.1113/jp277082] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/28/2019] [Indexed: 01/07/2023] Open
Abstract
KEY POINTS The embryonic PHOX2B-progenitor domain generates neuronal and glial cells which together are involved in chemosensory control of breathing and sleep homeostasis. Ablating PHOX2B-derived astrocytes significantly contributes to secondary hypoxic respiratory depression as well as abnormalities in sleep homeostasis. PHOX2B-derived astrocyte ablation results in axonal pathologies in the retrotrapezoid nucleus. ABSTRACT We identify in mice a population of ∼800 retrotrapezoid nucleus (RTN) astrocytes derived from PHOX2B-positive, OLIG3-negative progenitor cells, that interact with PHOX2B-expressing RTN chemosensory neurons. PHOX2B-derived astrocyte ablation during early life results in adult-onset O2 chemoreflex deficiency. These animals also display changes in sleep homeostasis, including fragmented sleep and disturbances in delta power after sleep deprivation, all without observable changes in anxiety or social behaviours. Ultrastructural evaluation of the RTN demonstrates that PHOX2B-derived astrocyte ablation results in features characteristic of degenerative neuro-axonal dystrophy, including abnormally dilated axon terminals and increased amounts of synapses containing autophagic vacuoles/phagosomes. We conclude that PHOX2B-derived astrocytes are necessary for maintaining a functional O2 chemosensory reflex in the adult, modulate sleep homeostasis, and are key regulators of synaptic integrity in the RTN region, which is necessary for the chemosensory control of breathing. These data also highlight how defects in embryonic development may manifest as neurodegenerative pathology in an adult.
Collapse
Affiliation(s)
| | - Talita M. Silva
- Department of Physiology and BiophysicsInstitute of Biomedical ScienceUniversity of Sao PauloSao PauloBrazil
| | - Summer R. Fair
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Jillian Liu
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Srinivasan Tupal
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Behiye Kaya
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Aaron Cowgill
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Salil Mahajan
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Phelipe E. Silva
- Department of Physiology and BiophysicsInstitute of Biomedical ScienceUniversity of Sao PauloSao PauloBrazil
| | - Yangyang Wang
- Department of NeuroscienceThe Ohio State University College of MedicineColumbusOHUSA
- The Ohio State University Mathematical Biosciences InstituteColumbusOHUSA
| | - Angela R. Blissett
- Department of Mechanical and Aerospace EngineeringThe Ohio State University College of EngineeringColumbusOHUSA
| | - Mustafa Göksel
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Jeremy C. Borniger
- Department of NeuroscienceThe Ohio State University College of MedicineColumbusOHUSA
| | - Ning Zhang
- Department of NeuroscienceWest Virginia UniversityWVUSA
| | - Silvio A. Fernandes‐Junior
- The Ohio State University Campus Microscopy and Imaging FacilityColumbusOHUSA
- Department of PharmacologyInstitute of Biomedical ScienceUniversity of São PauloSao PauloBrazil
| | - Fay Catacutan
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Michele J. Alves
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | | | - Vishnu Sundaresean
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| | - Jens Rekling
- Department of NeuroscienceUniversity of CopenhagenCopenhagenDenmark
| | - Ana C. Takakura
- Department of PharmacologyInstitute of Biomedical ScienceUniversity of São PauloSao PauloBrazil
| | - Thiago S. Moreira
- Department of Physiology and BiophysicsInstitute of Biomedical ScienceUniversity of Sao PauloSao PauloBrazil
| | - José J. Otero
- Department of PathologyThe Ohio State University College of MedicineColumbusOHUSA
| |
Collapse
|
29
|
Gruner HN, Kim M, Mastick GS. Robo1 and 2 Repellent Receptors Cooperate to Guide Facial Neuron Cell Migration and Axon Projections in the Embryonic Mouse Hindbrain. Neuroscience 2019; 402:116-129. [PMID: 30685539 PMCID: PMC6435285 DOI: 10.1016/j.neuroscience.2019.01.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 01/12/2019] [Accepted: 01/14/2019] [Indexed: 11/19/2022]
Abstract
The facial nerve is necessary for our ability to eat, speak, and make facial expressions. Both the axons and cell bodies of the facial nerve undergo a complex embryonic developmental pattern involving migration of the cell bodies caudally and tangentially through rhombomeres, and simultaneously the axons projecting to exit the hindbrain to form the facial nerve. Our goal in this study was to test the functions of the chemorepulsive receptors Robo1 and Robo2 in facial neuron migration and axon projection by analyzing genetically marked motor neurons in double-mutant mouse embryos through the migration time course, E10.0-E13.5. In Robo1/2 double mutants, axon projection and cell body migration errors were more severe than in single mutants. Most axons did not make it to their motor exit point, and instead projected into and longitudinally within the floor plate. Surprisingly, some facial neurons had multiple axons exiting and projecting into the floor plate. At the same time, a subset of mutant facial cell bodies failed to migrate caudally, and instead either streamed dorsally toward the exit point or shifted into the floor plate. We conclude that Robo1 and Robo2 have redundant functions to guide multiple aspects of the complex cell migration of the facial nucleus, as well as regulating axon trajectories and suppressing formation of ectopic axons.
Collapse
Affiliation(s)
- Hannah N. Gruner
- Department of Biology, University of Nevada, 1664 N Virginia St, Reno, NV 89557, USA.
| | - Minkyung Kim
- Department of Biology, University of Nevada, 1664 N Virginia St, Reno, NV 89557, USA.
| | - Grant S. Mastick
- Department of Biology, University of Nevada, 1664 N Virginia St, Reno, NV 89557, USA.
| |
Collapse
|
30
|
Frank MM, Goodrich LV. Talking back: Development of the olivocochlear efferent system. WILEY INTERDISCIPLINARY REVIEWS. DEVELOPMENTAL BIOLOGY 2018; 7:e324. [PMID: 29944783 PMCID: PMC6185769 DOI: 10.1002/wdev.324] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/27/2018] [Accepted: 05/17/2018] [Indexed: 02/02/2023]
Abstract
Developing sensory systems must coordinate the growth of neural circuitry spanning from receptors in the peripheral nervous system (PNS) to multilayered networks within the central nervous system (CNS). This breadth presents particular challenges, as nascent processes must navigate across the CNS-PNS boundary and coalesce into a tightly intermingled wiring pattern, thereby enabling reliable integration from the PNS to the CNS and back. In the auditory system, feedforward spiral ganglion neurons (SGNs) from the periphery collect sound information via tonotopically organized connections in the cochlea and transmit this information to the brainstem for processing via the VIII cranial nerve. In turn, feedback olivocochlear neurons (OCNs) housed in the auditory brainstem send projections into the periphery, also through the VIII nerve. OCNs are motor neuron-like efferent cells that influence auditory processing within the cochlea and protect against noise damage in adult animals. These aligned feedforward and feedback systems develop in parallel, with SGN central axons reaching the developing auditory brainstem around the same time that the OCN axons extend out toward the developing inner ear. Recent findings have begun to unravel the genetic and molecular mechanisms that guide OCN development, from their origins in a generic pool of motor neuron precursors to their specialized roles as modulators of cochlear activity. One recurrent theme is the importance of efferent-afferent interactions, as afferent SGNs guide OCNs to their final locations within the sensory epithelium, and efferent OCNs shape the activity of the developing auditory system. This article is categorized under: Nervous System Development > Vertebrates: Regional Development.
Collapse
|
31
|
Ernsberger U, Rohrer H. Sympathetic tales: subdivisons of the autonomic nervous system and the impact of developmental studies. Neural Dev 2018; 13:20. [PMID: 30213267 PMCID: PMC6137933 DOI: 10.1186/s13064-018-0117-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/12/2018] [Indexed: 02/06/2023] Open
Abstract
Remarkable progress in a range of biomedical disciplines has promoted the understanding of the cellular components of the autonomic nervous system and their differentiation during development to a critical level. Characterization of the gene expression fingerprints of individual neurons and identification of the key regulators of autonomic neuron differentiation enables us to comprehend the development of different sets of autonomic neurons. Their individual functional properties emerge as a consequence of differential gene expression initiated by the action of specific developmental regulators. In this review, we delineate the anatomical and physiological observations that led to the subdivision into sympathetic and parasympathetic domains and analyze how the recent molecular insights melt into and challenge the classical description of the autonomic nervous system.
Collapse
Affiliation(s)
- Uwe Ernsberger
- Institute for Clinical Neuroanatomy, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany
| | - Hermann Rohrer
- Institute for Clinical Neuroanatomy, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany
| |
Collapse
|
32
|
Cardani S, Di Lascio S, Belperio D, Di Biase E, Ceccherini I, Benfante R, Fornasari D. Desogestrel down-regulates PHOX2B and its target genes in progesterone responsive neuroblastoma cells. Exp Cell Res 2018; 370:671-679. [PMID: 30036539 DOI: 10.1016/j.yexcr.2018.07.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/18/2018] [Accepted: 07/19/2018] [Indexed: 10/28/2022]
Abstract
The paired-like homeobox 2B gene (PHOX2B) encodes a key transcription factor that plays a role in the development of the autonomic nervous system and the neural structures involved in controlling breathing. In humans, PHOX2B over-expression plays a role in the pathogenesis of tumours arising from the sympathetic nervous system such as neuroblastomas, and heterozygous PHOX2B mutations cause Congenital Central Hypoventilation Syndrome (CCHS), a life-threatening neurocristopathy characterised by the defective autonomic control of breathing and involving altered CO2/H+ chemosensitivity. The recovery of CO2/H+ chemosensitivity and increased ventilation have been observed in two CCHS patients using the potent contraceptive progestin desogestrel. Given the central role of PHOX2B in the pathogenesis of CCHS, and the progesterone-mediated effects observed in the disease, we generated progesterone-responsive neuroblastoma cells, and evaluated the effects of 3-Ketodesogestrel (3-KDG), the biologically active metabolite of desogestrel, on the expression of PHOX2B and its target genes. Our findings demonstrate that, through progesterone nuclear receptor PR-B, 3-KDG down-regulates PHOX2B gene expression, by a post-transcriptional mechanism, and its target genes and open up the possibility that this mechanism may contribute to the positive effects observed in some CCHS patients.
Collapse
Affiliation(s)
- Silvia Cardani
- Dept. of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, via Vanvitelli 32, 2019 Milan, Italy
| | - Simona Di Lascio
- Dept. of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, via Vanvitelli 32, 2019 Milan, Italy
| | - Debora Belperio
- Dept. of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, via Vanvitelli 32, 2019 Milan, Italy
| | - Erika Di Biase
- Dept. of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, via Vanvitelli 32, 2019 Milan, Italy
| | - Isabella Ceccherini
- Laboratorio di Genetica Molecolare, Istituto Giannina Gaslini, Largo G. Gaslini 5, 16148 Genoa, Italy
| | - Roberta Benfante
- Dept. of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, via Vanvitelli 32, 2019 Milan, Italy; CNR -Neuroscience Institute, via Vanvitelli 32, 20129 Milan, Italy.
| | - Diego Fornasari
- Dept. of Medical Biotechnology and Translational Medicine (BIOMETRA), Università degli Studi di Milano, via Vanvitelli 32, 2019 Milan, Italy; CNR -Neuroscience Institute, via Vanvitelli 32, 20129 Milan, Italy.
| |
Collapse
|
33
|
Puelles L, Tvrdik P, Martínez-de-la-torre M. The Postmigratory Alar Topography of Visceral Cranial Nerve Efferents Challenges the Classical Model of Hindbrain Columns. Anat Rec (Hoboken) 2018; 302:485-504. [DOI: 10.1002/ar.23830] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Luis Puelles
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, School of Medicine; University of Murcia; Murcia 30071 Spain
| | - Petr Tvrdik
- Department of Neurosurgery-Physiology; University of Utah; Salt Lake City, Utah 84112
| | - Margaret Martínez-de-la-torre
- Department of Human Anatomy and Psychobiology and IMIB-Arrixaca Institute, School of Medicine; University of Murcia; Murcia 30071 Spain
| |
Collapse
|
34
|
Watson C, Tvrdik P. Spinal Accessory Motor Neurons in the Mouse: A Special Type of Branchial Motor Neuron? Anat Rec (Hoboken) 2018; 302:505-511. [DOI: 10.1002/ar.23822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 10/10/2017] [Accepted: 10/10/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Charles Watson
- School of Animal Biology; University of Western Australia; Perth Australia
- Neuroscience Research Australia; Sydney Australia
| | - Petr Tvrdik
- Department of Neurosurgery; University of Utah; Salt Lake City Utah
| |
Collapse
|
35
|
Molecular specification of facial branchial motor neurons in vertebrates. Dev Biol 2018; 436:5-13. [PMID: 29391164 DOI: 10.1016/j.ydbio.2018.01.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 01/27/2018] [Accepted: 01/28/2018] [Indexed: 02/02/2023]
Abstract
Orofacial muscles are critical for life-sustaining behaviors, such as feeding and breathing. Centuries of work by neuroanatomists and surgeons resulted in the mapping of bulbar motor neurons in the brainstem and the course of the cranial nerves that carry their axons. Despite the sophisticated understanding of the anatomy of the region, the molecular mechanisms that dictate the development and maturation of facial motor neurons remain poorly understood. This fundamental problem has been recently revisited by physiologists with novel techniques of studying the rhythmic contraction of orofacial muscles in relationship to breathing. The molecular understanding of facial motor neuron development will not only lead to the comprehension of the neural basis of facial expression but may also unlock new avenues to generate stem cell-derived replacements. This review summarizes the current understanding of molecular programs involved in facial motor neuron generation, migration, and maturation, including neural circuit assembly.
Collapse
|
36
|
Dominici C, Rappeneau Q, Zelina P, Fouquet S, Chédotal A. Non-cell autonomous control of precerebellar neuron migration by Slit and Robo proteins. Development 2018; 145:dev150375. [PMID: 29343636 DOI: 10.1242/dev.150375] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 12/11/2017] [Indexed: 02/05/2023]
Abstract
During development, precerebellar neurons migrate tangentially from the dorsal hindbrain to the floor plate. Their axons cross it but their cell bodies stop their ventral migration upon reaching the midline. It has previously been shown that Slit chemorepellents and their receptors, Robo1 and Robo2, might control the migration of precerebellar neurons in a repulsive manner. Here, we have used a conditional knockout strategy in mice to test this hypothesis. We show that the targeted inactivation of the expression of Robo1 and Robo2 receptors in precerebellar neurons does not perturb their migration and that they still stop at the midline. The selective ablation of the expression of all three Slit proteins in floor-plate cells has no effect on pontine neurons and only induces the migration of a small subset of inferior olivary neurons across the floor plate. Likewise, we show that the expression of Slit proteins in the facial nucleus is dispensable for pontine neuron migration. Together, these results show that Robo1 and Robo2 receptors act non-cell autonomously in migrating precerebellar neurons and that floor-plate signals, other than Slit proteins, must exist to prevent midline crossing.
Collapse
Affiliation(s)
- Chloé Dominici
- Sorbonne Universités, UPMC Paris 06, INSERM, CNRS, Institut de la Vision 75012, Paris, France
| | - Quentin Rappeneau
- Sorbonne Universités, UPMC Paris 06, INSERM, CNRS, Institut de la Vision 75012, Paris, France
| | - Pavol Zelina
- Sorbonne Universités, UPMC Paris 06, INSERM, CNRS, Institut de la Vision 75012, Paris, France
| | - Stéphane Fouquet
- Sorbonne Universités, UPMC Paris 06, INSERM, CNRS, Institut de la Vision 75012, Paris, France
| | - Alain Chédotal
- Sorbonne Universités, UPMC Paris 06, INSERM, CNRS, Institut de la Vision 75012, Paris, France
| |
Collapse
|
37
|
Di Lascio S, Benfante R, Di Zanni E, Cardani S, Adamo A, Fornasari D, Ceccherini I, Bachetti T. Structural and functional differences in PHOX2B frameshift mutations underlie isolated or syndromic congenital central hypoventilation syndrome. Hum Mutat 2017; 39:219-236. [PMID: 29098737 PMCID: PMC5846889 DOI: 10.1002/humu.23365] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/24/2017] [Accepted: 10/29/2017] [Indexed: 12/13/2022]
Abstract
Heterozygous mutations in the PHOX2B gene are causative of congenital central hypoventilation syndrome (CCHS), a neurocristopathy characterized by defective autonomic control of breathing due to the impaired differentiation of neural crest cells. Among PHOX2B mutations, polyalanine (polyAla) expansions are almost exclusively associated with isolated CCHS, whereas frameshift variants, although less frequent, are often more severe than polyAla expansions and identified in syndromic CCHS. This article provides a complete review of all the frameshift mutations identified in cases of isolated and syndromic CCHS reported in the literature as well as those identified by us and not yet published. These were considered in terms of both their structure, whether the underlying indels induced frameshifts of either 1 or 2 steps ("frame 2" and "frame 3" mutations respectively), and clinical associations. Furthermore, we evaluated the structural and functional effects of one "frame 3" mutation identified in a patient with isolated CCHS, and one "frame 2" mutation identified in a patient with syndromic CCHS, also affected with Hirschsprung's disease and neuroblastoma. The data thus obtained confirm that the type of translational frame affects the severity of the transcriptional dysfunction and the predisposition to isolated or syndromic CCHS.
Collapse
Affiliation(s)
- Simona Di Lascio
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Roberta Benfante
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy.,CNR- Neuroscience Institute, Milan, Italy
| | | | - Silvia Cardani
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Annalisa Adamo
- UOC Genetica Medica, Istituto Giannina Gaslini, Genoa, Italy
| | - Diego Fornasari
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy.,CNR- Neuroscience Institute, Milan, Italy
| | | | | |
Collapse
|
38
|
Espinosa-Medina I, Saha O, Boismoreau F, Chettouh Z, Rossi F, Richardson WD, Brunet JF. The sacral autonomic outflow is sympathetic. Science 2017; 354:893-897. [PMID: 27856909 DOI: 10.1126/science.aah5454] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 10/14/2016] [Indexed: 12/16/2022]
Abstract
A kinship between cranial and pelvic visceral nerves of vertebrates has been accepted for a century. Accordingly, sacral preganglionic neurons are considered parasympathetic, as are their targets in the pelvic ganglia that prominently control rectal, bladder, and genital functions. Here, we uncover 15 phenotypic and ontogenetic features that distinguish pre- and postganglionic neurons of the cranial parasympathetic outflow from those of the thoracolumbar sympathetic outflow in mice. By every single one, the sacral outflow is indistinguishable from the thoracolumbar outflow. Thus, the parasympathetic nervous system receives input from cranial nerves exclusively and the sympathetic nervous system from spinal nerves, thoracic to sacral inclusively. This simplified, bipartite architecture offers a new framework to understand pelvic neurophysiology as well as development and evolution of the autonomic nervous system.
Collapse
Affiliation(s)
- I Espinosa-Medina
- Institut de Biologie de l'École Normale Supérieure (IBENS), INSERM, CNRS, École Normale Supérieure, Paris Sciences et Lettres Research University, Paris, 75005 France
| | - O Saha
- Institut de Biologie de l'École Normale Supérieure (IBENS), INSERM, CNRS, École Normale Supérieure, Paris Sciences et Lettres Research University, Paris, 75005 France
| | - F Boismoreau
- Institut de Biologie de l'École Normale Supérieure (IBENS), INSERM, CNRS, École Normale Supérieure, Paris Sciences et Lettres Research University, Paris, 75005 France
| | - Z Chettouh
- Institut de Biologie de l'École Normale Supérieure (IBENS), INSERM, CNRS, École Normale Supérieure, Paris Sciences et Lettres Research University, Paris, 75005 France
| | - F Rossi
- Institut de Biologie de l'École Normale Supérieure (IBENS), INSERM, CNRS, École Normale Supérieure, Paris Sciences et Lettres Research University, Paris, 75005 France
| | - W D Richardson
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - J-F Brunet
- Institut de Biologie de l'École Normale Supérieure (IBENS), INSERM, CNRS, École Normale Supérieure, Paris Sciences et Lettres Research University, Paris, 75005 France.
| |
Collapse
|
39
|
Di Bonito M, Studer M. Cellular and Molecular Underpinnings of Neuronal Assembly in the Central Auditory System during Mouse Development. Front Neural Circuits 2017; 11:18. [PMID: 28469562 PMCID: PMC5395578 DOI: 10.3389/fncir.2017.00018] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/01/2017] [Indexed: 11/13/2022] Open
Abstract
During development, the organization of the auditory system into distinct functional subcircuits depends on the spatially and temporally ordered sequence of neuronal specification, differentiation, migration and connectivity. Regional patterning along the antero-posterior axis and neuronal subtype specification along the dorso-ventral axis intersect to determine proper neuronal fate and assembly of rhombomere-specific auditory subcircuits. By taking advantage of the increasing number of transgenic mouse lines, recent studies have expanded the knowledge of developmental mechanisms involved in the formation and refinement of the auditory system. Here, we summarize several findings dealing with the molecular and cellular mechanisms that underlie the assembly of central auditory subcircuits during mouse development, focusing primarily on the rhombomeric and dorso-ventral origin of auditory nuclei and their associated molecular genetic pathways.
Collapse
|
40
|
Cain JT, Kim DI, Quast M, Shivega WG, Patrick RJ, Moser C, Reuter S, Perez M, Myers A, Weimer JM, Roux KJ, Landsverk M. Nonsense pathogenic variants in exon 1 of PHOX2B lead to translational reinitiation in congenital central hypoventilation syndrome. Am J Med Genet A 2017; 173:1200-1207. [PMID: 28371199 DOI: 10.1002/ajmg.a.38162] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 11/15/2016] [Accepted: 01/09/2017] [Indexed: 11/06/2022]
Abstract
Pathogenic variants in PHOX2B lead to congenital central hypoventilation syndrome (CCHS), a rare disorder of the nervous system characterized by autonomic dysregulation and hypoventilation typically presenting in the neonatal period, although a milder late-onset (LO) presentation has been reported. More than 90% of cases are caused by polyalanine repeat mutations (PARMs) in the C-terminus of the protein; however non-polyalanine repeat mutations (NPARMs) have been reported. Most NPARMs are located in exon 3 of PHOX2B and result in a more severe clinical presentation including Hirschsprung disease (HSCR) and/or peripheral neuroblastic tumors (PNTs). A previously reported nonsense pathogenic variant in exon 1 of a patient with LO-CCHS and no HSCR or PNTs leads to translational reinitiation at a downstream AUG codon producing an N-terminally truncated protein. Here we report additional individuals with nonsense pathogenic variants in exon 1 of PHOX2B. In vitro analyses were used to determine if these and other reported nonsense variants in PHOX2B exon 1 produced N-terminally truncated proteins. We found that all tested nonsense variants in PHOX2B exon 1 produced a truncated protein of the same size. This truncated protein localized to the nucleus and transactivated a target promoter. These data suggest that nonsense pathogenic variants in the first exon of PHOX2B likely escape nonsense mediated decay (NMD) and produce N-terminally truncated proteins functionally distinct from those produced by the more common PARMs.
Collapse
Affiliation(s)
- Jacob T Cain
- Children's Health Research Center, Sanford Research, Sioux Falls, South Dakota
| | - Dae I Kim
- Children's Health Research Center, Sanford Research, Sioux Falls, South Dakota
| | - Megan Quast
- Children's Health Research Center, Sanford Research, Sioux Falls, South Dakota
| | - Winnie G Shivega
- Children's Health Research Center, Sanford Research, Sioux Falls, South Dakota
| | - Ryan J Patrick
- Children's Health Research Center, Sanford Research, Sioux Falls, South Dakota
| | - Chuanpit Moser
- Section of Pediatric Pulmonology, Sanford School of Medicine at the University of South Dakota, Sioux Falls, South Dakota
| | - Suzanne Reuter
- Section of Neonatal-Perinatal Medicine, Sanford School of Medicine at the University of South Dakota, Sioux Falls, South Dakota
| | - Myrza Perez
- Department of Pediatric Pulmonology, Kaiser Permanente, Roseville, California
| | - Angela Myers
- Department of Pediatrics, Sanford School of Medicine at the University of South Dakota, Sioux Falls, South Dakota
| | - Jill M Weimer
- Children's Health Research Center, Sanford Research, Sioux Falls, South Dakota.,Department of Pediatrics, Sanford School of Medicine at the University of South Dakota, Sioux Falls, South Dakota
| | - Kyle J Roux
- Children's Health Research Center, Sanford Research, Sioux Falls, South Dakota.,Department of Pediatrics, Sanford School of Medicine at the University of South Dakota, Sioux Falls, South Dakota
| | - Megan Landsverk
- Children's Health Research Center, Sanford Research, Sioux Falls, South Dakota.,Department of Pediatrics, Sanford School of Medicine at the University of South Dakota, Sioux Falls, South Dakota
| |
Collapse
|
41
|
ISL1-based LIM complexes control Slit2 transcription in developing cranial motor neurons. Sci Rep 2016; 6:36491. [PMID: 27819291 PMCID: PMC5098159 DOI: 10.1038/srep36491] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/17/2016] [Indexed: 01/02/2023] Open
Abstract
LIM-homeodomain (HD) transcription factors form a multimeric complex and assign neuronal subtype identities, as demonstrated by the hexameric ISL1-LHX3 complex which gives rise to somatic motor (SM) neurons. However, the roles of combinatorial LIM code in motor neuron diversification and their subsequent differentiation is much less well understood. In the present study, we demonstrate that the ISL1 controls postmitotic cranial branchiomotor (BM) neurons including the positioning of the cell bodies and peripheral axon pathfinding. Unlike SM neurons, which transform into interneurons, BM neurons are normal in number and in marker expression in Isl1 mutant mice. Nevertheless, the movement of trigeminal and facial BM somata is stalled, and their peripheral axons are fewer or misrouted, with ectopic branches. Among genes whose expression level changes in previous ChIP-seq and microarray analyses in Isl1-deficient cell lines, we found that Slit2 transcript was almost absent from BM neurons of Isl1 mutants. Both ISL1-LHX3 and ISL1-LHX4 bound to the Slit2 enhancer and drove endogenous Slit2 expression in SM and BM neurons. Our findings suggest that combinations of ISL1 and LHX factors establish cell-type specificity and functional diversity in terms of motor neuron identities and/or axon development.
Collapse
|
42
|
Hernandez-Miranda LR, Müller T, Birchmeier C. The dorsal spinal cord and hindbrain: From developmental mechanisms to functional circuits. Dev Biol 2016; 432:34-42. [PMID: 27742210 DOI: 10.1016/j.ydbio.2016.10.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 09/07/2016] [Accepted: 10/10/2016] [Indexed: 11/29/2022]
Abstract
Neurons of the dorsal hindbrain and spinal cord are central in receiving, processing and relaying sensory perception and participate in the coordination of sensory-motor output. Numerous cellular and molecular mechanisms that underlie neuronal development in both regions of the nervous system are shared. We discuss here the mechanisms that generate neuronal diversity in the dorsal spinal cord and hindbrain, and emphasize similarities in patterning and neuronal specification. Insight into the developmental mechanisms has provided tools that can help to assign functions to small subpopulations of neurons. Hence, novel information on how mechanosensory or pain sensation is encoded under normal and neuropathic conditions has already emerged. Such studies show that the complex neuronal circuits that control perception of somatosensory and viscerosensory stimuli are becoming amenable to investigations.
Collapse
Affiliation(s)
- Luis R Hernandez-Miranda
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz-Association, Robert-Rössle-Str. 10, 13125 Berlin, Germany.
| | - Thomas Müller
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz-Association, Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Carmen Birchmeier
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz-Association, Robert-Rössle-Str. 10, 13125 Berlin, Germany.
| |
Collapse
|
43
|
Linares AJ, Lin CH, Damianov A, Adams KL, Novitch BG, Black DL. The splicing regulator PTBP1 controls the activity of the transcription factor Pbx1 during neuronal differentiation. eLife 2015; 4:e09268. [PMID: 26705333 PMCID: PMC4755740 DOI: 10.7554/elife.09268] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Accepted: 12/22/2015] [Indexed: 12/13/2022] Open
Abstract
The RNA-binding proteins PTBP1 and PTBP2 control programs of alternative splicing during neuronal development. PTBP2 was found to maintain embryonic splicing patterns of many synaptic and cytoskeletal proteins during differentiation of neuronal progenitor cells (NPCs) into early neurons. However, the role of the earlier PTBP1 program in embryonic stem cells (ESCs) and NPCs was not clear. We show that PTBP1 controls a program of neuronal gene expression that includes the transcription factor Pbx1. We identify exons specifically regulated by PTBP1 and not PTBP2 as mouse ESCs differentiate into NPCs. We find that PTBP1 represses Pbx1 exon 7 and the expression of the neuronal Pbx1a isoform in ESCs. Using CRISPR-Cas9 to delete regulatory elements for exon 7, we induce Pbx1a expression in ESCs, finding that this activates transcription of neuronal genes. Thus, PTBP1 controls the activity of Pbx1 to suppress its neuronal transcriptional program prior to induction of NPC development. DOI:http://dx.doi.org/10.7554/eLife.09268.001 The neurons that transmit information around the nervous system develop in several stages. Embryonic stem cells specialize to form neuronal progenitor cells, which then develop into neurons. These cell types have different characteristics, in part because they make different proteins or different versions of the same proteins. To make a protein, the DNA sequence of a gene is used to build a molecule of ribonucleic acid (RNA) that acts as a template for the protein. However, not all of this sequence codes for the protein. The non-coding regions must be removed from the RNA, and the remaining “exons” joined together to form the final “mRNA” template. Not all of the exons are necessarily included in the final mRNA molecule. By joining together different combinations of exons, several different versions of a protein can be produced from a single gene. This process is known as alternative splicing. One way that alternative splicing is controlled is through proteins that bind to RNA and determine which exons are included or excluded from the final mRNA molecule. PTBP1 is an RNA-binding protein that controls alternative splicing in embryonic stem cells and neuronal progenitor cells. Embryonic stem cells have the ability to develop into all the cells of the body. In contrast, neuronal progenitor cells are restricted in their development and only give rise to specialized cells of the nervous system. The role of PTBP1 in these properties was not clear. Linares et al. have now used a range of techniques to study the RNA molecules produced in these two cell types and how these RNAs change when PTBP1 is removed. This identified many RNAs whose splicing is regulated by PTBP1, including mRNAs of the gene that produces a protein called Pbx1, which is an important regulator of neuronal development. Further investigation revealed that PTBP1 prevents a particular exon being included in the mRNA template for Pbx1. This creates an embryonic stem cell form of Pbx1 that does not affect neuronal genes. Removal of PTBP1 allows splicing of the Pbx1 exon and produces a version of Pbx1 that is found in neuronal progenitor cells and which turns on neuronal genes. Thus, through its action on Pbx1, one role of PTBP1 is to enable stem cells to maintain their non-neuronal properties and prevent their premature development into neuronal progenitor cells. The gene for Pbx1 is only one of many genes controlled by PTBP1 at the level of splicing. One challenge for the future will be to understand how these genes work together in a common program that determines the properties of stem cells. Another question regards how the different Pbx1 proteins in stem cells and in neuronal progenitors can exert different effects in the cells where they are made. DOI:http://dx.doi.org/10.7554/eLife.09268.002
Collapse
Affiliation(s)
- Anthony J Linares
- Molecular Biology Institute Graduate Program, University of California, Los Angeles, Los Angeles, United States
| | - Chia-Ho Lin
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, United States
| | - Andrey Damianov
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, United States
| | - Katrina L Adams
- Molecular Biology Institute Graduate Program, University of California, Los Angeles, Los Angeles, United States.,Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Bennett G Novitch
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| | - Douglas L Black
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, United States.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, United States
| |
Collapse
|
44
|
Kim N, Park C, Jeong Y, Song MR. Functional Diversification of Motor Neuron-specific Isl1 Enhancers during Evolution. PLoS Genet 2015; 11:e1005560. [PMID: 26447474 PMCID: PMC4598079 DOI: 10.1371/journal.pgen.1005560] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 09/09/2015] [Indexed: 11/19/2022] Open
Abstract
Functional diversification of motor neurons has occurred in order to selectively control the movements of different body parts including head, trunk and limbs. Here we report that transcription of Isl1, a major gene necessary for motor neuron identity, is controlled by two enhancers, CREST1 (E1) and CREST2 (E2) that allow selective gene expression of Isl1 in motor neurons. Introduction of GFP reporters into the chick neural tube revealed that E1 is active in hindbrain motor neurons and spinal cord motor neurons, whereas E2 is active in the lateral motor column (LMC) of the spinal cord, which controls the limb muscles. Genome-wide ChIP-Seq analysis combined with reporter assays showed that Phox2 and the Isl1-Lhx3 complex bind to E1 and drive hindbrain and spinal cord-specific expression of Isl1, respectively. Interestingly, Lhx3 alone was sufficient to activate E1, and this may contribute to the initiation of Isl1 expression when progenitors have just developed into motor neurons. E2 was induced by onecut 1 (OC-1) factor that permits Isl1 expression in LMCm neurons. Interestingly, the core region of E1 has been conserved in evolution, even in the lamprey, a jawless vertebrate with primitive motor neurons. All E1 sequences from lamprey to mouse responded equally well to Phox2a and the Isl1-Lhx3 complex. Conversely, E2, the enhancer for limb-innervating motor neurons, was only found in tetrapod animals. This suggests that evolutionarily-conserved enhancers permit the diversification of motor neurons. During evolution, motor neurons became specialized to control movements of different body parts including head, trunk and limbs. Here we report that two enhancers of Isl1, E1 and E2, are active together with transcription factors in motor neurons. Surprisingly, E1 and its response to transcription factors has been conserved in evolution from the lamprey to man, whereas E2 is only found in animals with limbs. Our study provides an evolutionary example of how functional diversification of motor neurons is achieved by a dynamic interplay between enhancers and transcription factors.
Collapse
Affiliation(s)
- Namhee Kim
- School of Life Sciences, Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, Republic of Korea
| | - Chungoo Park
- School of Biological Sciences and Technology, Chonnam National University, Yongbong-ro, Buk-gu, Gwangju, Republic of Korea
| | - Yongsu Jeong
- Department of Genetic Engineering, College of Life Sciences and Graduate School of Biotechnology, Kyung Hee University, Yongin-si, Republic of Korea
| | - Mi-Ryoung Song
- School of Life Sciences, Cell Dynamics Research Center, Gwangju Institute of Science and Technology, Oryong-dong, Buk-gu, Gwangju, Republic of Korea
- * E-mail:
| |
Collapse
|
45
|
Sajgo S, Ali S, Popescu O, Badea TC. Dynamic expression of transcription factor Brn3b during mouse cranial nerve development. J Comp Neurol 2015; 524:1033-61. [PMID: 26356988 DOI: 10.1002/cne.23890] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 08/18/2015] [Accepted: 08/31/2015] [Indexed: 01/23/2023]
Abstract
During development, transcription factor combinatorial codes define a large variety of morphologically and physiologically distinct neurons. Such a combinatorial code has been proposed for the differentiation of projection neurons of the somatic and visceral components of cranial nerves. It is possible that individual neuronal cell types are not specified by unique transcription factors but rather emerge through the intersection of their expression domains. Brn3a, Brn3b, and Brn3c, in combination with each other and/or transcription factors of other families, can define subgroups of retinal ganglion cells (RGC), spiral and vestibular ganglia, inner ear and vestibular hair cell neurons in the vestibuloacoustic system, and groups of somatosensory neurons in the dorsal root ganglia. The present study investigates the expression and potential role of the Brn3b transcription factor in cranial nerves and associated nuclei of the brainstem. We report the dynamic expression of Brn3b in the somatosensory component of cranial nerves II, V, VII, and VIII and visceromotor nuclei of nerves VII, IX, and X as well as other brainstem nuclei during different stages of development into adult stage. We find that genetically identified Brn3b(KO) RGC axons show correct but delayed pathfinding during the early stages of embryonic development. However, loss of Brn3b does not affect the anatomy of the other cranial nerves normally expressing this transcription factor.
Collapse
Affiliation(s)
- Szilard Sajgo
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, 20892.,Molecular Biology Center, Interdisciplinary Research Institute on Bio-Nano-Science, Babes-Bolyai University, Cluj-Napoca, Cluj, 400084, Romania
| | - Seid Ali
- National Eye Institute, National Institutes of Health, Bethesda, Maryland, 20892
| | - Octavian Popescu
- Molecular Biology Center, Interdisciplinary Research Institute on Bio-Nano-Science, Babes-Bolyai University, Cluj-Napoca, Cluj, 400084, Romania.,Institute of Biology, Romanian Academy, Bucharest, 060031, Romania
| | | |
Collapse
|
46
|
Hozumi A, Horie T, Sasakura Y. Neuronal map reveals the highly regionalized pattern of the juvenile central nervous system of the ascidian Ciona intestinalis. Dev Dyn 2015; 244:1375-93. [PMID: 26250096 DOI: 10.1002/dvdy.24317] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 07/28/2015] [Accepted: 08/02/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The dorsally located central nervous system (CNS) is an important hallmark of chordates. Among chordates, tunicate ascidians change their CNS remarkably by means of a metamorphosis from a highly regionalized larval CNS to an oval-shaped juvenile CNS without prominent morphological features. The neuronal organization of the CNS of ascidian tadpole larvae has been well described, but that in the CNS of postmetamorphosis juveniles has not been characterized well. RESULTS We investigated the number of neural cells, the number and position of differentiated neurons, and their axonal trajectories in the juvenile CNS of the ascidian Ciona intestinalis. The cell bodies of cholinergic, glutamatergic, and GABAergic/glycinergic neurons exhibited different localization patterns along the anterior-posterior axis in the juvenile CNS. Cholinergic neurons extended their axons toward the oral, atrial and body wall muscles and pharyngeal gill to regulate muscle contraction and ciliary movement. CONCLUSIONS Unlike its featureless shape, the juvenile CNS is highly patterned along the anterior-posterior axis. This patterning may be necessary for exerting multiple roles in the regulation of adult tissues distributed throughout the body. This basic information of the juvenile CNS of Ciona will allow in-depth studies of molecular mechanisms underlying the reconstruction of the CNS during ascidian metamorphosis.
Collapse
Affiliation(s)
- Akiko Hozumi
- Shimoda Marine Research Center, University of Tsukuba, Shimoda, Shizuoka, Japan
| | - Takeo Horie
- Japan Science and Technology Agency, PRESTO, Honcho, Kawaguchi, Saitama, Japan
| | - Yasunori Sasakura
- Shimoda Marine Research Center, University of Tsukuba, Shimoda, Shizuoka, Japan
| |
Collapse
|
47
|
Nobuta H, Cilio MR, Danhaive O, Tsai HH, Tupal S, Chang SM, Murnen A, Kreitzer F, Bravo V, Czeisler C, Gokozan HN, Gygli P, Bush S, Weese-Mayer DE, Conklin B, Yee SP, Huang EJ, Gray PA, Rowitch D, Otero JJ. Dysregulation of locus coeruleus development in congenital central hypoventilation syndrome. Acta Neuropathol 2015; 130:171-83. [PMID: 25975378 PMCID: PMC4503865 DOI: 10.1007/s00401-015-1441-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 05/01/2015] [Accepted: 05/02/2015] [Indexed: 12/29/2022]
Abstract
Human congenital central hypoventilation syndrome (CCHS), resulting from mutations in transcription factor PHOX2B, manifests with impaired responses to hypoxemia and hypercapnia especially during sleep. To identify brainstem structures developmentally affected in CCHS, we analyzed two postmortem neonatal-lethal cases with confirmed polyalanine repeat expansion (PARM) or Non-PARM (PHOX2B∆8) mutation of PHOX2B. Both human cases showed neuronal losses within the locus coeruleus (LC), which is important for central noradrenergic signaling. Using a conditionally active transgenic mouse model of the PHOX2B∆8 mutation, we found that early embryonic expression (<E10.5) caused failure of LC neuronal specification and perinatal respiratory lethality. In contrast, later onset (E11.5) of PHOX2B∆8 expression was not deleterious to LC development and perinatal respiratory lethality was rescued, despite failure of chemosensor retrotrapezoid nucleus formation. Our findings indicate that early-onset mutant PHOX2B expression inhibits LC neuronal development in CCHS. They further suggest that such mutations result in dysregulation of central noradrenergic signaling, and therefore, potential for early pharmacologic intervention in humans with CCHS.
Collapse
|
48
|
Rosin JM, Kurrasch DM, Cobb J. Shox2 is required for the proper development of the facial motor nucleus and the establishment of the facial nerves. BMC Neurosci 2015; 16:39. [PMID: 26156498 PMCID: PMC4495855 DOI: 10.1186/s12868-015-0176-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/12/2015] [Indexed: 11/10/2022] Open
Abstract
Background Axons from the visceral motor neurons (vMNs) project from nuclei in the hindbrain to innervate autonomic ganglia and branchial arch-derived muscles. Although much is known about the events that govern specification of somatic motor neurons, the genetic pathways responsible for the development of vMNs are less well characterized. We know that vMNs, like all motor neurons, depend on sonic hedgehog signaling for their generation. Similarly, the paired-like homeobox 2b (Phox2b) gene, which is expressed in both proliferating progenitors and post-mitotic motor neurons, is essential for the development of vMNs. Given that our previous study identified a novel role for the short stature homeobox 2 (Shox2) gene in the hindbrain, and since SHOX2 has been shown to regulate transcription of islet 1 (Isl1), an important regulator of vMN development, we sought to determine whether Shox2 is required for the proper development of the facial motor nucleus. Results Using a Nestin-Cre driver, we show that elimination of Shox2 throughout the brain results in elevated cell death in the facial motor nucleus at embryonic day 12.5 (E12.5) and E14.5, which correlates with impaired axonal projection properties of vMNs. We also observed changes in the spatial expression of the vMN cell fate factors Isl1 and Phox2b, and concomitant defects in Shh and Ptch1 expression in Shox2 mutants. Furthermore, we demonstrate that elimination of Shox2 results in the loss of dorsomedial and ventromedial subnuclei by postnatal day 0 (P0), which may explain the changes in physical activity and impaired feeding/nursing behavior in Shox2 mutants. Conclusions Combined, our data show that Shox2 is required for development of the facial motor nucleus and its associated facial (VII) nerves, and serves as a new molecular tool to probe the genetic programs of this understudied hindbrain region. Electronic supplementary material The online version of this article (doi:10.1186/s12868-015-0176-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jessica M Rosin
- Department of Biological Sciences, University of Calgary, 2500 University Drive N.W., BI286D, Calgary, AB, T2N 1N4, Canada.
| | - Deborah M Kurrasch
- Department of Medical Genetics, Alberta Children's Hospital Research Institute, University of Calgary, 3330 Hospital Drive N.W., Room HS2275, Calgary, AB, T2N 4N1, Canada.
| | - John Cobb
- Department of Biological Sciences, University of Calgary, 2500 University Drive N.W., BI286D, Calgary, AB, T2N 1N4, Canada.
| |
Collapse
|
49
|
Jarrar W, Dias JM, Ericson J, Arnold HH, Holz A. Nkx2.2 and Nkx2.9 are the key regulators to determine cell fate of branchial and visceral motor neurons in caudal hindbrain. PLoS One 2015; 10:e0124408. [PMID: 25919494 PMCID: PMC4412715 DOI: 10.1371/journal.pone.0124408] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 03/05/2015] [Indexed: 12/12/2022] Open
Abstract
Cranial motor nerves in vertebrates are comprised of the three principal subtypes of branchial, visceral, and somatic motor neurons, which develop in typical patterns along the anteroposterior and dorsoventral axes of hindbrain. Here we demonstrate that the formation of branchial and visceral motor neurons critically depends on the transcription factors Nkx2.2 and Nkx2.9, which together determine the cell fate of neuronal progenitor cells. Disruption of both genes in mouse embryos results in complete loss of the vagal and spinal accessory motor nerves, and partial loss of the facial and glossopharyngeal motor nerves, while the purely somatic hypoglossal and abducens motor nerves are not diminished. Cell lineage analysis in a genetically marked mouse line reveals that alterations of cranial nerves in Nkx2.2; Nkx2.9 double-deficient mouse embryos result from changes of cell fate in neuronal progenitor cells. As a consequence progenitors of branchiovisceral motor neurons in the ventral p3 domain of hindbrain are transformed to somatic motor neurons, which use ventral exit points to send axon trajectories to their targets. Cell fate transformation is limited to the caudal hindbrain, as the trigeminal nerve is not affected in double-mutant embryos suggesting that Nkx2.2 and Nkx2.9 proteins play no role in the development of branchiovisceral motor neurons in hindbrain rostral to rhombomere 4.
Collapse
Affiliation(s)
- Wassan Jarrar
- Cell and Molecular Biology, Zoological Institute, University of Braunschweig, Braunschweig, Germany
| | - Jose M. Dias
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Johan Ericson
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Hans-Henning Arnold
- Cell and Molecular Biology, Zoological Institute, University of Braunschweig, Braunschweig, Germany
- * E-mail: (AH); (HHA)
| | - Andreas Holz
- Cell and Molecular Biology, Zoological Institute, University of Braunschweig, Braunschweig, Germany
- * E-mail: (AH); (HHA)
| |
Collapse
|
50
|
Deterministic HOX patterning in human pluripotent stem cell-derived neuroectoderm. Stem Cell Reports 2015; 4:632-44. [PMID: 25843047 PMCID: PMC4400649 DOI: 10.1016/j.stemcr.2015.02.018] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 11/25/2022] Open
Abstract
Colinear HOX expression during hindbrain and spinal cord development diversifies and assigns regional neural phenotypes to discrete rhombomeric and vertebral domains. Despite the precision of HOX patterning in vivo, in vitro approaches for differentiating human pluripotent stem cells (hPSCs) to posterior neural fates coarsely pattern HOX expression thereby generating cultures broadly specified to hindbrain or spinal cord regions. Here, we demonstrate that successive activation of fibroblast growth factor, Wnt/β-catenin, and growth differentiation factor signaling during hPSC differentiation generates stable, homogenous SOX2+/Brachyury+ neuromesoderm that exhibits progressive, full colinear HOX activation over 7 days. Switching to retinoic acid treatment at any point during this process halts colinear HOX activation and transitions the neuromesoderm into SOX2+/PAX6+ neuroectoderm with predictable, discrete HOX gene/protein profiles that can be further differentiated into region-specific cells, e.g., motor neurons. This fully defined approach significantly expands capabilities to derive regional neural phenotypes from diverse hindbrain and spinal cord domains. Deterministic HOX expression in hPSC-derived neuromesoderm progenitors (NMPs) Wnt/β-catenin, FGF, and GDF signaling regulate HOX activation in NMPs Retinoic acid (RA) transitions NMPs to neuroectoderm and halts HOX activation Neural cells can be patterned to any rostrocaudal hindbrain or spinal cord domain
Collapse
|