1
|
Veltman CHJ, van der Ven LTM, Menegola E, Luijten M. A Pragmatic Workflow for Human Relevance Assessment of Toxicological Pathways and Associated New Approach Methodologies. Regul Toxicol Pharmacol 2025:105828. [PMID: 40228575 DOI: 10.1016/j.yrtph.2025.105828] [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: 12/04/2024] [Revised: 03/21/2025] [Accepted: 04/08/2025] [Indexed: 04/16/2025]
Abstract
Currently, safety assessments of chemical substances are predominantly based on animal data. Multiple considerations call for the use of alternative testing strategies that are based on new approach methodologies (NAMs). However, the human relevance of these testing strategies is usually uncertain. This necessitates a harmonized and accepted workflow for assessing their applicability for regulatory purposes. This report proposes such a workflow, applicable for assessing the human relevance of a toxicological pathway and the relevance of NAMs related to the different components of the pathway. The workflow starts with an established toxicological pathway, of which the adverse outcome is relevant for human health risk assessment and that has sufficient weight of evidence. Human relevance is assessed through three main questions, related to the different components (steps) of the pathway, the pathology of human syndromes that have a similar adverse outcome, and quantitative aspects. The latter comprise both interspecies differences and in vitro - in vivo differences. The combined evidence is scored as 'strong', 'moderate' or 'weak' support of human relevance, based on expert judgement. The workflow developed was tested in a case study, through application to an AOP describing craniofacial malformations after in utero exposure to triazoles. Based on evidence collected for two of the three main questions, the case study provided moderate to strong support for human relevance of both the various components of the AOP and its associated NAMs. Furthermore, it demonstrated that the workflow is a promising approach that allows for a more transparent scientific evaluation of human relevance of toxicological pathways and associated NAMs. Therefore, despite some areas for improvement, we consider the workflow an important step forward for application of AOPs and related NAMs in human health risk assessment.
Collapse
Affiliation(s)
- Christina H J Veltman
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Leo T M van der Ven
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Elena Menegola
- Department of Environmental Science and Policy, Università degli Studi di Milano, Milano, Italy
| | - Mirjam Luijten
- Centre for Health Protection, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.
| |
Collapse
|
2
|
Pang W, Zhu J, Yang K, Zhu X, Zhou W, Jiang L, Zhuang X, Liu Y, Wei J, Lu X, Yin Y, Chen Z, Xiang Y. Generation of human region-specific brain organoids with medullary spinal trigeminal nuclei. Cell Stem Cell 2024; 31:1501-1512.e8. [PMID: 39208804 DOI: 10.1016/j.stem.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/16/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Brain organoids with nucleus-specific identities provide unique platforms for studying human brain development and diseases at a finer resolution. Despite its essential role in vital body functions, the medulla of the hindbrain has seen a lack of in vitro models, let alone models resembling specific medullary nuclei, including the crucial spinal trigeminal nucleus (SpV) that relays peripheral sensory signals to the thalamus. Here, we report a method to differentiate human pluripotent stem cells into region-specific brain organoids resembling the dorsal domain of the medullary hindbrain. Importantly, organoids specifically recapitulated the development of the SpV derived from the dorsal medulla. We also developed an organoid system to create the trigeminothalamic projections between the SpV and the thalamus by fusing these organoids, namely human medullary SpV-like organoids (hmSpVOs), with organoids representing the thalamus (hThOs). Our study provides a platform for understanding SpV development, nucleus-based circuit organization, and related disorders in the human brain.
Collapse
Affiliation(s)
- Wei Pang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jinkui Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kexin Yang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaona Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Wei Zhou
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Linlin Jiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xuran Zhuang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yantong Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jianfeng Wei
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xiaoxiang Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yao Yin
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ziling Chen
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yangfei Xiang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China; Shanghai Clinical Research and Trial Center, Shanghai 201210, China.
| |
Collapse
|
3
|
Yuan X, Chen L, Saffen D. Allele-Specific Regulation of the Candidate Autism Liability Gene RAI1 by the Enhancer Variant rs4925102 ( C/G). Genes (Basel) 2024; 15:460. [PMID: 38674394 PMCID: PMC11049881 DOI: 10.3390/genes15040460] [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: 03/15/2024] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
Retinoic acid-induced 1 (RAI1) is a dosage-sensitive gene that causes autistic phenotypes when deleted or duplicated. Observations from clinical cases and animal models also suggest that changes of RAI1 expression levels contribute to autism. Previously, we used a bioinformatic approach to identify several single nucleotide polymorphisms (SNPs) located within the 5'-region of RAI1 that correlate with RAI1 mRNA expression in the human brain. In particular, the SNP rs4925102 was identified as a candidate cis-acting regulatory variant, the genotype of which may affect the binding of transcription factors that influence RAI1 mRNA expression. In this study, we provide experimental evidence based on reporter gene, chromatin immunoprecipitation (ChIP), and chromatin conformation capture (3C) assays that rs4925102 regulates RAI1 mRNA expression in an allele-specific manner in human cell lines, including the neuroblastoma-derived cell line SH-SY5Y. We also describe a statistically significant association between rs4925102 genotype and autism spectrum disorder (ASD) diagnosis in a case-control study and near-statistically significant association in an Autism Genome Project (AGP) transmission disequilibrium (TDT) study using Caucasian subjects.
Collapse
Affiliation(s)
- Xi Yuan
- Institutes of Brain Science, Fudan University, Shanghai 200032, China;
| | - Li Chen
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - David Saffen
- Institutes of Brain Science, Fudan University, Shanghai 200032, China;
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China
| |
Collapse
|
4
|
Fritzsch B. Evolution and development of extraocular motor neurons, nerves and muscles in vertebrates. Ann Anat 2024; 253:152225. [PMID: 38346566 PMCID: PMC11786961 DOI: 10.1016/j.aanat.2024.152225] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/16/2024] [Accepted: 02/05/2024] [Indexed: 02/17/2024]
Abstract
The purpose of this review is to analyze the origin of ocular motor neurons, define the pattern of innervation of nerve fibers that project to the extraocular eye muscles (EOMs), describe congenital disorders that alter the development of ocular motor neurons, and provide an overview of vestibular pathway inputs to ocular motor nuclei. Six eye muscles are innervated by axons of three ocular motor neurons, the oculomotor (CNIII), trochlear (CNIV), and abducens (CNVI) neurons. Ocular motor neurons (CNIII) originate in the midbrain and innervate the ipsilateral orbit, except for the superior rectus and the levator palpebrae, which are contralaterally innervated. Trochlear motor neurons (CNIV) originate at the midbrain-hindbrain junction and innervate the contralateral superior oblique muscle. Abducens motor neurons (CNVI) originate variously in the hindbrain of rhombomeres r4-6 that innervate the posterior (or lateral) rectus muscle and innervate the retractor bulbi. Genes allow a distinction between special somatic (CNIII, IV) and somatic (CNVI) ocular motor neurons. Development of ocular motor neurons and their axonal projections to the EOMs may be derailed by various genetic causes, resulting in the congenital cranial dysinnervation disorders. The ocular motor neurons innervate EOMs while the vestibular nuclei connect with the midbrain-brainstem motor neurons.
Collapse
Affiliation(s)
- Bernd Fritzsch
- Department of Neurological Sciences, University of Nebraska Medical Center, NE, USA.
| |
Collapse
|
5
|
Pun M, Pratt D, Nano PR, Joshi PK, Jiang L, Englinger B, Rao A, Cieslik M, Chinnaiyan AM, Aldape K, Pfister S, Filbin MG, Bhaduri A, Venneti S. Common molecular features of H3K27M DMGs and PFA ependymomas map to hindbrain developmental pathways. Acta Neuropathol Commun 2023; 11:25. [PMID: 36759899 PMCID: PMC9912509 DOI: 10.1186/s40478-023-01514-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 01/11/2023] [Indexed: 02/11/2023] Open
Abstract
Globally decreased histone 3, lysine 27 tri-methylation (H3K27me3) is a hallmark of H3K27-altered diffuse midline gliomas (DMGs) and group-A posterior fossa ependymomas (PFAs). H3K27-altered DMGs are largely characterized by lysine-to-methionine mutations in histone 3 at position 27 (H3K27M). Most PFAs overexpress EZH inhibitory protein (EZHIP), which possesses a region of similarity to the mutant H3K27M. Both H3K27M and EZHIP inhibit the function of the polycomb repressive complex 2 (PRC2) responsible for H3K27me3 deposition. These tumors often arise in neighboring regions of the brainstem and posterior fossa. In rare cases PFAs harbor H3K27M mutations, and DMGs overexpress EZHIP. These findings together raise the possibility that certain cell populations in the developing hindbrain/posterior fossa are especially sensitive to modulation of H3K27me3 states. We identified shared molecular features by comparing genomic, bulk transcriptomic, chromatin-based profiles, and single-cell RNA-sequencing (scRNA-seq) data from the two tumor classes. Our approach demonstrated that 1q gain, a key biomarker in PFAs, is prognostic in H3.1K27M, but not H3.3K27M gliomas. Conversely, Activin A Receptor Type 1 (ACVR1), which is associated with mutations in H3.1K27M gliomas, is overexpressed in a subset of PFAs with poor outcome. Despite diffuse H3K27me3 reduction, previous work shows that both tumors maintain genomic H3K27me3 deposition at select sites. We demonstrate heterogeneity in shared patterns of residual H3K27me3 for both tumors that largely segregated with inferred anatomic tumor origins and progenitor populations of tumor cells. In contrast, analysis of genes linked to H3K27 acetylation (H3K27ac)-marked enhancers showed higher expression in astrocytic-like tumor cells. Finally, common H3K27me3-marked genes mapped closely to expression patterns in the human developing hindbrain. Overall, our data demonstrate developmentally relevant molecular similarities between PFAs and H3K27M DMGs and support the overall hypothesis that deregulated mechanisms of hindbrain development are central to the biology of both tumors.
Collapse
Affiliation(s)
- Matthew Pun
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan, 3520E MSRB 1, 1150 W. Medical Center, Ann Arbor, MI, 41804, USA
- Chad Carr Pediatric Tumor Center, Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Drew Pratt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Dr., Room 2S235, Bethesda, MD, 20892, USA
| | - Patricia R Nano
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Piyush K Joshi
- Hopp Children's Cancer Center (KiTZ) Heidelberg, Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Li Jiang
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02115, USA
| | - Bernhard Englinger
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090, Vienna, Austria
- Center for Cancer Research and Comprehensive Cancer Center, Medical University Vienna, 1090, Vienna, Austria
| | - Arvind Rao
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Radiation Oncology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Marcin Cieslik
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Arul M Chinnaiyan
- Michigan Center for Translational Pathology, Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Department of Urology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
- Howard Hughes Medical Institute, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Dr., Room 2S235, Bethesda, MD, 20892, USA
| | - Stefan Pfister
- Hopp Children's Cancer Center (KiTZ) Heidelberg, Division of Pediatric Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), 69120, Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Mariella G Filbin
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA, 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Aparna Bhaduri
- Department of Biological Chemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Sriram Venneti
- Laboratory of Brain Tumor Metabolism and Epigenetics, Department of Pathology, University of Michigan, 3520E MSRB 1, 1150 W. Medical Center, Ann Arbor, MI, 41804, USA.
- Chad Carr Pediatric Tumor Center, Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA.
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
6
|
Glover JC, Fritzsch B. Molecular mechanisms governing development of the hindbrain choroid plexus and auditory projection: A validation of the seminal observations of Wilhelm His. IBRO Neurosci Rep 2022; 13:306-313. [PMID: 36247525 PMCID: PMC9561746 DOI: 10.1016/j.ibneur.2022.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 09/29/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022] Open
Abstract
Studies by His from 1868 to 1904 delineated the critical role of the dorsal roof plate in the development of the hindbrain choroid plexus, and of the rhombic lips in the development of hindbrain auditory centers. Modern molecular studies have confirmed these observations and placed them in a mechanistic context. Expression of the transcription factor Lmx1a/b is crucial to the development of the hindbrain choroid plexus, and also regulates the expression of Atoh1, a transcription factor that is essential for the formation of the cochlear hair cells and auditory nuclei. By contrast, development of the vestibular hair cells, vestibular ganglion and vestibular nuclei does not depend on Lmx1a/b. These findings demonstrate a common dependence on a specific gene for the hindbrain choroid plexus and the primary auditory projection from hair cells to sensory neurons to hindbrain nuclei. Thus, His' conclusions regarding the origins of specific hindbrain structures are borne out by molecular genetic experiments conducted more than a hundred years later.
Collapse
Affiliation(s)
- Joel C. Glover
- Department of Molecular Medicine, University of Oslo, Oslo, Norway
- Norwegian Center for Stem Cell Research, Oslo University Hospital, Oslo, Norway
- Sars International Centre for Marine Molecular Biology, University of Bergen, Bergen, Norway
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa, IA 52242, USA
| |
Collapse
|
7
|
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
|
8
|
Fritzsch B, Elliott KL, Yamoah EN. Neurosensory development of the four brainstem-projecting sensory systems and their integration in the telencephalon. Front Neural Circuits 2022; 16:913480. [PMID: 36213204 PMCID: PMC9539932 DOI: 10.3389/fncir.2022.913480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/23/2022] [Indexed: 11/18/2022] Open
Abstract
Somatosensory, taste, vestibular, and auditory information is first processed in the brainstem. From the brainstem, the respective information is relayed to specific regions within the cortex, where these inputs are further processed and integrated with other sensory systems to provide a comprehensive sensory experience. We provide the organization, genetics, and various neuronal connections of four sensory systems: trigeminal, taste, vestibular, and auditory systems. The development of trigeminal fibers is comparable to many sensory systems, for they project mostly contralaterally from the brainstem or spinal cord to the telencephalon. Taste bud information is primarily projected ipsilaterally through the thalamus to reach the insula. The vestibular fibers develop bilateral connections that eventually reach multiple areas of the cortex to provide a complex map. The auditory fibers project in a tonotopic contour to the auditory cortex. The spatial and tonotopic organization of trigeminal and auditory neuron projections are distinct from the taste and vestibular systems. The individual sensory projections within the cortex provide multi-sensory integration in the telencephalon that depends on context-dependent tertiary connections to integrate other cortical sensory systems across the four modalities.
Collapse
Affiliation(s)
- Bernd Fritzsch
- Department of Biology, The University of Iowa, Iowa City, IA, United States
- Department of Otolaryngology, The University of Iowa, Iowa City, IA, United States
- *Correspondence: Bernd Fritzsch,
| | - Karen L. Elliott
- Department of Biology, The University of Iowa, Iowa City, IA, United States
| | - Ebenezer N. Yamoah
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada, Reno, Reno, NV, United States
| |
Collapse
|
9
|
Kiani K, Sanford EM, Goyal Y, Raj A. Changes in chromatin accessibility are not concordant with transcriptional changes for single-factor perturbations. Mol Syst Biol 2022; 18:e10979. [PMID: 36069349 PMCID: PMC9450098 DOI: 10.15252/msb.202210979] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 08/11/2022] [Accepted: 08/19/2022] [Indexed: 11/23/2022] Open
Abstract
A major goal in the field of transcriptional regulation is the mapping of changes in the binding of transcription factors to the resultant changes in gene expression. Recently, methods for measuring chromatin accessibility have enabled us to measure changes in accessibility across the genome, which are thought to correspond to transcription factor-binding events. In concert with RNA-sequencing, these data in principle enable such mappings; however, few studies have looked at their concordance over short-duration treatments with specific perturbations. Here, we used tandem, bulk ATAC-seq, and RNA-seq measurements from MCF-7 breast carcinoma cells to systematically evaluate the concordance between changes in accessibility and changes in expression in response to retinoic acid and TGF-β. We found two classes of genes whose expression showed a significant change: those that showed some changes in the accessibility of nearby chromatin, and those that showed virtually no change despite strong changes in expression. The peaks associated with genes in the former group had lower baseline accessibility prior to exposure to signal. Focusing the analysis specifically on peaks with motifs for transcription factors associated with retinoic acid and TGF-β signaling did not reduce the lack of correspondence. Analysis of paired chromatin accessibility and gene expression data from distinct paths along the hematopoietic differentiation trajectory showed a much stronger correspondence, suggesting that the multifactorial biological processes associated with differentiation may lead to changes in chromatin accessibility that reflect rather than driving altered transcriptional status. Together, these results show many gene expression changes can happen independently of changes in the accessibility of local chromatin in the context of a single-factor perturbation.
Collapse
Affiliation(s)
- Karun Kiani
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Eric M Sanford
- Genomics and Computational Biology Graduate Group, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Yogesh Goyal
- Department of Bioengineering, School of Engineering and Applied SciencesUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Cell and Developmental Biology, Feinberg School of MedicineNorthwestern UniversityChicagoIllinoisUSA
- Center for Synthetic BiologyNorthwestern UniversityChicagoIllinoisUSA
| | - Arjun Raj
- Department of Bioengineering, School of Engineering and Applied SciencesUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Genetics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
10
|
Direct Reprograming of Mouse Fibroblasts into Dermal Papilla Cells via Small Molecules. Int J Mol Sci 2022; 23:ijms23084213. [PMID: 35457029 PMCID: PMC9030401 DOI: 10.3390/ijms23084213] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/29/2022] [Accepted: 04/07/2022] [Indexed: 01/27/2023] Open
Abstract
The reprogramming of somatic fibroblasts into alternative cell linages could provide a promising source of cells for regenerative medicine and cell therapy. However, the direct conversion of fibroblasts into other functional cell types is still challenging. In this study, we show that dermal-papilla-cell-like cells (DPC-LCs) can be generated by treating fibroblasts, including L929 mouse fibroblast cell lines and somatic mouse fibroblasts, with small molecules. Based on alkaline phosphatase activity and other molecular markers, different compounds or their combinations are needed for converting the two different fibroblasts into DPC-LCs. Notably, we found that TTNPB alone can efficiently convert primary adult mouse fibroblasts into DPC-LCs. DPC-LCs generated from mouse fibroblasts showed a stronger hair-inducing capacity. Transcriptome analysis reveals that expression of genes associated with a hair-inducing capacity are increased in DPC-LCs. This pharmacological approach to generating functional dermal papilla cells may have many important implications for hair follicle regeneration and hair loss therapy.
Collapse
|
11
|
Cho K, Lee SM, Heo J, Kwon YM, Chung D, Yu WJ, Bae SS, Choi G, Lee DS, Kim Y. Retinaldehyde Dehydrogenase Inhibition-Related Adverse Outcome Pathway: Potential Risk of Retinoic Acid Synthesis Inhibition during Embryogenesis. Toxins (Basel) 2021; 13:toxins13110739. [PMID: 34822523 PMCID: PMC8623920 DOI: 10.3390/toxins13110739] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 11/16/2022] Open
Abstract
Retinoic acid (RA) is one of the factors crucial for cell growth, differentiation, and embryogenesis; it interacts with the retinoic acid receptor and retinoic acid X receptor to eventually regulate target gene expression in chordates. RA is transformed from retinaldehyde via oxidization by retinaldehyde dehydrogenase (RALDH), which belongs to the family of oxidoreductases. Several chemicals, including disulphiram, diethylaminobenzaldehyde, and SB-210661, can effectively inhibit RALDH activity, potentially causing reproductive and developmental toxicity. The modes of action can be sequentially explained based on the molecular initiating event toward key events, and finally the adverse outcomes. Adverse outcome pathway (AOP) is a conceptual and theoretical framework that describes the sequential chain of casually liked events at different biological levels from molecular events to adverse effects. In the present review, we discussed a recently registered AOP (AOP297; inhibition of retinaldehyde dehydrogenase leads to population decline) to explain and support the weight of evidence for RALDH inhibition-related developmental toxicity using the existing knowledge.
Collapse
Affiliation(s)
- Kichul Cho
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Korea; (K.C.); (Y.M.K.); (D.C.); (W.-J.Y.); (S.S.B.); (G.C.)
| | - Sang-Moo Lee
- Department of Applied Bioscience, Dong-A University, Busan 49315, Korea;
| | - Jina Heo
- Department of Growth Engine Research, Chungbuk Research Institute (CRI), Chungju 28517, Korea;
| | - Yong Min Kwon
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Korea; (K.C.); (Y.M.K.); (D.C.); (W.-J.Y.); (S.S.B.); (G.C.)
| | - Dawoon Chung
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Korea; (K.C.); (Y.M.K.); (D.C.); (W.-J.Y.); (S.S.B.); (G.C.)
| | - Woon-Jong Yu
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Korea; (K.C.); (Y.M.K.); (D.C.); (W.-J.Y.); (S.S.B.); (G.C.)
| | - Seung Seob Bae
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Korea; (K.C.); (Y.M.K.); (D.C.); (W.-J.Y.); (S.S.B.); (G.C.)
| | - Grace Choi
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Korea; (K.C.); (Y.M.K.); (D.C.); (W.-J.Y.); (S.S.B.); (G.C.)
| | - Dae-Sung Lee
- Department of Genetic Resources Research, National Marine Biodiversity Institute of Korea (MABIK), Seocheon 33662, Korea; (K.C.); (Y.M.K.); (D.C.); (W.-J.Y.); (S.S.B.); (G.C.)
- Correspondence: (D.-S.L.); (Y.K.)
| | - Youngjun Kim
- Environmental Safety Group, Korea Institute of Science and Technology (KIST) Europe, Campus E 7.1, 66123 Saarbrücken, Germany
- Correspondence: (D.-S.L.); (Y.K.)
| |
Collapse
|
12
|
Zhou H, Chen Y, Hu Y, Gao S, Lu W, He Y. Administration of All-Trans Retinoic Acid to Pregnant Sows Improves the Developmental Defects of Hoxa1 -/- Fetal Pigs. Front Vet Sci 2021; 7:618660. [PMID: 33506002 PMCID: PMC7829359 DOI: 10.3389/fvets.2020.618660] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/09/2020] [Indexed: 12/16/2022] Open
Abstract
Hoxa1 mutation adversely affect fetal pig development, but whether all-trans retinoic acid (ATRA) administration to Hoxa1+/− pregnant sows can improve Hoxa1−/− fetal pig development defects has not been reported. A total of 24 healthy Hoxa1+/− sows were mated with a healthy Hoxa1+/− boar and randomly assigned to one control group and nine experiment groups. ATRA was orally administered to pregnant sows at the doses of 0, 4, 5, or 6 mg/kg maternal body weight on 12, 13, and 14 days post coitum (dpc), respectively, and a total of 146 live piglets were delivered including 37 Hoxa1−/− piglets and 109 non-Hoxa1−/− piglets. Results indicated that Hoxa1−/− piglets delivered by sows in control group had bilateral microtia, canal atresia and ear's internal defects, and had lower birth liveweight and external ear score than non-Hoxa1−/− neonatal piglets (P < 0.05). Maternal administration with ATRA can effectively correct the development defects of Hoxa1−/− fetal pigs, Hoxa1−/− neonatal piglets delivered by sows administered ATRA at a dose of 4 mg/kg body weight on 14 dpc had higher birth liveweight (P > 0.05) and higher scores of external ear (P < 0.05) compared to Hoxa1−/− neonatal piglets from the control group, but had no significantly difference in terms of birth liveweight and external ear integrity than non-Hoxa1−/− piglets from the control group (P > 0.05). The time of ATRA administration significantly affected Hoxa1−/− fetal development (P < 0.05). Administration of ATRA to Hoxa1+/− pregnant sows at 4 mg/kg body weight on 14 dpc can effectively improve the birth liveweight and ear defects of Hoxa1−/− piglets.
Collapse
Affiliation(s)
- Haimei Zhou
- Jiangxi Province Key Laboratory of Animal Nutrition/Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang, China.,Department of Animal Science, Jiangxi Agricultural Engineering College, Zhangshu, China
| | - Yixin Chen
- Jiangxi Province Key Laboratory of Animal Nutrition/Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang, China
| | - Yongqiang Hu
- Jiangxi Province Key Laboratory of Animal Nutrition/Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang, China
| | - Shan Gao
- Jiangxi Province Key Laboratory of Animal Nutrition/Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang, China
| | - Wei Lu
- Jiangxi Province Key Laboratory of Animal Nutrition/Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang, China
| | - Yuyong He
- Jiangxi Province Key Laboratory of Animal Nutrition/Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang, China
| |
Collapse
|
13
|
Sasai N, Kadoya M, Ong Lee Chen A. Neural induction: Historical views and application to pluripotent stem cells. Dev Growth Differ 2021; 63:26-37. [PMID: 33289091 DOI: 10.1111/dgd.12703] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/22/2020] [Accepted: 11/02/2020] [Indexed: 12/20/2022]
Abstract
Embryonic stem (ES) cells are a useful experimental material to recapitulate the differentiation steps of early embryos, which are usually invisible and inaccessible from outside of the body, especially in mammals. ES cells have greatly facilitated the analyses of gene expression profiles and cell characteristics. In addition, understanding the mechanisms during neural differentiation is important for clinical purposes, such as developing new therapeutic methods or regenerative medicine. As neurons have very limited regenerative ability, neurodegenerative diseases are usually intractable, and patients suffer from the disease throughout their lifetimes. The functional cells generated from ES cells in vitro could replace degenerative areas by transplantation. In this review, we will first demonstrate the historical views and widely accepted concepts regarding the molecular mechanisms of neural induction and positional information to produce the specific types of neurons in model animals. Next, we will describe how these concepts have recently been applied to the research in the establishment of the methodology of neural differentiation from mammalian ES cells. Finally, we will focus on examples of the applications of differentiation systems to clinical purposes. Overall, the discussion will focus on how historical developmental studies are applied to state-of-the-art stem cell research.
Collapse
Affiliation(s)
- Noriaki Sasai
- Developmental Biomedical Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Minori Kadoya
- Developmental Biomedical Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Agnes Ong Lee Chen
- Developmental Biomedical Science, Nara Institute of Science and Technology, Ikoma, Japan
| |
Collapse
|
14
|
Rossillo M, Ringstad N. Development of specialized sensory neurons engages a nuclear receptor required for functional plasticity. Genes Dev 2020; 34:1666-1679. [PMID: 33184226 PMCID: PMC7706712 DOI: 10.1101/gad.342212.120] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
In this study, Rossillo and Ringstad sought to determine mechanisms that support the physiology and plasticity of BAG neurons, which are specialized neurons that sense the respiratory gas carbon dioxide (CO2) and, in a context-dependent manner, switch from mediating avoidance of CO2 to supporting CO2 attraction in C. elegans. They used tandem ChIP-seq and cell targeted RNA-seq to identify gene targets of the transcription factor ETS-5, which is required for BAG development, and their functional screen of ETS-5 targets revealed that NHR-6, the sole C. elegans NR4A-type nuclear receptor, is required for BAG-mediated avoidance of CO2 and regulates expression of a subset of BAG-specific genes. During development, the nervous system generates neurons that serve highly specialized roles and, accordingly, possess unique functional attributes. The chemosensory BAG neurons of C. elegans are striking exemplars of this. BAGs sense the respiratory gas carbon dioxide (CO2) and, in a context-dependent manner, switch from mediating avoidance of CO2 to supporting CO2 attraction. To determine mechanisms that support the physiology and plasticity of BAG neurons, we used tandem ChIP-seq and cell targeted RNA-seq to identify gene targets of the transcription factor ETS-5, which is required for BAG development. A functional screen of ETS-5 targets revealed that NHR-6, the sole C. elegans NR4A-type nuclear receptor, is required for BAG-mediated avoidance of CO2 and regulates expression of a subset of BAG-specific genes. Unlike ets-5 mutants, which are defective for both attraction to and avoidance of CO2, nhr-6 mutants are fully competent for attraction. These data indicate that the remarkable ability of BAGs to adaptively assign positive or negative valence to a chemosensory stimulus requires a gene-regulatory program supported by an evolutionarily conserved type of nuclear receptor. We suggest that NHR-6 might be an example of a developmental mechanism for modular encoding of functional plasticity in the nervous system.
Collapse
Affiliation(s)
- Mary Rossillo
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, Neuroscience Institute, New York University School of Medicine, New York, New York 10016, USA
| | - Niels Ringstad
- Skirball Institute of Biomolecular Medicine, Department of Cell Biology, Neuroscience Institute, New York University School of Medicine, New York, New York 10016, USA
| |
Collapse
|
15
|
Devoto C, Lai C, Qu BX, Guedes VA, Leete J, Wilde E, Walker WC, Diaz-Arrastia R, Kenney K, Gill J. Exosomal MicroRNAs in Military Personnel with Mild Traumatic Brain Injury: Preliminary Results from the Chronic Effects of Neurotrauma Consortium Biomarker Discovery Project. J Neurotrauma 2020; 37:2482-2492. [PMID: 32458732 PMCID: PMC7698974 DOI: 10.1089/neu.2019.6933] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic symptoms after mild traumatic brain injury (mTBI) are common among veterans and service members, and represent a significant source of morbidity, with those who sustain multiple mTBIs at greatest risk. Exosomal micro-RNAs (miRNAs), mediators of intercellular communication, may be involved in chronic TBI symptom persistence. Exosomal miRNA (exomiR) was extracted from 153 participants enrolled in the Chronic Effect of Neurotrauma Consortium (CENC) longitudinal study (no TBI, n = 35; ≥ 3 mTBIs (rTBI), n = 45; 1-2 mTBIs, n = 73). Analyses were performed with nCounter® Human miRNA Expression Panels and Ingenuity Pathway Analysis (IPA) for identification of gene networks associated with TBI. Generalized linear models were used to analyze the predictive value of exomiR dysregulation and remote neurobehavioral symptoms. Compared with controls, there were 17 dysregulated exomiRs in the entire mTBI group and 32 dysregulated exomiRs in the rTBI group. Two miRNAs, hsa-miR-139-5p and hsa-miR-18a-5p, were significantly differentially expressed in the rTBI and 1-2 mTBI groups. IPA analyses showed that these dysregulated exomiRs correlated with pathways of inflammatory regulation, neurological disease, and cell development. Within the rTBI group, exomiRs correlated with gene activity for hub-genes of tumor protein TP53, insulin-like growth factor 1 receptor, and transforming growth factor beta. TBI history and neurobehavioral symptom survey scores negatively and significantly correlated with hsa-miR-103a-3p expression. Participants with remote mTBI have distinct exomiR profiles, which are significantly linked to inflammatory and neuronal repair pathways. These profiles suggest that analysis of exosomal miRNA expression may provide novel insights into the underlying pathobiology of chronic TBI symptom persistence.
Collapse
Affiliation(s)
- Christina Devoto
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, Maryland, USA
- Center for Neuroscience and Rehabilitation Medicine, Uniformed Services University of the Health Sciences and National Institutes of Health, Bethesda, Maryland, USA
| | - Chen Lai
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Bao-Xi Qu
- Department of Neurology, Uniformed Services University of the Health Sciences and National Institutes of Health, Bethesda, Maryland, USA
- CENC Biorepository, Uniformed Services University of the Health Sciences, Twinbrook, Rockville, Maryland, USA
| | - Vivian A. Guedes
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Jacqueline Leete
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Elisabeth Wilde
- CENC Imaging Core, University of Utah, Salt Lake City, Utah, USA
| | - William C. Walker
- Department of Physical Medicine and Rehabilitation, Virginia Commonwealth University, Richmond, Virginia, USA
| | - Ramon Diaz-Arrastia
- Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kimbra Kenney
- Department of Neurology, Uniformed Services University of the Health Sciences and National Institutes of Health, Bethesda, Maryland, USA
- CENC Biorepository, Uniformed Services University of the Health Sciences, Twinbrook, Rockville, Maryland, USA
| | - Jessica Gill
- Tissue Injury Branch, National Institutes of Nursing Research, National Institutes of Health, Bethesda, Maryland, USA
- Center for Neuroscience and Rehabilitation Medicine, Uniformed Services University of the Health Sciences and National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
16
|
Kukreja S, Udaykumar N, Yogesh B, Sen J. Retinoic acid signaling regulates proliferation and lamina formation in the developing chick optic tectum. Dev Biol 2020; 467:95-107. [PMID: 32919944 DOI: 10.1016/j.ydbio.2020.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 09/03/2020] [Accepted: 09/06/2020] [Indexed: 01/05/2023]
Abstract
The retinotectal system has been extensively studied for investigating the mechanism(s) for topographic map formation. The optic tectum, which is composed of multiple laminae, is the major retino recipient structure in the developing avian brain. Laminar development of the tectum results from cell proliferation, differentiation and migration, coordinated in strict temporal and spatial patterns. However, the molecular mechanisms that orchestrate these complex developmental events, have not been fully elucidated. In this study, we have identified the presence of differential retinoic acid (RA) signaling along the rostro-caudal and dorsoventral axis of the tectum. We show for the first time that loss of RA signaling in the anterior optic tectum, leads to an increase in cell proliferation and gross changes in the morphology manifested as defects in lamination. Detailed analysis points to delayed migration of cells as the plausible cause for the defects in lamina formation. Thus, we conclude that in the optic tectum, RA signaling is involved in maintaining cell proliferation and in regulating the formation of the tectal laminae.
Collapse
Affiliation(s)
- Shweta Kukreja
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, Uttar Pradesh, India; Present address: Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, USA
| | - Niveda Udaykumar
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, Uttar Pradesh, India
| | - Baba Yogesh
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, Uttar Pradesh, India; Present address: Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Jonaki Sen
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Kanpur, 208016, Uttar Pradesh, India.
| |
Collapse
|
17
|
Desai D, Pethe P. Polycomb repressive complex 1: Regulators of neurogenesis from embryonic to adult stage. J Cell Physiol 2020; 235:4031-4045. [PMID: 31608994 DOI: 10.1002/jcp.29299] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/27/2019] [Indexed: 02/05/2023]
Abstract
Development of vertebrate nervous system is a complex process which involves differential gene expression and disruptions in this process or in the mature brain, may lead to neurological disorders and diseases. Extensive work that spanned several decades using rodent models and recent work on stem cells have helped uncover the intricate process of neuronal differentiation and maturation. There are various morphological changes, genetic and epigenetic modifications which occur during normal mammalian neural development, one of the chromatin modifications that controls vital gene expression are the posttranslational modifications on histone proteins, that controls accessibility of translational machinery. Among the histone modifiers, polycomb group proteins (PcGs), such as Ezh2, Eed and Suz12 form large protein complexes-polycomb repressive complex 2 (PRC2); while Ring1b and Bmi1 proteins form core of PRC1 along with accessory proteins such as Cbx, Hph, Rybp and Pcgfs catalyse histone modifications such as H3K27me3 and H2AK119ub1. PRC1 proteins are known to play critical role in X chromosome inactivation in females but they also repress the expression of key developmental genes and tightly regulate the mammalian neuronal development. In this review we have discussed the signalling pathways, morphogens and nuclear factors that initiate, regulate and maintain cells of the nervous system. Further, we have extensively reviewed the recent literature on the role of Ring1b and Bmi1 in mammalian neuronal development and differentiation; as well as highlighted questions that are still unanswered.
Collapse
Affiliation(s)
- Divya Desai
- Department of Biological Sciences, Sunandan Divatia School of Science (SDSOS), Narsee Monjee Institute of Management Studies (NMIMS) deemed-to-be University, Mumbai, India
| | - Prasad Pethe
- Symbiosis Centre for Stem Cell Research (SCSCR), Symbiosis International University (SIU), Pune, India
| |
Collapse
|
18
|
Lu Z, Xie Y, Huang H, Jiang K, Zhou B, Wang F, Chen T. Hair follicle stem cells regulate retinoid metabolism to maintain the self-renewal niche for melanocyte stem cells. eLife 2020; 9:e52712. [PMID: 31898934 PMCID: PMC6970533 DOI: 10.7554/elife.52712] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 01/03/2020] [Indexed: 12/18/2022] Open
Abstract
Metabolites are major biological parameters sensed by many cell types in vivo, whether they function as signaling mediators of SC and niche cross talk to regulate tissue regeneration is largely unknown. We show here that deletion of the Notch pathway co-factor RBP-J specifically in mouse HFSCs triggers adjacent McSCs to precociously differentiate in their shared niche. Transcriptome screen and in vivo functional studies revealed that the elevated level of retinoic acid (RA) caused by de-repression of RA metabolic process genes as a result of RBP-J deletion in HFSCs triggers ectopic McSCs differentiation in the niche. Mechanistically the increased level of RA sensitizes McSCs to differentiation signal KIT-ligand by increasing its c-Kit receptor protein level in vivo. Using genetic approach, we further pinpointed HFSCs as the source of KIT-ligand in the niche. We discover that HFSCs regulate the metabolite RA level in vivo to allow self-renewal of neighboring McSCs.
Collapse
Affiliation(s)
- Zhiwei Lu
- Peking Union Medical CollegeBeijingChina
- National Institute of Biological SciencesBeijingChina
| | - Yuhua Xie
- National Institute of Biological SciencesBeijingChina
| | - Huanwei Huang
- National Institute of Biological SciencesBeijingChina
| | - Kaiju Jiang
- National Institute of Biological SciencesBeijingChina
| | - Bin Zhou
- Institute of Biochemistry and Cell BiologyShanghai Institutes for Biological Sciences, University of Chinese Academy of SciencesBeijingChina
| | - Fengchao Wang
- National Institute of Biological SciencesBeijingChina
| | - Ting Chen
- National Institute of Biological SciencesBeijingChina
- Tsinghua Institute of Multidisciplinary Biomedical ResearchTsinghua UniversityBeijingChina
| |
Collapse
|
19
|
Cardeña-Núñez S, Sánchez-Guardado LÓ, Hidalgo-Sánchez M. Cyp1B1 expression patterns in the developing chick inner ear. Dev Dyn 2019; 249:410-424. [PMID: 31400045 DOI: 10.1002/dvdy.99] [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: 06/24/2019] [Revised: 07/26/2019] [Accepted: 07/26/2019] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Retinoic acid (RA) plays an important role in organogenesis as a paracrine signal through transcriptional regulation of an increasing number of known downstream target genes, regulating cell proliferation, and differentiation. During the development of the inner ear, RA directly governs the morphogenesis and specification processes mainly by means of RA-synthesizing retinaldehyde dehydrogenase (RALDH) enzymes. Interestingly, CYP1B1, a cytochrome P450 enzyme, is able to mediate the oxidative metabolisms also leading to RA generation, its expression patterns being associated with many known sites of RA activity. RESULTS This study describes for the first time the presence of CYP1B1 in the developing chick inner ear as a RALDH-independent RA-signaling mechanism. In our in situ hybridization analysis, Cyp1B1 expression was first observed in a domain located in the ventromedial wall of the otic anlagen, being included within the rostralmost aspect of an Fgf10-positive pan-sensory domain. As development proceeds, all identified Fgf10-positive areas were Cyp1B1 stained, with all sensory patches being Cyp1B1 positive at stage HH34, except the macula neglecta. CONCLUSIONS Cyp1B1 expression suggested a possible contribution of CYP1B1 action in the specification of the lateral-to-medial and dorsal-to-ventral axes of the developing chick inner ear.
Collapse
Affiliation(s)
- Sheila Cardeña-Núñez
- Department of Cell Biology, School of Science, University of Extremadura, Badajoz, Spain
| | - Luis Ó Sánchez-Guardado
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California
| | - Matías Hidalgo-Sánchez
- Department of Cell Biology, School of Science, University of Extremadura, Badajoz, Spain
| |
Collapse
|
20
|
Lin Y, Yu J, Wu J, Wang S, Zhang T. Abnormal level of CUL4B-mediated histone H2A ubiquitination causes disruptive HOX gene expression. Epigenetics Chromatin 2019; 12:22. [PMID: 30992047 PMCID: PMC6466687 DOI: 10.1186/s13072-019-0268-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/04/2019] [Indexed: 12/17/2022] Open
Abstract
Background Neural tube defects (NTDs) are common birth defects involving the central nervous system. Recent studies on the etiology of human NTDs have raised the possibility that epigenetic regulation could be involved in determining susceptibility to them. Results Here, we show that the H2AK119ub1 E3 ligase CUL4B is required for the activation of retinoic acid (RA)-inducible developmentally critical homeobox (HOX) genes in NT2/D1 embryonal carcinoma cells. RA treatment led to attenuation of H2AK119ub1 due to decrease in CUL4B, further affecting HOX gene regulation. Furthermore, we found that CUL4B interacted directly with RORγ and negatively regulated its transcriptional activity. Interestingly, knockdown of RORγ decreased the expression of HOX genes along with increased H2AK119ub1 occupancy levels, at HOX gene sites in N2/D1 cells. In addition, upregulation of HOX genes was observed along with lower levels of CUL4B-mediated H2AK119ub1 in both mouse and human anencephaly NTD cases. Notably, the expression of HOXA10 genes was negatively correlated with CUL4B levels in human anencephaly NTD cases. Conclusions Our results indicate that abnormal HOX gene expression induced by aberrant CUL4B-mediated H2AK119ub1 levels may be a risk factor for NTDs, and highlight the need for further analysis of genome-wide epigenetic modifications in NTDs. Electronic supplementary material The online version of this article (10.1186/s13072-019-0268-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ye Lin
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China.,Graduate Schools of Peking Union Medical College, Beijing, 100730, China
| | - Juan Yu
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jianxin Wu
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China.,Graduate Schools of Peking Union Medical College, Beijing, 100730, China
| | - Shan Wang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China. .,Institute of Basic Medical Sciences, Chinese Academy of Medical Science, Beijing, 100730, China.
| | - Ting Zhang
- Beijing Municipal Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing, 100020, China. .,Graduate Schools of Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
21
|
Nadadhur AG, Leferink PS, Holmes D, Hinz L, Cornelissen-Steijger P, Gasparotto L, Heine VM. Patterning factors during neural progenitor induction determine regional identity and differentiation potential in vitro. Stem Cell Res 2018; 32:25-34. [PMID: 30172094 DOI: 10.1016/j.scr.2018.08.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Revised: 08/13/2018] [Accepted: 08/22/2018] [Indexed: 12/20/2022] Open
Abstract
The neural tube consists of neural progenitors (NPs) that acquire different characteristics during gestation due to patterning factors. However, the influence of such patterning factors on human pluripotent stem cells (hPSCs) during in vitro neural differentiation is often unclear. This study compared neural induction protocols involving in vitro patterning with single SMAD inhibition (SSI), retinoic acid (RA) administration and dual SMAD inhibition (DSI). While the derived NP cells expressed known NP markers, they differed in their NP expression profile and differentiation potential. Cortical neuronal cells generated from 1) SSI NPs exhibited less mature neuronal phenotypes, 2) RA NPs exhibited an increased GABAergic phenotype, and 3) DSI NPs exhibited greater expression of glutamatergic lineage markers. Further, although all NPs generated astrocytes, astrocytes derived from the RA-induced NPs had the highest GFAP expression. Differences between NP populations included differential expression of regional identity markers HOXB4, LBX1, OTX1 and GSX2, which persisted into mature neural cell stages. This study suggests that patterning factors regulate how potential NPs may differentiate into specific neuronal and glial cell types in vitro. This challenges the utility of generic neural induction procedures, while highlighting the importance of carefully selecting specific NP protocols.
Collapse
Affiliation(s)
- Aishwarya G Nadadhur
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Prisca S Leferink
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Dwayne Holmes
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Lisa Hinz
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Paulien Cornelissen-Steijger
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Lisa Gasparotto
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands
| | - Vivi M Heine
- Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands; Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, the Netherlands.
| |
Collapse
|
22
|
Zou H, Lan Z, Zhou M, Lu W. Promoter methylation and Hoxd4 regulate UII mRNA tissue-specific expression in olive flounder (paralichthys olivaceus). Gen Comp Endocrinol 2018. [PMID: 29522756 DOI: 10.1016/j.ygcen.2018.03.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The peptide urotensin II (UII) mediates multiple physiology effects in mammals and fishes, and UII expression shows a tissue-specific pattern. However the mechanism is still unknown. In the present study high level of UII mRNA was detected in the caudal neurosecretory system (CNSS) of the olive flounder when compared to other tissues. We examined whether epigenetic mechanisms of DNA methylation are involved in UII gene expression. Methylation DNA immune precipitation (MeDIP) assay showed low methylation of UII promoter in CNSS tissue compared with muscle and spinal cord. Methylation of UII promoter was further assessed through bisulphate sequencing analysis. Low level methylation (31%) in CpG island of UII promoter was detected in CNSS tissue, while methylation status in muscle and spinal cord was 89% and 91%, respectively. In addition, high conserved sites of Hoxd4 in UII promoter were found. Activation of Hoxd4 mRNA using transretinoic acid (RA) resulted in 18-fold increase of UII mRNA expression in CNSS and high locomotor activity in medaka, confirming that Hoxd4 is also involved in UII gene transcriptional regulation. Taken together, our data provide the first evidence of the epigenetic mechanism of promoter methylation in transcriptional regulation of UII expression in a tissue-specific manner, and Hoxd4 may also participate in UII gene transcription in flounder.
Collapse
Affiliation(s)
- Huafeng Zou
- National Demonstration Center for Experimental Fisheries Science Education (Shanghai Ocean University), Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai 201306, China
| | - Zhaohui Lan
- National Demonstration Center for Experimental Fisheries Science Education (Shanghai Ocean University), Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai 201306, China
| | - Mo Zhou
- National Demonstration Center for Experimental Fisheries Science Education (Shanghai Ocean University), Shanghai 201306, China
| | - Weiqun Lu
- National Demonstration Center for Experimental Fisheries Science Education (Shanghai Ocean University), Shanghai 201306, China; Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai 201306, China; International Research Center for Marine Biosciences at Shanghai Ocean University, Ministry of Science and Technology, Shanghai 201306, China.
| |
Collapse
|
23
|
Glover JC, Elliott KL, Erives A, Chizhikov VV, Fritzsch B. Wilhelm His' lasting insights into hindbrain and cranial ganglia development and evolution. Dev Biol 2018; 444 Suppl 1:S14-S24. [PMID: 29447907 DOI: 10.1016/j.ydbio.2018.02.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 02/03/2018] [Accepted: 02/04/2018] [Indexed: 01/04/2023]
Abstract
Wilhelm His (1831-1904) provided lasting insights into the development of the central and peripheral nervous system using innovative technologies such as the microtome, which he invented. 150 years after his resurrection of the classical germ layer theory of Wolff, von Baer and Remak, his description of the developmental origin of cranial and spinal ganglia from a distinct cell population, now known as the neural crest, has stood the test of time and more recently sparked tremendous advances regarding the molecular development of these important cells. In addition to his 1868 treatise on 'Zwischenstrang' (now neural crest), his work on the development of the human hindbrain published in 1890 provided novel ideas that more than 100 years later form the basis for penetrating molecular investigations of the regionalization of the hindbrain neural tube and of the migration and differentiation of its constituent neuron populations. In the first part of this review we briefly summarize the major discoveries of Wilhelm His and his impact on the field of embryology. In the second part we relate His' observations to current knowledge about the molecular underpinnings of hindbrain development and evolution. We conclude with the proposition, present already in rudimentary form in the writings of His, that a primordial spinal cord-like organization has been molecularly supplemented to generate hindbrain 'neomorphs' such as the cerebellum and the auditory and vestibular nuclei and their associated afferents and sensory organs.
Collapse
Affiliation(s)
- Joel C Glover
- Department of Molecular Medicine, University of Oslo, Oslo, Norway; Norwegian Center for Stem Cell Research, Oslo University Hospital, Oslo, Norway; Sars International Centre for Marine Molecular Biology, University of Bergen, Bergen, Norway.
| | - Karen L Elliott
- Department of Biology, University of Iowa, Iowa, IA 52242, USA
| | - Albert Erives
- Department of Biology, University of Iowa, Iowa, IA 52242, USA
| | - Victor V Chizhikov
- The University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Bernd Fritzsch
- Department of Biology, University of Iowa, Iowa, IA 52242, USA.
| |
Collapse
|
24
|
Dubey A, Rose RE, Jones DR, Saint-Jeannet JP. Generating retinoic acid gradients by local degradation during craniofacial development: One cell's cue is another cell's poison. Genesis 2018; 56:10.1002/dvg.23091. [PMID: 29330906 PMCID: PMC5818312 DOI: 10.1002/dvg.23091] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/08/2018] [Accepted: 01/09/2018] [Indexed: 01/02/2023]
Abstract
Retinoic acid (RA) is a vital morphogen for early patterning and organogenesis in the developing embryo. RA is a diffusible, lipophilic molecule that signals via nuclear RA receptor heterodimeric units that regulate gene expression by interacting with RA response elements in promoters of a significant number of genes. For precise RA signaling, a robust gradient of the morphogen is required. The developing embryo contains regions that produce RA, and specific intracellular concentrations of RA are created through local degradation mediated by Cyp26 enzymes. In order to elucidate the mechanisms by which RA executes precise developmental programs, the kinetics of RA metabolism must be clearly understood. Recent advances in techniques for endogenous RA detection and quantification have paved the way for mechanistic studies to shed light on downstream gene expression regulation coordinated by RA. It is increasingly coming to light that RA signaling operates not only at precise concentrations but also employs mechanisms of degradation and feedback inhibition to self-regulate its levels. A global gradient of RA throughout the embryo is often found concurrently with several local gradients, created by juxtaposed domains of RA synthesis and degradation. The existence of such local gradients has been found especially critical for the proper development of craniofacial structures that arise from the neural crest and the cranial placode populations. In this review, we summarize the current understanding of how local gradients of RA are established in the embryo and their impact on craniofacial development.
Collapse
Affiliation(s)
- Aditi Dubey
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry
| | - Rebecca E. Rose
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, New York, NY, USA
| | - Drew R. Jones
- Department of Biochemistry and Molecular Pharmacology, New York University Langone Health, New York, NY, USA
| | | |
Collapse
|
25
|
Chagnaud BP, Engelmann J, Fritzsch B, Glover JC, Straka H. Sensing External and Self-Motion with Hair Cells: A Comparison of the Lateral Line and Vestibular Systems from a Developmental and Evolutionary Perspective. BRAIN, BEHAVIOR AND EVOLUTION 2017; 90:98-116. [PMID: 28988233 DOI: 10.1159/000456646] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Detection of motion is a feature essential to any living animal. In vertebrates, mechanosensory hair cells organized into the lateral line and vestibular systems are used to detect external water or head/body motion, respectively. While the neuronal components to detect these physical attributes are similar between the two sensory systems, the organizational pattern of the receptors in the periphery and the distribution of hindbrain afferent and efferent projections are adapted to the specific functions of the respective system. Here we provide a concise review comparing the functional organization of the vestibular and lateral line systems from the development of the organs to the wiring from the periphery and the first processing stages. The goal of this review is to highlight the similarities and differences to demonstrate how evolution caused a common neuronal substrate to adapt to different functions, one for the detection of external water stimuli and the generation of sensory maps and the other for the detection of self-motion and the generation of motor commands for immediate behavioral reactions.
Collapse
Affiliation(s)
- Boris P Chagnaud
- Ludwig-Maximilians-Universität München, Department Biology II, Division of Neurobiology, Martinsried-Planegg, Germany
| | | | | | | | | |
Collapse
|
26
|
Roles of Retinoic Acid Signaling in Shaping the Neuronal Architecture of the Developing Amphioxus Nervous System. Mol Neurobiol 2017; 55:5210-5229. [PMID: 28875454 DOI: 10.1007/s12035-017-0727-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 08/08/2017] [Indexed: 02/01/2023]
Abstract
The morphogen retinoic acid (RA) patterns vertebrate nervous systems and drives neurogenesis, but how these functions evolved remains elusive. Here, we show that RA signaling plays stage- and tissue-specific roles during the formation of neural cell populations with serotonin, dopamine, and GABA neurotransmitter phenotypes in amphioxus, a proxy for the ancestral chordate. Our data suggest that RA signaling restricts the specification of dopamine-containing cells in the ectoderm and of GABA neurons in the neural tube, probably by regulating Hox1 and Hox3 gene expression, respectively. The two Hox genes thus appear to serve distinct functions rather than to participate in a combinatorial Hox code. We were further able to correlate the RA signaling-dependent mispatterning of hindbrain GABA neurons with concomitant motor impairments. Taken together, these data provide new insights into how RA signaling and Hox genes contribute to nervous system as well as to motor control development in amphioxus and hence shed light on the evolution of these functions within vertebrates.
Collapse
|
27
|
Metzler MA, Sandell LL. Enzymatic Metabolism of Vitamin A in Developing Vertebrate Embryos. Nutrients 2016; 8:E812. [PMID: 27983671 PMCID: PMC5188467 DOI: 10.3390/nu8120812] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/09/2016] [Accepted: 12/13/2016] [Indexed: 12/16/2022] Open
Abstract
Embryonic development is orchestrated by a small number of signaling pathways, one of which is the retinoic acid (RA) signaling pathway. Vitamin A is essential for vertebrate embryonic development because it is the molecular precursor of the essential signaling molecule RA. The level and distribution of RA signaling within a developing embryo must be tightly regulated; too much, or too little, or abnormal distribution, all disrupt embryonic development. Precise regulation of RA signaling during embryogenesis is achieved by proteins involved in vitamin A metabolism, retinoid transport, nuclear signaling, and RA catabolism. The reversible first step in conversion of the precursor vitamin A to the active retinoid RA is mediated by retinol dehydrogenase 10 (RDH10) and dehydrogenase/reductase (SDR family) member 3 (DHRS3), two related membrane-bound proteins that functionally activate each other to mediate the interconversion of retinol and retinal. Alcohol dehydrogenase (ADH) enzymes do not contribute to RA production under normal conditions during embryogenesis. Genes involved in vitamin A metabolism and RA catabolism are expressed in tissue-specific patterns and are subject to feedback regulation. Mutations in genes encoding these proteins disrupt morphogenesis of many systems in a developing embryo. Together these observations demonstrate the importance of vitamin A metabolism in regulating RA signaling during embryonic development in vertebrates.
Collapse
Affiliation(s)
- Melissa A Metzler
- Department of Molecular, Cellular and Craniofacial Biology, University of Louisville, Louisville, KY 40201, USA.
| | - Lisa L Sandell
- Department of Molecular, Cellular and Craniofacial Biology, University of Louisville, Louisville, KY 40201, USA.
| |
Collapse
|
28
|
Salehi H, Amirpour N, Niapour A, Razavi S. An Overview of Neural Differentiation Potential of Human Adipose Derived Stem Cells. Stem Cell Rev Rep 2016; 12:26-41. [PMID: 26490462 DOI: 10.1007/s12015-015-9631-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There is wide interest in application of adult stem cells due to easy to obtain with a minimal patient discomfort, capable of producing cell numbers in large quantities and their immunocompatible properties without restriction by ethical concerns. Among these stem cells, multipotent mesenchymal stem cells (MSCs) from human adipose tissue are considered as an ideal source for various regenerative medicine. In spite of mesodermal origin of human adipose-derived stem cells (hADSCs), these cells have differentiation potential toward mesodermal and non-mesodermal lineages. Up to now, several studies have shown that hADSCs can undergo transdifferentiation and produce cells outside of their lineage, especially into neural cells when they are transferred to a specific cell environment. The purpose of this literature review is to provide an overview of the existing state of knowledge of the differentiation potential of hADSCs, specifically their ability to give rise to neuronal cells. The following review discusses different protocols considered for differentiation of hADSCs to neural cells, the neural markers that are used in each procedure and possible mechanisms that are involved in this differentiation.
Collapse
|
29
|
New Insights Into the Roles of Retinoic Acid Signaling in Nervous System Development and the Establishment of Neurotransmitter Systems. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 330:1-84. [PMID: 28215529 DOI: 10.1016/bs.ircmb.2016.09.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Secreted chiefly from the underlying mesoderm, the morphogen retinoic acid (RA) is well known to contribute to the specification, patterning, and differentiation of neural progenitors in the developing vertebrate nervous system. Furthermore, RA influences the subtype identity and neurotransmitter phenotype of subsets of maturing neurons, although relatively little is known about how these functions are mediated. This review provides a comprehensive overview of the roles played by RA signaling during the formation of the central and peripheral nervous systems of vertebrates and highlights its effects on the differentiation of several neurotransmitter systems. In addition, the evolutionary history of the RA signaling system is discussed, revealing both conserved properties and alternate modes of RA action. It is proposed that comparative approaches should be employed systematically to expand our knowledge of the context-dependent cellular mechanisms controlled by the multifunctional signaling molecule RA.
Collapse
|
30
|
LaMantia AS, Moody SA, Maynard TM, Karpinski BA, Zohn IE, Mendelowitz D, Lee NH, Popratiloff A. Hard to swallow: Developmental biological insights into pediatric dysphagia. Dev Biol 2015; 409:329-42. [PMID: 26554723 DOI: 10.1016/j.ydbio.2015.09.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 09/10/2015] [Accepted: 09/15/2015] [Indexed: 12/16/2022]
Abstract
Pediatric dysphagia-feeding and swallowing difficulties that begin at birth, last throughout childhood, and continue into maturity--is one of the most common, least understood complications in children with developmental disorders. We argue that a major cause of pediatric dysphagia is altered hindbrain patterning during pre-natal development. Such changes can compromise craniofacial structures including oropharyngeal muscles and skeletal elements as well as motor and sensory circuits necessary for normal feeding and swallowing. Animal models of developmental disorders that include pediatric dysphagia in their phenotypic spectrum can provide mechanistic insight into pathogenesis of feeding and swallowing difficulties. A fairly common human genetic developmental disorder, DiGeorge/22q11.2 Deletion Syndrome (22q11DS) includes a substantial incidence of pediatric dysphagia in its phenotypic spectrum. Infant mice carrying a parallel deletion to 22q11DS patients have feeding and swallowing difficulties that approximate those seen in pediatric dysphagia. Altered hindbrain patterning, craniofacial malformations, and changes in cranial nerve growth prefigure these difficulties. Thus, in addition to craniofacial and pharyngeal anomalies that arise independently of altered neural development, pediatric dysphagia may result from disrupted hindbrain patterning and its impact on peripheral and central neural circuit development critical for feeding and swallowing. The mechanisms that disrupt hindbrain patterning and circuitry may provide a foundation to develop novel therapeutic approaches for improved clinical management of pediatric dysphagia.
Collapse
Affiliation(s)
- Anthony-Samuel LaMantia
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Department of Pharmacology and Physiology, George Washington University, School of Medicine and Health Sciences, Washington D.C., USA
| | - Sally A Moody
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, Washington D.C., USA
| | - Thomas M Maynard
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Department of Pharmacology and Physiology, George Washington University, School of Medicine and Health Sciences, Washington D.C., USA
| | - Beverly A Karpinski
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Department of Pharmacology and Physiology, George Washington University, School of Medicine and Health Sciences, Washington D.C., USA
| | - Irene E Zohn
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Center for Neuroscience Research, Children's National Health System, Washington D.C., USA
| | - David Mendelowitz
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Department of Pharmacology and Physiology, George Washington University, School of Medicine and Health Sciences, Washington D.C., USA
| | - Norman H Lee
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Department of Pharmacology and Physiology, George Washington University, School of Medicine and Health Sciences, Washington D.C., USA
| | - Anastas Popratiloff
- Institute for Neuroscience, The George Washington University School of Medicine and Health Sciences, Washington D.C., USA; Department of Anatomy and Regenerative Biology, George Washington University, School of Medicine and Health Sciences, Washington D.C., USA
| |
Collapse
|
31
|
Terriente J, Pujades C. Cell segregation in the vertebrate hindbrain: a matter of boundaries. Cell Mol Life Sci 2015; 72:3721-30. [PMID: 26089248 PMCID: PMC11113478 DOI: 10.1007/s00018-015-1953-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 05/06/2015] [Accepted: 06/08/2015] [Indexed: 02/07/2023]
Abstract
Segregating cells into compartments during embryonic development is essential for growth and pattern formation. In the developing hindbrain, boundaries separate molecularly, physically and neuroanatomically distinct segments called rhombomeres. After rhombomeric cells have acquired their identity, interhombomeric boundaries restrict cell intermingling between adjacent rhombomeres and act as signaling centers to pattern the surrounding tissue. Several works have stressed the relevance of Eph/ephrin signaling in rhombomeric cell sorting. Recent data have unveiled the role of this pathway in the assembly of actomyosin cables as an important mechanism for keeping cells from different rhombomeres segregated. In this Review, we will provide a short summary of recent evidences gathered in different systems suggesting that physical actomyosin barriers can be a general mechanism for tissue separation. We will discuss current evidences supporting a model where cell-cell signaling pathways, such as Eph/ephrin, govern compartmental cell sorting through modulation of the actomyosin cytoskeleton and cell adhesive properties to prevent cell intermingling.
Collapse
Affiliation(s)
- Javier Terriente
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, PRBB, Dr Aiguader 88, 08003, Barcelona, Spain.
| | - Cristina Pujades
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, PRBB, Dr Aiguader 88, 08003, Barcelona, Spain.
| |
Collapse
|
32
|
Chang JT, Lehtinen MK, Sive H. Zebrafish cerebrospinal fluid mediates cell survival through a retinoid signaling pathway. Dev Neurobiol 2015; 76:75-92. [PMID: 25980532 PMCID: PMC4644717 DOI: 10.1002/dneu.22300] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 11/07/2022]
Abstract
Cerebrospinal fluid (CSF) includes conserved factors whose function is largely unexplored. To assess the role of CSF during embryonic development, CSF was repeatedly drained from embryonic zebrafish brain ventricles soon after their inflation. Removal of CSF increased cell death in the diencephalon, indicating a survival function. Factors within the CSF are required for neuroepithelial cell survival as injected mouse CSF but not artificial CSF could prevent cell death after CSF depletion. Mass spectrometry analysis of the CSF identified retinol binding protein 4 (Rbp4), which transports retinol, the precursor to retinoic acid (RA). Consistent with a role for Rbp4 in cell survival, inhibition of Rbp4 or RA synthesis increased neuroepithelial cell death. Conversely, ventricle injection of exogenous human RBP4 plus retinol, or RA alone prevented cell death after CSF depletion. Zebrafish rbp4 is highly expressed in the yolk syncytial layer, suggesting Rbp4 protein and retinol/RA precursors can be transported into the CSF from the yolk. In accord with this suggestion, injection of human RBP4 protein into the yolk prevents neuroepithelial cell death in rbp4 loss‐of‐function embryos. Together, these data support the model that Rbp4 and RA precursors are present within the CSF and used for synthesis of RA, which promotes embryonic neuroepithelial survival. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 76: 75–92, 2016
Collapse
Affiliation(s)
- Jessica T Chang
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, Massachusetts, 02142.,Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, Massachusetts, 02115
| | - Hazel Sive
- Whitehead Institute for Biomedical Research, Nine Cambridge Center, Cambridge, Massachusetts, 02142.,Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139
| |
Collapse
|
33
|
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
|
34
|
Meechan DW, Maynard TM, Tucker ES, Fernandez A, Karpinski BA, Rothblat LA, LaMantia AS. Modeling a model: Mouse genetics, 22q11.2 Deletion Syndrome, and disorders of cortical circuit development. Prog Neurobiol 2015; 130:1-28. [PMID: 25866365 DOI: 10.1016/j.pneurobio.2015.03.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 03/24/2015] [Accepted: 03/29/2015] [Indexed: 12/21/2022]
Abstract
Understanding the developmental etiology of autistic spectrum disorders, attention deficit/hyperactivity disorder and schizophrenia remains a major challenge for establishing new diagnostic and therapeutic approaches to these common, difficult-to-treat diseases that compromise neural circuits in the cerebral cortex. One aspect of this challenge is the breadth and overlap of ASD, ADHD, and SCZ deficits; another is the complexity of mutations associated with each, and a third is the difficulty of analyzing disrupted development in at-risk or affected human fetuses. The identification of distinct genetic syndromes that include behavioral deficits similar to those in ASD, ADHC and SCZ provides a critical starting point for meeting this challenge. We summarize clinical and behavioral impairments in children and adults with one such genetic syndrome, the 22q11.2 Deletion Syndrome, routinely called 22q11DS, caused by micro-deletions of between 1.5 and 3.0 MB on human chromosome 22. Among many syndromic features, including cardiovascular and craniofacial anomalies, 22q11DS patients have a high incidence of brain structural, functional, and behavioral deficits that reflect cerebral cortical dysfunction and fall within the spectrum that defines ASD, ADHD, and SCZ. We show that developmental pathogenesis underlying this apparent genetic "model" syndrome in patients can be defined and analyzed mechanistically using genomically accurate mouse models of the deletion that causes 22q11DS. We conclude that "modeling a model", in this case 22q11DS as a model for idiopathic ASD, ADHD and SCZ, as well as other behavioral disorders like anxiety frequently seen in 22q11DS patients, in genetically engineered mice provides a foundation for understanding the causes and improving diagnosis and therapy for these disorders of cortical circuit development.
Collapse
Affiliation(s)
- Daniel W Meechan
- Institute for Neuroscience, Department of Pharmacology & Physiology, The George Washington University, Washington, DC, United States
| | - Thomas M Maynard
- Institute for Neuroscience, Department of Pharmacology & Physiology, The George Washington University, Washington, DC, United States
| | - Eric S Tucker
- Department of Neurobiology and Anatomy, Neuroscience Graduate Program, and Center for Neuroscience, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Alejandra Fernandez
- Institute for Neuroscience, Department of Pharmacology & Physiology, The George Washington University, Washington, DC, United States
| | - Beverly A Karpinski
- Institute for Neuroscience, Department of Pharmacology & Physiology, The George Washington University, Washington, DC, United States
| | - Lawrence A Rothblat
- Institute for Neuroscience, Department of Pharmacology & Physiology, The George Washington University, Washington, DC, United States; Department of Psychology, The George Washington University, Washington, DC, United States
| | - Anthony-S LaMantia
- Institute for Neuroscience, Department of Pharmacology & Physiology, The George Washington University, Washington, DC, United States.
| |
Collapse
|
35
|
Janesick A, Wu SC, Blumberg B. Retinoic acid signaling and neuronal differentiation. Cell Mol Life Sci 2015; 72:1559-76. [PMID: 25558812 PMCID: PMC11113123 DOI: 10.1007/s00018-014-1815-9] [Citation(s) in RCA: 209] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 12/15/2014] [Accepted: 12/19/2014] [Indexed: 01/13/2023]
Abstract
The identification of neurological symptoms caused by vitamin A deficiency pointed to a critical, early developmental role of vitamin A and its metabolite, retinoic acid (RA). The ability of RA to induce post-mitotic, neural phenotypes in various stem cells, in vitro, served as early evidence that RA is involved in the switch between proliferation and differentiation. In vivo studies have expanded this "opposing signal" model, and the number of primary neurons an embryo develops is now known to depend critically on the levels and spatial distribution of RA. The proneural and neurogenic transcription factors that control the exit of neural progenitors from the cell cycle and allow primary neurons to develop are partly elucidated, but the downstream effectors of RA receptor (RAR) signaling (many of which are putative cell cycle regulators) remain largely unidentified. The molecular mechanisms underlying RA-induced primary neurogenesis in anamniote embryos are starting to be revealed; however, these data have been not been extended to amniote embryos. There is growing evidence that bona fide RARs are found in some mollusks and other invertebrates, but little is known about their necessity or functions in neurogenesis. One normal function of RA is to regulate the cell cycle to halt proliferation, and loss of RA signaling is associated with dedifferentiation and the development of cancer. Identifying the genes and pathways that mediate cell cycle exit downstream of RA will be critical for our understanding of how to target tumor differentiation. Overall, elucidating the molecular details of RAR-regulated neurogenesis will be decisive for developing and understanding neural proliferation-differentiation switches throughout development.
Collapse
Affiliation(s)
- Amanda Janesick
- Department of Developmental and Cell Biology, 2011 Biological Sciences 3, University of California, Irvine, 92697-2300 USA
| | - Stephanie Cherie Wu
- Department of Developmental and Cell Biology, 2011 Biological Sciences 3, University of California, Irvine, 92697-2300 USA
| | - Bruce Blumberg
- Department of Developmental and Cell Biology, 2011 Biological Sciences 3, University of California, Irvine, 92697-2300 USA
- Department of Pharmaceutical Sciences, University of California, Irvine, USA
| |
Collapse
|
36
|
Wai HA, Kawakami K, Wada H, Müller F, Vernallis AB, Brown G, Johnson WEB. The development and growth of tissues derived from cranial neural crest and primitive mesoderm is dependent on the ligation status of retinoic acid receptor γ: evidence that retinoic acid receptor γ functions to maintain stem/progenitor cells in the absence of retinoic acid. Stem Cells Dev 2015; 24:507-19. [PMID: 25233141 PMCID: PMC4313414 DOI: 10.1089/scd.2014.0235] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 09/18/2014] [Indexed: 12/21/2022] Open
Abstract
Retinoic acid (RA) signaling is important to normal development. However, the function of the different RA receptors (RARs)--RARα, RARβ, and RARγ--is as yet unclear. We have used wild-type and transgenic zebrafish to examine the role of RARγ. Treatment of zebrafish embryos with an RARγ-specific agonist reduced somite formation and axial length, which was associated with a loss of hoxb13a expression and less-clear alterations in hoxc11a or myoD expression. Treatment with the RARγ agonist also disrupted formation of tissues arising from cranial neural crest, including cranial bones and anterior neural ganglia. There was a loss of Sox 9-immunopositive neural crest stem/progenitor cells in the same anterior regions. Pectoral fin outgrowth was blocked by RARγ agonist treatment. However, there was no loss of Tbx-5-immunopositive lateral plate mesodermal stem/progenitor cells and the block was reversed by agonist washout or by cotreatment with an RARγ antagonist. Regeneration of the caudal fin was also blocked by RARγ agonist treatment, which was associated with a loss of canonical Wnt signaling. This regenerative response was restored by agonist washout or cotreatment with the RARγ antagonist. These findings suggest that RARγ plays an essential role in maintaining stem/progenitor cells during embryonic development and tissue regeneration when the receptor is in its nonligated state.
Collapse
Affiliation(s)
- Htoo Aung Wai
- Life and Health Sciences, Aston University, Birmingham, United Kingdom
| | - Koichi Kawakami
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Japan
| | - Hironori Wada
- Division of Molecular and Developmental Biology, National Institute of Genetics, Mishima, Japan
| | - Ferenc Müller
- School of Clinical and Experimental Medicine, University of Birmingham, United Kingdom
| | | | - Geoffrey Brown
- School of Immunity and Infection, University of Birmingham, United Kingdom
| | | |
Collapse
|
37
|
Willaredt MA, Schlüter T, Nothwang HG. The gene regulatory networks underlying formation of the auditory hindbrain. Cell Mol Life Sci 2015; 72:519-535. [PMID: 25332098 PMCID: PMC11113740 DOI: 10.1007/s00018-014-1759-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 09/24/2014] [Accepted: 10/09/2014] [Indexed: 01/28/2023]
Abstract
Development and evolution of auditory hindbrain nuclei are two major unsolved issues in hearing research. Recent characterization of transgenic mice identified the rhombomeric origins of mammalian auditory nuclei and unraveled genes involved in their formation. Here, we provide an overview on these data by assembling them into rhombomere-specific gene regulatory networks (GRNs), as they underlie developmental and evolutionary processes. To explore evolutionary mechanisms, we compare the GRNs operating in the mammalian auditory hindbrain with data available from the inner ear and other vertebrate groups. Finally, we propose that the availability of genomic sequences from all major vertebrate taxa and novel genetic techniques for non-model organisms provide an unprecedented opportunity to investigate development and evolution of the auditory hindbrain by comparative molecular approaches. The dissection of the molecular mechanisms leading to auditory structures will also provide an important framework for auditory processing disorders, a clinical problem difficult to tackle so far. These data will, therefore, foster basic and clinical hearing research alike.
Collapse
Affiliation(s)
- Marc A Willaredt
- Neurogenetics group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany.
| | - Tina Schlüter
- Neurogenetics group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany
| | - Hans Gerd Nothwang
- Neurogenetics group, Center of Excellence Hearing4All, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany.
- Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, 26111, Oldenburg, Germany.
| |
Collapse
|
38
|
di Masi A, Leboffe L, De Marinis E, Pagano F, Cicconi L, Rochette-Egly C, Lo-Coco F, Ascenzi P, Nervi C. Retinoic acid receptors: from molecular mechanisms to cancer therapy. Mol Aspects Med 2015; 41:1-115. [PMID: 25543955 DOI: 10.1016/j.mam.2014.12.003] [Citation(s) in RCA: 256] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 12/15/2014] [Indexed: 02/07/2023]
Abstract
Retinoic acid (RA), the major bioactive metabolite of retinol or vitamin A, induces a spectrum of pleiotropic effects in cell growth and differentiation that are relevant for embryonic development and adult physiology. The RA activity is mediated primarily by members of the retinoic acid receptor (RAR) subfamily, namely RARα, RARβ and RARγ, which belong to the nuclear receptor (NR) superfamily of transcription factors. RARs form heterodimers with members of the retinoid X receptor (RXR) subfamily and act as ligand-regulated transcription factors through binding specific RA response elements (RAREs) located in target genes promoters. RARs also have non-genomic effects and activate kinase signaling pathways, which fine-tune the transcription of the RA target genes. The disruption of RA signaling pathways is thought to underlie the etiology of a number of hematological and non-hematological malignancies, including leukemias, skin cancer, head/neck cancer, lung cancer, breast cancer, ovarian cancer, prostate cancer, renal cell carcinoma, pancreatic cancer, liver cancer, glioblastoma and neuroblastoma. Of note, RA and its derivatives (retinoids) are employed as potential chemotherapeutic or chemopreventive agents because of their differentiation, anti-proliferative, pro-apoptotic, and anti-oxidant effects. In humans, retinoids reverse premalignant epithelial lesions, induce the differentiation of myeloid normal and leukemic cells, and prevent lung, liver, and breast cancer. Here, we provide an overview of the biochemical and molecular mechanisms that regulate the RA and retinoid signaling pathways. Moreover, mechanisms through which deregulation of RA signaling pathways ultimately impact on cancer are examined. Finally, the therapeutic effects of retinoids are reported.
Collapse
Affiliation(s)
- Alessandra di Masi
- Department of Science, Roma Tre University, Viale Guglielmo Marconi 446, Roma I-00146, Italy
| | - Loris Leboffe
- Department of Science, Roma Tre University, Viale Guglielmo Marconi 446, Roma I-00146, Italy
| | - Elisabetta De Marinis
- Department of Medical and Surgical Sciences and Biotechnologies, University of Roma "La Sapienza", Corso della Repubblica 79, Latina I-04100
| | - Francesca Pagano
- Department of Medical and Surgical Sciences and Biotechnologies, University of Roma "La Sapienza", Corso della Repubblica 79, Latina I-04100
| | - Laura Cicconi
- Department of Biomedicine and Prevention, University of Roma "Tor Vergata", Via Montpellier 1, Roma I-00133, Italy; Laboratory of Neuro-Oncohematology, Santa Lucia Foundation, Via Ardeatina, 306, Roma I-00142, Italy
| | - Cécile Rochette-Egly
- Department of Functional Genomics and Cancer, IGBMC, CNRS UMR 7104 - Inserm U 964, University of Strasbourg, 1 rue Laurent Fries, BP10142, Illkirch Cedex F-67404, France.
| | - Francesco Lo-Coco
- Department of Biomedicine and Prevention, University of Roma "Tor Vergata", Via Montpellier 1, Roma I-00133, Italy; Laboratory of Neuro-Oncohematology, Santa Lucia Foundation, Via Ardeatina, 306, Roma I-00142, Italy.
| | - Paolo Ascenzi
- Interdepartmental Laboratory for Electron Microscopy, Roma Tre University, Via della Vasca Navale 79, Roma I-00146, Italy.
| | - Clara Nervi
- Department of Medical and Surgical Sciences and Biotechnologies, University of Roma "La Sapienza", Corso della Repubblica 79, Latina I-04100.
| |
Collapse
|
39
|
Lee K, Skromne I. Retinoic acid regulates size, pattern and alignment of tissues at the head-trunk transition. Development 2015; 141:4375-84. [PMID: 25371368 DOI: 10.1242/dev.109603] [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] [Indexed: 11/20/2022]
Abstract
At the head-trunk transition, hindbrain and spinal cord alignment to occipital and vertebral bones is crucial for coherent neural and skeletal system organization. Changes in neural or mesodermal tissue configuration arising from defects in the specification, patterning or relative axial placement of territories can severely compromise their integration and function. Here, we show that coordination of neural and mesodermal tissue at the zebrafish head-trunk transition crucially depends on two novel activities of the signaling factor retinoic acid (RA): one specifying the size and the other specifying the axial position relative to mesodermal structures of the hindbrain territory. These activities are each independent but coordinated with the well-established function of RA in hindbrain patterning. Using neural and mesodermal landmarks we demonstrate that the functions of RA in aligning neural and mesodermal tissues temporally precede the specification of hindbrain and spinal cord territories and the activation of hox transcription. Using cell transplantation assays we show that RA activity in the neuroepithelium regulates hindbrain patterning directly and territory size specification indirectly. This indirect function is partially dependent on Wnts but independent of FGFs. Importantly, RA specifies and patterns the hindbrain territory by antagonizing the activity of the spinal cord specification gene cdx4; loss of Cdx4 rescues the defects associated with the loss of RA, including the reduction in hindbrain size and the loss of posterior rhombomeres. We propose that at the head-trunk transition, RA coordinates specification, patterning and alignment of neural and mesodermal tissues that are essential for the organization and function of the neural and skeletal systems.
Collapse
Affiliation(s)
- Keun Lee
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA
| | - Isaac Skromne
- Department of Biology, University of Miami, Coral Gables, FL 33146, USA
| |
Collapse
|
40
|
Gutierrez-Mazariegos J, Schubert M, Laudet V. Evolution of retinoic acid receptors and retinoic acid signaling. Subcell Biochem 2014; 70:55-73. [PMID: 24962881 DOI: 10.1007/978-94-017-9050-5_4] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Retinoic acid (RA) is a vitamin A-derived morphogen controlling important developmental processes in vertebrates, and more generally in chordates, including axial patterning and tissue formation and differentiation. In the embryo, endogenous RA levels are controlled by RA synthesizing and degrading enzymes and the RA signal is transduced by two retinoid receptors: the retinoic acid receptor (RAR) and the retinoid X receptor (RXR). Both RAR and RXR are members of the nuclear receptor superfamily of ligand-activated transcription factors and mainly act as heterodimers to activate the transcription of target genes in the presence of their ligand, all-trans RA. This signaling pathway was long thought to be a chordate innovation, however, recent findings of gene homologs involved in RA signaling in the genomes of a wide variety of non-chordate animals, including ambulacrarians (sea urchins and acorn worms) and lophotrochozoans (annelids and mollusks), challenged this traditional view and suggested that the RA signaling pathway might have a more ancient evolutionary origin than previously thought. In this chapter, we discuss the evolutionary history of the RA signaling pathway, and more particularly of the RARs, which might have experienced independent gene losses and duplications in different animal lineages. In sum, the available data reveal novel insights into the origin of the RA signaling pathway as well as into the evolutionary history of the RARs.
Collapse
Affiliation(s)
- Juliana Gutierrez-Mazariegos
- Molecular Zoology Team, Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, INRA, Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364, Lyon Cedex 07, France,
| | | | | |
Collapse
|
41
|
Knutson DC, Clagett-Dame M. A complex RARE is required for the majority of Nedd9 embryonic expression. Transgenic Res 2014; 24:123-34. [PMID: 25120220 PMCID: PMC4274375 DOI: 10.1007/s11248-014-9825-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 08/01/2014] [Indexed: 11/16/2022]
Abstract
Neural precursor cell expressed, developmentally down-regulated 9 (Nedd9, Casl, Hef1, p105cas, Ef1) is a scaffolding protein that assembles complexes involved in regulating cell adhesion, migration, division, and survival. Nedd9 is found very early in the developing embryonic nervous system. A highly conserved complex retinoic acid response element (RARE) is located 485 base pairs (bp) upstream of exon 2B in the promoter of the Nedd9 gene. Mice transgenic for a 5.2 kilobase (kb) region of the 2B Nedd9 promoter containing the RARE upstream of a lacZ reporter gene [Nedd9(RARE)-lacZ] show a large subset of the normal endogenous Nedd9 expression including that in the caudal hindbrain neuroepithelium, spinal cord, dorsal root ganglia (drg) and migrating neural crest (ncc). However, the transgenic mice do not recapitulate the native Nedd9 expression pattern in presumptive rhombomeres (pr) 3 and 5 of the early hindbrain, the base of the neuroepithelium in the midbrain, nor the forebrain telencephalon. Thus, the 5.2 kb region containing the intact RARE drives a large subset of Nedd9 expression, with additional sequences outside of this region needed to define the full complement of expression. When the 5.2 kb construct is modified (eight point mutations) to eliminate responsiveness of the RARE to all-trans retinoic acid (atRA) [Nedd9(mutRARE)-lacZ], virtually all β-galactosidase (β-gal, lacZ) expression is lost. Exposure of Nedd9(RARE)-lacZ transgenic embryos to excess atRA at embryonic day 8.0 (E8.0) leads to rostral ectopic transgene expression within 6 h whereas the Nedd9(mutRARE)-lacZ mutant does not show this effect. Thus the RARE upstream of the Nedd9 2B promoter is necessary for much of the endogenous gene expression during early development as well as ectopic expression in response to atRA.
Collapse
Affiliation(s)
- Danielle C Knutson
- Department of Biochemistry, University of Wisconsin-Madison, 433 Babcock Drive, Madison, WI, 53706, USA
| | | |
Collapse
|
42
|
Sukiban J, Bräunig P, Mey J, Bui-Göbbels K. Retinoic acid as a survival factor in neuronal development of the grasshopper, Locusta migratoria. Cell Tissue Res 2014; 358:303-12. [PMID: 25107605 DOI: 10.1007/s00441-014-1957-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 07/03/2014] [Indexed: 12/23/2022]
Abstract
Based on experience with cell cultures of adult insect neurons, we develop a serum-free culture system for embryonic locust neurons. Influences of trophic substances on survival and neurite outgrowth of developing neurons are investigated. For the first time, a positive trophic effect of 9-cis retinoic acid (9-cis RA) was shown in vitro on embryonic neurons of an insect. We observed longer cell survival of 50 % developmental stage neurons in cultures supplemented with 0.3 nM 9-cis RA. Furthermore, an influence on neuron morphology was revealed, as the addition of 9-cis RA to cell culture medium led to an increase in the number of neurites per cell. Although an RA receptor gene, LmRXR (Locusta migratoria retinoid X receptor), was expressed in the central nervous system throughout development, the influence of 9-cis RA on neuronal survival and outgrowth was restricted to 50 % stage embryonic cells.
Collapse
Affiliation(s)
- Jeyathevy Sukiban
- Institut für Biologie II (Zoologie), RWTH Aachen University, Worringer Weg 3, 52074, Aachen, Germany
| | | | | | | |
Collapse
|
43
|
Tahir R, Kennedy A, Elsea SH, Dickinson AJ. Retinoic acid induced-1 (Rai1) regulates craniofacial and brain development in Xenopus. Mech Dev 2014; 133:91-104. [PMID: 24878353 DOI: 10.1016/j.mod.2014.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 05/16/2014] [Accepted: 05/19/2014] [Indexed: 12/18/2022]
Abstract
Retinoic acid induced-1 (RAI1) is an important yet understudied histone code reader that when mutated in humans results in Smith-Magenis syndrome (SMS), a neurobehavioral disorder accompanied by signature craniofacial abnormalities. Despite previous studies in mouse and human cell models, very little is known about the function of RAI1 during embryonic development. In the present study, we have turned to the model vertebrates Xenopus laevis and Xenopus tropicalis to better understand the developmental roles of Rai1. First we demonstrate that the Rai1 protein sequence is conserved in frogs, especially in known functional domains. By in situ hybridization we revealed expression of rai1 in the developing craniofacial tissues and the nervous system. Knockdown of Rai1 using antisense morpholinos resulted in defects in the developing brain and face. In particular, Rai1 morphants display midface hypoplasia and malformed mouth shape analogous to defects in humans with SMS. These craniofacial defects were accompanied with aberrant neural crest migration and reduction in the size of facial cartilage elements. Rai1 morphants also had defects in axon patterns and decreased forebrain ventricle size. Such brain defects correlated with a decrease in the neurotrophic factor, bdnf, and increased forebrain apoptosis. Our results emphasize a critical role of Rai1 for normal neural and craniofacial development, and further the current understanding of potential mechanisms that cause SMS.
Collapse
Affiliation(s)
- Raiha Tahir
- Center of the Study of Biological Complexity, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Allyson Kennedy
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Sarah H Elsea
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, MS NAB2015, Houston, TX 77030, USA
| | - Amanda J Dickinson
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA.
| |
Collapse
|
44
|
Matilla-Dueñas A, Ashizawa T, Brice A, Magri S, McFarland KN, Pandolfo M, Pulst SM, Riess O, Rubinsztein DC, Schmidt J, Schmidt T, Scoles DR, Stevanin G, Taroni F, Underwood BR, Sánchez I. Consensus paper: pathological mechanisms underlying neurodegeneration in spinocerebellar ataxias. CEREBELLUM (LONDON, ENGLAND) 2014; 13:269-302. [PMID: 24307138 PMCID: PMC3943639 DOI: 10.1007/s12311-013-0539-y] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Intensive scientific research devoted in the recent years to understand the molecular mechanisms or neurodegeneration in spinocerebellar ataxias (SCAs) are identifying new pathways and targets providing new insights and a better understanding of the molecular pathogenesis in these diseases. In this consensus manuscript, the authors discuss their current views on the identified molecular processes causing or modulating the neurodegenerative phenotype in spinocerebellar ataxias with the common opinion of translating the new knowledge acquired into candidate targets for therapy. The following topics are discussed: transcription dysregulation, protein aggregation, autophagy, ion channels, the role of mitochondria, RNA toxicity, modulators of neurodegeneration and current therapeutic approaches. Overall point of consensus includes the common vision of neurodegeneration in SCAs as a multifactorial, progressive and reversible process, at least in early stages. Specific points of consensus include the role of the dysregulation of protein folding, transcription, bioenergetics, calcium handling and eventual cell death with apoptotic features of neurons during SCA disease progression. Unresolved questions include how the dysregulation of these pathways triggers the onset of symptoms and mediates disease progression since this understanding may allow effective treatments of SCAs within the window of reversibility to prevent early neuronal damage. Common opinions also include the need for clinical detection of early neuronal dysfunction, for more basic research to decipher the early neurodegenerative process in SCAs in order to give rise to new concepts for treatment strategies and for the translation of the results to preclinical studies and, thereafter, in clinical practice.
Collapse
Affiliation(s)
- A Matilla-Dueñas
- Health Sciences Research Institute Germans Trias i Pujol (IGTP), Ctra. de Can Ruti, Camí de les Escoles s/n, Badalona, Barcelona, Spain,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Das BC, Thapa P, Karki R, Das S, Mahapatra S, Liu TC, Torregroza I, Wallace DP, Kambhampati S, Van Veldhuizen P, Verma A, Ray SK, Evans T. Retinoic acid signaling pathways in development and diseases. Bioorg Med Chem 2014; 22:673-83. [PMID: 24393720 PMCID: PMC4447240 DOI: 10.1016/j.bmc.2013.11.025] [Citation(s) in RCA: 176] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 11/04/2013] [Accepted: 11/13/2013] [Indexed: 02/07/2023]
Abstract
Retinoids comprise a group of compounds each composed of three basic parts: a trimethylated cyclohexene ring that is a bulky hydrophobic group, a conjugated tetraene side chain that functions as a linker unit, and a polar carbon-oxygen functional group. Biochemical conversion of carotenoid or other retinoids to retinoic acid (RA) is essential for normal regulation of a wide range of biological processes including development, differentiation, proliferation, and apoptosis. Retinoids regulate various physiological outputs by binding to nuclear receptors called retinoic acid receptors (RARs) and retinoid X receptors (RXRs), which themselves are DNA-binding transcriptional regulators. The functional response of RA and their receptors are modulated by a host of coactivators and corepressors. Retinoids are essential in the development and function of several organ systems; however, deregulated retinoid signaling can contribute to serious diseases. Several natural and synthetic retinoids are in clinical use or undergoing trials for treating specific diseases including cancer. In this review, we provide a broad overview on the importance of retinoids in development and various diseases, highlighting various retinoids in the drug discovery process, ranging all the way from retinoid chemistry to clinical uses and imaging.
Collapse
Affiliation(s)
- Bhaskar C Das
- Division of Hematology and Oncology, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA; Molecular Bio-nanotechnology, Imaging and Therapeutic Research Unit, Veteran Affairs Medical Center, Kansas City, MO 64128, USA; Department of Surgery, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA; The Kidney Institute, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA.
| | - Pritam Thapa
- Division of Hematology and Oncology, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA; Molecular Bio-nanotechnology, Imaging and Therapeutic Research Unit, Veteran Affairs Medical Center, Kansas City, MO 64128, USA
| | - Radha Karki
- Division of Hematology and Oncology, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA; Molecular Bio-nanotechnology, Imaging and Therapeutic Research Unit, Veteran Affairs Medical Center, Kansas City, MO 64128, USA
| | - Sasmita Das
- Division of Hematology and Oncology, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA; Molecular Bio-nanotechnology, Imaging and Therapeutic Research Unit, Veteran Affairs Medical Center, Kansas City, MO 64128, USA
| | - Sweta Mahapatra
- Division of Hematology and Oncology, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA; Molecular Bio-nanotechnology, Imaging and Therapeutic Research Unit, Veteran Affairs Medical Center, Kansas City, MO 64128, USA
| | - Ting-Chun Liu
- Department of Surgery, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Ingrid Torregroza
- Department of Surgery, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA
| | - Darren P Wallace
- The Kidney Institute, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA
| | - Suman Kambhampati
- Division of Hematology and Oncology, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA; Molecular Bio-nanotechnology, Imaging and Therapeutic Research Unit, Veteran Affairs Medical Center, Kansas City, MO 64128, USA
| | - Peter Van Veldhuizen
- Division of Hematology and Oncology, Department of Internal Medicine, Kansas University Medical Center, Kansas City, KS 66103, USA; Molecular Bio-nanotechnology, Imaging and Therapeutic Research Unit, Veteran Affairs Medical Center, Kansas City, MO 64128, USA
| | - Amit Verma
- Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Swapan K Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Todd Evans
- Department of Surgery, Weill Cornell Medical College of Cornell University, New York, NY 10065, USA.
| |
Collapse
|
46
|
Affiliation(s)
- Pengxiang Huang
- Metabolic Signaling and Disease Program, Sanford-Burnham Medical Research Institute, Orlando, FL 32827, USA
| | - Vikas Chandra
- Metabolic Signaling and Disease Program, Sanford-Burnham Medical Research Institute, Orlando, FL 32827, USA
| | - Fraydoon Rastinejad
- Metabolic Signaling and Disease Program, Sanford-Burnham Medical Research Institute, Orlando, FL 32827, USA
| |
Collapse
|
47
|
Karpinski BA, Maynard TM, Fralish MS, Nuwayhid S, Zohn IE, Moody SA, LaMantia AS. Dysphagia and disrupted cranial nerve development in a mouse model of DiGeorge (22q11) deletion syndrome. Dis Model Mech 2013; 7:245-57. [PMID: 24357327 PMCID: PMC3917245 DOI: 10.1242/dmm.012484] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We assessed feeding-related developmental anomalies in the LgDel mouse model of chromosome 22q11 deletion syndrome (22q11DS), a common developmental disorder that frequently includes perinatal dysphagia – debilitating feeding, swallowing and nutrition difficulties from birth onward – within its phenotypic spectrum. LgDel pups gain significantly less weight during the first postnatal weeks, and have several signs of respiratory infections due to food aspiration. Most 22q11 genes are expressed in anlagen of craniofacial and brainstem regions critical for feeding and swallowing, and diminished expression in LgDel embryos apparently compromises development of these regions. Palate and jaw anomalies indicate divergent oro-facial morphogenesis. Altered expression and patterning of hindbrain transcriptional regulators, especially those related to retinoic acid (RA) signaling, prefigures these disruptions. Subsequently, gene expression, axon growth and sensory ganglion formation in the trigeminal (V), glossopharyngeal (IX) or vagus (X) cranial nerves (CNs) that innervate targets essential for feeding, swallowing and digestion are disrupted. Posterior CN IX and X ganglia anomalies primarily reflect diminished dosage of the 22q11DS candidate gene Tbx1. Genetic modification of RA signaling in LgDel embryos rescues the anterior CN V phenotype and returns expression levels or pattern of RA-sensitive genes to those in wild-type embryos. Thus, diminished 22q11 gene dosage, including but not limited to Tbx1, disrupts oro-facial and CN development by modifying RA-modulated anterior-posterior hindbrain differentiation. These disruptions likely contribute to dysphagia in infants and young children with 22q11DS.
Collapse
Affiliation(s)
- Beverly A Karpinski
- Department of Anatomy and Regenerative Biology, The George Washington University School of Medicine and Health Sciences, Washington DC 20037, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Perreault MC, Glover JC. Glutamatergic reticulospinal neurons in the mouse: developmental origins, axon projections, and functional connectivity. Ann N Y Acad Sci 2013; 1279:80-9. [PMID: 23531005 DOI: 10.1111/nyas.12054] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Subcortical descending glutamatergic neurons, such as reticulospinal (RS) neurons, play decisive roles in the initiation and control of many motor behaviors in mammals. However, little is known about the mechanisms used by RS neurons to control spinal motor networks because most of the neuronal elements involved have not been identified and characterized. In this review, we compare, in the embryonic mouse, the timing of developmental events that lead to the formation of synaptic connections between RS and spinal cord neurons. We then summarize our recent research in the postnatal mouse on the organization of synaptic connections between RS neurons and lumbar axial motoneurons (MNs), hindlimb MNs, and commissural interneurons. Finally, we give a brief account of some of the most recent studies on the intrinsic capabilities for plasticity of the mammalian RS system. The present review should give an updated insight into how functional specificity in RS motor networks emerges.
Collapse
|
49
|
Kayam G, Kohl A, Magen Z, Peretz Y, Weisinger K, Bar A, Novikov O, Brodski C, Sela-Donenfeld D. A novel role for Pax6 in the segmental organization of the hindbrain. Development 2013; 140:2190-202. [PMID: 23578930 DOI: 10.1242/dev.089136] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Complex patterns and networks of genes coordinate rhombomeric identities, hindbrain segmentation and neuronal differentiation and are responsible for later brainstem functions. Pax6 is a highly conserved transcription factor crucial for neuronal development, yet little is known regarding its early roles during hindbrain segmentation. We show that Pax6 expression is highly dynamic in rhombomeres, suggesting an early function in the hindbrain. Utilization of multiple gain- and loss-of-function approaches in chick and mice revealed that loss of Pax6 disrupts the sharp expression borders of Krox20, Kreisler, Hoxa2, Hoxb1 and EphA and leads to their expansion into adjacent territories, whereas excess Pax6 reduces these expression domains. A mutual negative cross-talk between Pax6 and Krox20 allows these genes to be co-expressed in the hindbrain through regulation of the Krox20-repressor gene Nab1 by Pax6. Rhombomere boundaries are also distorted upon Pax6 manipulations, suggesting a mechanism by which Pax6 acts to set hindbrain segmentation. Finally, FGF signaling acts upstream of the Pax6-Krox20 network to regulate Pax6 segmental expression. This study unravels a novel role for Pax6 in the segmental organization of the early hindbrain and provides new evidence for its significance in regional organization along the central nervous system.
Collapse
Affiliation(s)
- Galya Kayam
- Koret School of Veterinary Medicine, The Hebrew University of Jerusalem, The Robert H. Smith Faculty of Agriculture, Food and Environment, 76100 Rehovot, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Lara-Ramírez R, Zieger E, Schubert M. Retinoic acid signaling in spinal cord development. Int J Biochem Cell Biol 2013; 45:1302-13. [PMID: 23579094 DOI: 10.1016/j.biocel.2013.04.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Revised: 03/25/2013] [Accepted: 04/02/2013] [Indexed: 12/13/2022]
Abstract
Retinoic acid (RA) is an important signaling molecule mediating intercellular communication through vertebrate development. Here, we present and discuss recent information on the roles of the RA signaling pathway in spinal cord development. RA is an important player in the patterning and definition of the spinal cord territory from very early stages of development, even before the appearance of the neural plate and further serves a role in the patterning of the spinal cord both along the dorsoventral and anteroposterior axes, particularly in the promotion of neuronal differentiation. It is thus required to establish a variety of neuronal cell types at specific positions of the spinal cord. The main goal of this review is to gather information from vertebrate models, including fish, frogs, chicken and mice, and to put this information in a comparative context in an effort to visualize how the RA pathway was incorporated into the evolving vertebrate spinal cord and to identify mechanisms that are both common and different in the various vertebrate models. In doing so, we try to reconstruct how spinal cord development has been regulated by the RA signaling cascade through vertebrate diversification, highlighting areas which require further studies to obtain a better understanding of the evolutionary events that shaped this structure in the vertebrate lineage.
Collapse
Affiliation(s)
- Ricardo Lara-Ramírez
- Laboratoire de Biologie du Développement de Villefranche-sur-Mer, UMR 7009 - CNRS/UPMC, EvoInSiDe Team, Observatoire Océanologique, 181 Chemin du Lazaret, BP 28, 06230 Villefranche-sur-Mer, France
| | | | | |
Collapse
|