1
|
Leonard CE, McIntosh A, Taneyhill LA. The transcriptional landscape of the developing chick trigeminal ganglion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.20.604400. [PMID: 39211243 PMCID: PMC11361123 DOI: 10.1101/2024.07.20.604400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The trigeminal ganglion is a critical structure in the peripheral nervous system, responsible for transmitting sensations of touch, pain, and temperature from craniofacial regions to the brain. Trigeminal ganglion development depends upon intrinsic cellular programming as well as extrinsic signals exchanged by diverse cell populations. With its complex anatomy and dual cellular origin from cranial placodes and neural crest cells, the trigeminal ganglion offers a rich context for examining diverse biological processes, including cell migration, fate determination, adhesion, and axon guidance. Avian models have, so far, enabled key insights into craniofacial and peripheral nervous system development. Yet, the molecular mechanisms driving trigeminal ganglion formation and subsequent nerve growth remain elusive. In this study, we performed RNA-sequencing at multiple stages of chick trigeminal ganglion development and generated a novel transcriptomic dataset that has been curated to illustrate temporally dynamic gene expression patterns. This publicly available resource identifies major pathways involved in trigeminal gangliogenesis, particularly with respect to the condensation and maturation of placode-derived neurons, thus inviting new lines of research into the essential processes governing trigeminal ganglion development.
Collapse
|
2
|
Deng Y, Lin Y, Chen S, Xiang Y, Chen H, Qi S, Oh HS, Das B, Komazin-Meredith G, Pesola JM, Knipe DM, Coen DM, Pan D. Neuronal miR-9 promotes HSV-1 epigenetic silencing and latency by repressing Oct-1 and Onecut family genes. Nat Commun 2024; 15:1991. [PMID: 38443365 PMCID: PMC10914762 DOI: 10.1038/s41467-024-46057-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 02/07/2024] [Indexed: 03/07/2024] Open
Abstract
Herpes simplex virus 1 (HSV-1) latent infection entails repression of viral lytic genes in neurons. By functional screening using luciferase-expressing HSV-1, we identify ten neuron-specific microRNAs potentially repressing HSV-1 neuronal replication. Transfection of miR-9, the most active candidate from the screen, decreases HSV-1 replication and gene expression in Neuro-2a cells. Ectopic expression of miR-9 from lentivirus or recombinant HSV-1 suppresses HSV-1 replication in male primary mouse neurons in culture and mouse trigeminal ganglia in vivo, and reactivation from latency in the primary neurons. Target prediction and validation identify transcription factors Oct-1, a known co-activator of HSV transcription, and all three Onecut family members as miR-9 targets. Knockdown of ONECUT2 decreases HSV-1 yields in Neuro-2a cells. Overexpression of each ONECUT protein increases HSV-1 replication in Neuro-2a cells, human induced pluripotent stem cell-derived neurons, and primary mouse neurons, and accelerates reactivation from latency in the mouse neurons. Mutagenesis, ChIP-seq, RNA-seq, ChIP-qPCR and ATAC-seq results suggest that ONECUT2 can nonspecifically bind to viral genes via its CUT domain, globally stimulate viral gene transcription, reduce viral heterochromatin and enhance the accessibility of viral chromatin. Thus, neuronal miR-9 promotes viral epigenetic silencing and latency by targeting multiple host transcription factors important for lytic gene activation.
Collapse
Affiliation(s)
- Yue Deng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory for Microbial Biochemistry and Metabolic Engineering, Hangzhou, Zhejiang, China
| | - Yuqi Lin
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory for Microbial Biochemistry and Metabolic Engineering, Hangzhou, Zhejiang, China
| | - Siyu Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory for Microbial Biochemistry and Metabolic Engineering, Hangzhou, Zhejiang, China
| | - Yuhang Xiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory for Microbial Biochemistry and Metabolic Engineering, Hangzhou, Zhejiang, China
| | - Hongjia Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory for Microbial Biochemistry and Metabolic Engineering, Hangzhou, Zhejiang, China
| | - Shuyuan Qi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory for Microbial Biochemistry and Metabolic Engineering, Hangzhou, Zhejiang, China
| | - Hyung Suk Oh
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Biswajit Das
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Gloria Komazin-Meredith
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA, USA
| | - Jean M Pesola
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - David M Knipe
- Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Donald M Coen
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Dongli Pan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Department of Medical Microbiology and Parasitology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Key Laboratory for Microbial Biochemistry and Metabolic Engineering, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Nagel S, Rand U, Pommerenke C, Meyer C. Transcriptional Landscape of CUT-Class Homeobox Genes in Blastic Plasmacytoid Dendritic Cell Neoplasm. Int J Mol Sci 2024; 25:2764. [PMID: 38474011 PMCID: PMC10932245 DOI: 10.3390/ijms25052764] [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: 01/15/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Homeobox genes encode developmental transcription factors regulating tissue-specific differentiation processes and drive cancerogenesis when deregulated. Dendritic cells (DCs) are myeloid immune cells occurring as two types, either conventional or plasmacytoid DCs. Recently, we showed that the expression of NKL-subclass homeobox gene VENTX is restricted to conventional DCs, regulating developmental genes. Here, we identified and investigated homeobox genes specifically expressed in plasmacytoid DCs (pDCs) and derived blastic plasmacytoid dendritic cell neoplasm (BPDCN). We analyzed gene expression data, performed RQ-PCR, protein analyses by Western blot and immuno-cytology, siRNA-mediated knockdown assays and subsequent RNA-sequencing and live-cell imaging. Screening of public gene expression data revealed restricted activity of the CUT-class homeobox gene CUX2 in pDCs. An extended analysis of this homeobox gene class in myelopoiesis showed that additional CUX2 activity was restricted to myeloid progenitors, while BPDCN patients aberrantly expressed ONECUT2, which remained silent in the complete myeloid compartment. ONECUT2 expressing BPDCN cell line CAL-1 served as a model to investigate its regulation and oncogenic activity. The ONECUT2 locus at 18q21 was duplicated and activated by IRF4, AUTS2 and TNF-signaling and repressed by BMP4-, TGFb- and IL13-signalling. Functional analyses of ONECUT2 revealed the inhibition of pDC differentiation and of CDKN1C and CASP1 expression, while SMAD3 and EPAS1 were activated. EPAS1 in turn enhanced survival under hypoxic conditions which thus may support dendritic tumor cells residing in hypoxic skin lesions. Collectively, we revealed physiological and aberrant activities of CUT-class homeobox genes in myelopoiesis including pDCs and in BPDCN, respectively. Our data may aid in the diagnosis of BPDCN patients and reveal novel therapeutic targets for this fatal malignancy.
Collapse
Affiliation(s)
- Stefan Nagel
- Department of Human and Animal Cell Lines, Leibniz-Institute DSMZ, 38124 Braunschweig, Germany
| | | | | | | |
Collapse
|
4
|
Barrett MS, Bauer TC, Li MH, Hegarty DM, Mota CMD, Amaefuna CJ, Ingram SL, Habecker BA, Aicher SA. Ischemia-reperfusion myocardial infarction induces remodeling of left cardiac-projecting stellate ganglia neurons. Am J Physiol Heart Circ Physiol 2024; 326:H166-H179. [PMID: 37947434 PMCID: PMC11213476 DOI: 10.1152/ajpheart.00582.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/23/2023] [Accepted: 11/08/2023] [Indexed: 11/12/2023]
Abstract
Neurons in the stellate ganglion (SG) provide sympathetic innervation to the heart, brown adipose tissue (BAT), and other organs. Sympathetic innervation to the heart becomes hyperactive following myocardial infarction (MI). The impact of MI on the morphology of cardiac sympathetic neurons is not known, but we hypothesized that MI would stimulate increased cell and dendritic tree size in cardiac neurons. In this study, we examined the effects of ischemia-reperfusion MI on sympathetic neurons using dual retrograde tracing methods to allow detailed characterization of cardiac- and BAT-projecting neurons. Different fluorescently conjugated cholera toxin subunit B (CTb) tracers were injected into the pericardium and the interscapular BAT pads, respectively. Experimental animals received a 45-min occlusion of the left anterior descending coronary artery and controls received sham surgery. One week later, hearts were collected for assessment of MI infarct and SGs were collected for morphological or electrophysiological analysis. Cardiac-projecting SG neurons from MI mice had smaller cell bodies and shorter dendritic trees compared with sham animals, specifically on the left side ipsilateral to the MI. BAT-projecting neurons were not altered by MI, demonstrating the subpopulation specificity of the response. The normal size and distribution differences between BAT- and cardiac-projecting stellate ganglion neurons were not altered by MI. Patch-clamp recordings from cardiac-projecting left SG neurons revealed increased spontaneous excitatory postsynaptic currents despite the decrease in cell and dendritic tree size. Thus, increased dendritic tree size does not contribute to the enhanced sympathetic neural activity seen after MI.NEW & NOTEWORTHY Myocardial infarction (MI) causes structural and functional changes specifically in stellate ganglion neurons that project to the heart, but not in cells that project to brown adipose fat tissue.
Collapse
Affiliation(s)
- Madeleine S Barrett
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Temerity C Bauer
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Ming-Hua Li
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Deborah M Hegarty
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Clarissa M D Mota
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Chimezie J Amaefuna
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Susan L Ingram
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Beth A Habecker
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| | - Sue A Aicher
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, United States
| |
Collapse
|
5
|
Tamura M, Ishikawa R, Nakanishi Y, Pascual-Anaya J, Fukui M, Saitou T, Sugahara F, Rijli FM, Kuratani S, Suzuki DG, Murakami Y. Comparative analysis of Hmx expression and the distribution of neuronal somata in the trigeminal ganglion in lamprey and shark: insights into the homology of the trigeminal nerve branches and the evolutionary origin of the vertebrate jaw. ZOOLOGICAL LETTERS 2023; 9:23. [PMID: 38049907 PMCID: PMC10696661 DOI: 10.1186/s40851-023-00222-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/25/2023] [Indexed: 12/06/2023]
Abstract
The evolutionary origin of the jaw remains one of the most enigmatic events in vertebrate evolution. The trigeminal nerve is a key component for understanding jaw evolution, as it plays a crucial role as a sensorimotor interface for the effective manipulation of the jaw. This nerve is also found in the lamprey, an extant jawless vertebrate. The trigeminal nerve has three major branches in both the lamprey and jawed vertebrates. Although each of these branches was classically thought to be homologous between these two taxa, this homology is now in doubt. In the present study, we compared expression patterns of Hmx, a candidate genetic marker of the mandibular nerve (rV3, the third branch of the trigeminal nerve in jawed vertebrates), and the distribution of neuronal somata of trigeminal nerve branches in the trigeminal ganglion in lamprey and shark. We first confirmed the conserved expression pattern of Hmx1 in the shark rV3 neuronal somata, which are distributed in the caudal part of the trigeminal ganglion. By contrast, lamprey Hmx genes showed peculiar expression patterns, with expression in the ventrocaudal part of the trigeminal ganglion similar to Hmx1 expression in jawed vertebrates, which labeled the neuronal somata of the second branch. Based on these results, we propose two alternative hypotheses regarding the homology of the trigeminal nerve branches, providing new insights into the evolutionary origin of the vertebrate jaw.
Collapse
Affiliation(s)
- Motoki Tamura
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572, Ibaraki, Japan
- Graduate School of Science and Engineering, Ehime University, 2-5 Bunkyo-cho, Matsuyama, 790-8577, Japan
| | - Ryota Ishikawa
- Graduate School of Science and Engineering, Ehime University, 2-5 Bunkyo-cho, Matsuyama, 790-8577, Japan
| | - Yuki Nakanishi
- Graduate School of Science and Engineering, Ehime University, 2-5 Bunkyo-cho, Matsuyama, 790-8577, Japan
| | - Juan Pascual-Anaya
- Department of Animal Biology, Faculty of Science, University of Málaga, Campus de Teatinos s/n, Málaga, 29071, Spain
| | - Makiko Fukui
- Graduate School of Science and Engineering, Ehime University, 2-5 Bunkyo-cho, Matsuyama, 790-8577, Japan
| | - Takashi Saitou
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Toon, 791-0295, Japan
| | - Fumiaki Sugahara
- Division of Biology, Hyogo Medical University, Nishinomiya, 663-8501, Hyogo, Japan
- Evolutionary Morphology Laboratory, RIKEN Cluster for Pioneering Research (CPR), 2-2-3 Minatojima-minami, Chuo-ku, Kobe, 650-0047, Japan
| | - Filippo M Rijli
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel, 4058, Switzerland
- University of Basel, Basel, Switzerland
| | - Shigeru Kuratani
- Evolutionary Morphology Laboratory, RIKEN Cluster for Pioneering Research (CPR), 2-2-3 Minatojima-minami, Chuo-ku, Kobe, 650-0047, Japan
| | - Daichi G Suzuki
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8572, Ibaraki, Japan
| | - Yasunori Murakami
- Graduate School of Science and Engineering, Ehime University, 2-5 Bunkyo-cho, Matsuyama, 790-8577, Japan.
| |
Collapse
|
6
|
Koutsioumpa C, Santiago C, Jacobs K, Lehnert BP, Barrera V, Hutchinson JN, Schmelyun D, Lehoczky JA, Paul DL, Ginty DD. Skin-type-dependent development of murine mechanosensory neurons. Dev Cell 2023; 58:2032-2047.e6. [PMID: 37607547 PMCID: PMC10615785 DOI: 10.1016/j.devcel.2023.07.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/26/2023] [Accepted: 07/27/2023] [Indexed: 08/24/2023]
Abstract
Mechanosensory neurons innervating the skin underlie our sense of touch. Fast-conducting, rapidly adapting mechanoreceptors innervating glabrous (non-hairy) skin form Meissner corpuscles, while in hairy skin, they associate with hair follicles, forming longitudinal lanceolate endings. How mechanoreceptors develop axonal endings appropriate for their skin targets is unknown. We report that mechanoreceptor morphologies across different skin regions are indistinguishable during early development but diverge post-natally, in parallel with skin maturation. Neurons terminating along the glabrous and hairy skin border exhibit hybrid morphologies, forming both Meissner corpuscles and lanceolate endings. Additionally, molecular profiles of neonatal glabrous and hairy skin-innervating neurons largely overlap. In mouse mutants with ectopic glabrous skin, mechanosensory neurons form end-organs appropriate for the altered skin type. Finally, BMP5 and BMP7 are enriched in glabrous skin, and signaling through type I bone morphogenetic protein (BMP) receptors in neurons is critical for Meissner corpuscle morphology. Thus, mechanoreceptor morphogenesis is flexibly instructed by target tissues.
Collapse
Affiliation(s)
- Charalampia Koutsioumpa
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Celine Santiago
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Kiani Jacobs
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Brendan P Lehnert
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Victor Barrera
- Bioinformatics Core, Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - John N Hutchinson
- Bioinformatics Core, Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Dhane Schmelyun
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Jessica A Lehoczky
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - David L Paul
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - David D Ginty
- Department of Neurobiology, Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Leyva-Díaz E. CUT homeobox genes: transcriptional regulation of neuronal specification and beyond. Front Cell Neurosci 2023; 17:1233830. [PMID: 37744879 PMCID: PMC10515288 DOI: 10.3389/fncel.2023.1233830] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
CUT homeobox genes represent a captivating gene class fulfilling critical functions in the development and maintenance of multiple cell types across a wide range of organisms. They belong to the larger group of homeobox genes, which encode transcription factors responsible for regulating gene expression patterns during development. CUT homeobox genes exhibit two distinct and conserved DNA binding domains, a homeodomain accompanied by one or more CUT domains. Numerous studies have shown the involvement of CUT homeobox genes in diverse developmental processes such as body axis formation, organogenesis, tissue patterning and neuronal specification. They govern these processes by exerting control over gene expression through their transcriptional regulatory activities, which they accomplish by a combination of classic and unconventional interactions with the DNA. Intriguingly, apart from their roles as transcriptional regulators, they also serve as accessory factors in DNA repair pathways through protein-protein interactions. They are highly conserved across species, highlighting their fundamental importance in developmental biology. Remarkably, evolutionary analysis has revealed that CUT homeobox genes have experienced an extraordinary degree of rearrangements and diversification compared to other classes of homeobox genes, including the emergence of a novel gene family in vertebrates. Investigating the functions and regulatory networks of CUT homeobox genes provides significant understanding into the molecular mechanisms underlying embryonic development and tissue homeostasis. Furthermore, aberrant expression or mutations in CUT homeobox genes have been associated with various human diseases, highlighting their relevance beyond developmental processes. This review will overview the well known roles of CUT homeobox genes in nervous system development, as well as their functions in other tissues across phylogeny.
Collapse
|
8
|
Ueta Y, Miyata M. Functional and structural synaptic remodeling mechanisms underlying somatotopic organization and reorganization in the thalamus. Neurosci Biobehav Rev 2023; 152:105332. [PMID: 37524138 DOI: 10.1016/j.neubiorev.2023.105332] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/09/2023] [Accepted: 07/27/2023] [Indexed: 08/02/2023]
Abstract
The somatosensory system organizes the topographic representation of body maps, termed somatotopy, at all levels of an ascending hierarchy. Postnatal maturation of somatotopy establishes optimal somatosensation, whereas deafferentation in adults reorganizes somatotopy, which underlies pathological somatosensation, such as phantom pain and complex regional pain syndrome. Here, we focus on the mouse whisker somatosensory thalamus to study how sensory experience shapes the fine topography of afferent connectivity during the critical period and what mechanisms remodel it and drive a large-scale somatotopic reorganization after peripheral nerve injury. We will review our findings that, following peripheral nerve injury in adults, lemniscal afferent synapses onto thalamic neurons are remodeled back to immature configuration, as if the critical period reopens. The remodeling process is initiated with local activation of microglia in the brainstem somatosensory nucleus downstream to injured nerves and heterosynaptically controlled by input from GABAergic and cortical neurons to thalamic neurons. These fruits of thalamic studies complement well-studied cortical mechanisms of somatotopic organization and reorganization and unveil potential intervention points in treating pathological somatosensation.
Collapse
Affiliation(s)
- Yoshifumi Ueta
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women's Medical University, Tokyo 162-8666, Japan
| | - Mariko Miyata
- Division of Neurophysiology, Department of Physiology, School of Medicine, Tokyo Women's Medical University, Tokyo 162-8666, Japan.
| |
Collapse
|
9
|
Ostler JB, Jones C. The Bovine Herpesvirus 1 Latency-Reactivation Cycle, a Chronic Problem in the Cattle Industry. Viruses 2023; 15:552. [PMID: 36851767 PMCID: PMC9966457 DOI: 10.3390/v15020552] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/19/2023] Open
Abstract
Bovine alphaherpesvirus 1 (BoHV-1) is a persistent and recurring disease that affects cattle worldwide. It is a major contributor to bovine respiratory disease and reproductive failure in the US. A major complication of BoHV-1 arises from the lifelong latent infection established in the sensory ganglia of the peripheral nervous system following acute infection. Lifelong latency is marked by periodic reactivation from latency that leads to virus transmission and transient immunosuppression. Physiological and environmental stress, along with hormone fluctuations, can drive virus reactivation from latency, allowing the virus to spread rapidly. This review discusses the mechanisms of the latency/reactivation cycle, with particular emphasis on how different hormones directly regulate BoHV-1 gene expression and productive infection. Glucocorticoids, including the synthetic corticosteroid dexamethasone, are major effectors of the stress response. Stress directly regulates BoHV-1 gene expression through multiple pathways, including β-catenin dependent Wnt signaling, and the glucocorticoid receptor. Related type 1 nuclear hormone receptors, the androgen and progesterone receptors, also drive BoHV-1 gene expression and productive infection. These receptors form feed-forward transcription loops with the stress-induced Krüppel-like transcription factors KLF4 and KLF15. Understanding these molecular pathways is critical for developing novel therapeutics designed to block reactivation and reduce virus spread and disease.
Collapse
Affiliation(s)
| | - Clinton Jones
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
10
|
Ogawa C, Mikawa S, Li S, Hayashi Y, Masumoto K, Sato K. BMP10 expression in the adult rat central nervous system. J Chem Neuroanat 2022; 121:102084. [PMID: 35182716 DOI: 10.1016/j.jchemneu.2022.102084] [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: 09/02/2021] [Revised: 02/09/2022] [Accepted: 02/14/2022] [Indexed: 11/19/2022]
Abstract
Bone morphogenetic protein 10 (BMP10), is a member of the transforming growth factor β (TGFβ) superfamily. Although BMP10 plays pivotal roles during development, including vascular development and cardiogenesis, little information is available for BMP10 expression in the central nervous system (CNS). We, thus, investigated BMP10 expression in the adult rat CNS using immunohistochemistry. BMP10 was intensely expressed in most neurons and their axons. Furthermore, we found that astrocytes and ependymal cells also express BMP10 protein. These data indicate that BMP10 is widely expressed throughout the adult CNS, and this abundant expression strongly supports the idea that BMP10 also plays important roles in the adult CNS.
Collapse
Affiliation(s)
- Chikara Ogawa
- Department of Dentistry and Oral and Maxillofacial Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan; Department of Organ & Tissue Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Sumiko Mikawa
- Department of Organ & Tissue Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Shuo Li
- Department of Organ & Tissue Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan; Department of Orthopedic Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Yutaro Hayashi
- Department of Dentistry and Oral and Maxillofacial Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Kazuma Masumoto
- Department of Dentistry and Oral and Maxillofacial Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Kohji Sato
- Department of Organ & Tissue Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan.
| |
Collapse
|
11
|
The cellular and molecular basis of somatosensory neuron development. Neuron 2021; 109:3736-3757. [PMID: 34592169 DOI: 10.1016/j.neuron.2021.09.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 11/23/2022]
Abstract
Primary somatosensory neurons convey salient information about our external environment and internal state to the CNS, allowing us to detect, perceive, and react to a wide range of innocuous and noxious stimuli. Pseudo-unipolar in shape, and among the largest (longest) cells of most mammals, dorsal root ganglia (DRG) somatosensory neurons have peripheral axons that extend into skin, muscle, viscera, or bone and central axons that innervate the spinal cord and brainstem, where they synaptically engage the central somatosensory circuitry. Here, we review the diversity of mammalian DRG neuron subtypes and the intrinsic and extrinsic mechanisms that control their development. We describe classical and contemporary advances that frame our understanding of DRG neurogenesis, transcriptional specification of DRG neurons, and the establishment of morphological, physiological, and synaptic diversification across somatosensory neuron subtypes.
Collapse
|
12
|
Maynard TM, Horvath A, Bernot JP, Karpinski BA, Tavares ALP, Shah A, Zheng Q, Spurr L, Olender J, Moody SA, Fraser CM, LaMantia AS, Lee NH. Transcriptional dysregulation in developing trigeminal sensory neurons in the LgDel mouse model of DiGeorge 22q11.2 deletion syndrome. Hum Mol Genet 2021; 29:1002-1017. [PMID: 32047912 PMCID: PMC7158380 DOI: 10.1093/hmg/ddaa024] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 01/12/2020] [Accepted: 02/04/2020] [Indexed: 12/13/2022] Open
Abstract
LgDel mice, which model the heterozygous deletion of genes at human chromosome 22q11.2 associated with DiGeorge/22q11.2 deletion syndrome (22q11DS), have cranial nerve and craniofacial dysfunction as well as disrupted suckling, feeding and swallowing, similar to key 22q11DS phenotypes. Divergent trigeminal nerve (CN V) differentiation and altered trigeminal ganglion (CNgV) cellular composition prefigure these disruptions in LgDel embryos. We therefore asked whether a distinct transcriptional state in a specific population of early differentiating LgDel cranial sensory neurons, those in CNgV, a major source of innervation for appropriate oropharyngeal function, underlies this departure from typical development. LgDel versus wild-type (WT) CNgV transcriptomes differ significantly at E10.5 just after the ganglion has coalesced. Some changes parallel altered proportions of cranial placode versus cranial neural crest-derived CNgV cells. Others are consistent with a shift in anterior-posterior patterning associated with divergent LgDel cranial nerve differentiation. The most robust quantitative distinction, however, is statistically verifiable increased variability of expression levels for most of the over 17 000 genes expressed in common in LgDel versus WT CNgV. Thus, quantitative expression changes of functionally relevant genes and increased stochastic variation across the entire CNgV transcriptome at the onset of CN V differentiation prefigure subsequent disruption of cranial nerve differentiation and oropharyngeal function in LgDel mice.
Collapse
Affiliation(s)
- Thomas M Maynard
- Fralin Biomedical Research Institute, Virginia Tech-Carilion School of Medicine, Roanoke, VA, 24016 USA.,Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Anelia Horvath
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA.,McCormick Genomics and Proteomics Center, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - James P Bernot
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Beverly A Karpinski
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Andre L P Tavares
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Ankita Shah
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Qianqian Zheng
- Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Liam Spurr
- Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Jacqueline Olender
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Sally A Moody
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| | - Claire M Fraser
- Institute for Genome Sciences, University of Maryland, Baltimore, Baltimore, MD, USA
| | - Anthony-S LaMantia
- Fralin Biomedical Research Institute, Virginia Tech-Carilion School of Medicine, Roanoke, VA, 24016 USA.,Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Anatomy and Cell Biology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA.,Department of Biological Sciences, College of Science, Virginia Tech, Blacksburg VA, 24061, USA.,Department of Pediatrics, Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
| | - Norman H Lee
- Institute for Neuroscience, The George Washington University, Washington, DC 20037, USA.,Department of Pharmacology and Physiology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
| |
Collapse
|
13
|
Ogawa C, Mikawa S, Hayashi Y, Masumoto K, Katou F, Sato K. BMP9 expression in the adult rat brain. J Chem Neuroanat 2021; 113:101933. [PMID: 33582251 DOI: 10.1016/j.jchemneu.2021.101933] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 01/29/2023]
Abstract
Bone morphogenetic protein 9 (BMP9), also known as growth differentiation factor 2 (GDF2), is a member of the transforming growth factor β (TGF β) superfamily. Although BMP9 plays pivotal roles during development, including angiogenesis, hematopoiesis, hepatogenesis, osteogenesis, and glucose metabolism, little information is available for BMP9 expression in the central nervous system (CNS). We, thus, investigated BMP9 expression in the adult rat CNS using immunohistochemistry. BMP9 was intensely expressed in most neurons and their axons. Furthermore, we found that oligodendrocytes and ependymal cells also express BMP9 protein. These data indicate that BMP9 is widely expressed throughout the adult CNS, and this abundant expression strongly supports the idea that BMP9 also plays important roles in the adult brain.
Collapse
Affiliation(s)
- Chikara Ogawa
- Department of Dentistry and Oral and Maxillofacial Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan; Department of Organ & Tissue Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Sumiko Mikawa
- Department of Organ & Tissue Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Yutaro Hayashi
- Department of Dentistry and Oral and Maxillofacial Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Kazuma Masumoto
- Department of Dentistry and Oral and Maxillofacial Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Fuminori Katou
- Department of Dentistry and Oral and Maxillofacial Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Kohji Sato
- Department of Organ & Tissue Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan.
| |
Collapse
|
14
|
Berndt AJ, Othonos KM, Lian T, Flibotte S, Miao M, Bhuiyan SA, Cho RY, Fong JS, Hur SA, Pavlidis P, Allan DW. A low affinity cis-regulatory BMP response element restricts target gene activation to subsets of Drosophila neurons. eLife 2020; 9:59650. [PMID: 33124981 PMCID: PMC7669266 DOI: 10.7554/elife.59650] [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: 06/03/2020] [Accepted: 10/29/2020] [Indexed: 11/19/2022] Open
Abstract
Retrograde BMP signaling and canonical pMad/Medea-mediated transcription regulate diverse target genes across subsets of Drosophila efferent neurons, to differentiate neuropeptidergic neurons and promote motor neuron terminal maturation. How a common BMP signal regulates diverse target genes across many neuronal subsets remains largely unresolved, although available evidence implicates subset-specific transcription factor codes rather than differences in BMP signaling. Here we examine the cis-regulatory mechanisms restricting BMP-induced FMRFa neuropeptide expression to Tv4-neurons. We find that pMad/Medea bind at an atypical, low affinity motif in the FMRFa enhancer. Converting this motif to high affinity caused ectopic enhancer activity and eliminated Tv4-neuron expression. In silico searches identified additional motif instances functional in other efferent neurons, implicating broader functions for this motif in BMP-dependent enhancer activity. Thus, differential interpretation of a common BMP signal, conferred by low affinity pMad/Medea binding motifs, can contribute to the specification of BMP target genes in efferent neuron subsets.
Collapse
Affiliation(s)
- Anthony Je Berndt
- Department of Food & Fuel for the 21st Century, University of California San Diego, San Diego, United States
| | - Katerina M Othonos
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Tianshun Lian
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Stephane Flibotte
- UBC/LSI Bioinformatics Facility, University of British Columbia, Vancouver, Canada
| | - Mo Miao
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | | | - Raymond Y Cho
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Justin S Fong
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Seo Am Hur
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Paul Pavlidis
- Department of Psychiatry, University of British Columbia, Vancouver, Canada
| | - Douglas W Allan
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| |
Collapse
|
15
|
Naik AS, Lin JM, Taroc EZM, Katreddi RR, Frias JA, Lemus AA, Sammons MA, Forni PE. Smad4-dependent morphogenic signals control the maturation and axonal targeting of basal vomeronasal sensory neurons to the accessory olfactory bulb. Development 2020; 147:147/8/dev184036. [PMID: 32341026 PMCID: PMC7197725 DOI: 10.1242/dev.184036] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 03/10/2020] [Indexed: 12/31/2022]
Abstract
The vomeronasal organ (VNO) contains two main types of vomeronasal sensory neurons (VSNs) that express distinct vomeronasal receptor (VR) genes and localize to specific regions of the neuroepithelium. Morphogenic signals are crucial in defining neuronal identity and network formation; however, if and what signals control maturation and homeostasis of VSNs is largely unexplored. Here, we found transforming growth factor β (TGFβ) and bone morphogenetic protein (BMP) signal transduction in postnatal mice, with BMP signaling being restricted to basal VSNs and at the marginal zones of the VNO: the site of neurogenesis. Using different Smad4 conditional knockout mouse models, we disrupted canonical TGFβ/BMP signaling in either maturing basal VSNs (bVSNs) or all mature VSNs. Smad4 loss of function in immature bVSNs compromises dendritic knob formation, pheromone induced activation, correct glomeruli formation in the accessory olfactory bulb (AOB) and survival. However, Smad4 loss of function in all mature VSNs only compromises correct glomeruli formation in the posterior AOB. Our results indicate that Smad4-mediated signaling drives the functional maturation and connectivity of basal VSNs. Summary: Genetic disruption of TGFβ/BMP signaling in maturing basal vomeronasal sensory neurons (VSNs) or in all mature VSNs indicates that Smad4 signaling drives maturation and connectivity of basal VSNs.
Collapse
Affiliation(s)
- Ankana S Naik
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Jennifer M Lin
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Ed Zandro M Taroc
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Raghu R Katreddi
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Jesus A Frias
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Alex A Lemus
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Morgan A Sammons
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| | - Paolo E Forni
- Department of Biological Sciences; The RNA Institute; University at Albany, State University of New York, Albany, NY 12222, USA
| |
Collapse
|
16
|
Seo EH, Kim HJ, Kim JH, Lim B, Park JL, Kim SY, Lee SI, Jeong HY, Song KS, Kim YS. ONECUT2 upregulation is associated with CpG hypomethylation at promoter-proximal DNA in gastric cancer and triggers ACSL5. Int J Cancer 2020; 146:3354-3368. [PMID: 32129880 PMCID: PMC7217064 DOI: 10.1002/ijc.32946] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 02/16/2020] [Accepted: 02/19/2020] [Indexed: 12/14/2022]
Abstract
Many studies have focused on global hypomethylation or hypermethylation of tumor suppressor genes, but less is known about the impact of promoter hypomethylation of oncogenes. We previously showed that promoter methylation may gradually increase or decrease during the transition from gastric mucosa (GM) to intestinal metaplasia (IM) to gastric cancer (GC). In our study, we focused on regional CpG hypomethylation of the promoter‐proximal DNA of the transcription factor ONECUT2 (OC2) in IM and GC cells. We validated the hypomethylation of promoter‐proximal DNA of OC2 in 160 primary GCs, in which methylation level correlated negatively with OC2 mRNA level. IM and GC cells stained positively for OC2, whereas GM cells did not. Stable transfection of OC2 in GC cells promoted colony formation, cell migration, invasion and proliferation. Moreover, OC2 knockdown with a short hairpin RNA suppressed tumorigenesis in nude mice. In addition, chromatin immunoprecipitation coupled with DNA sequencing and RNA‐seq analyses revealed that OC2 triggered ACSL5, which is strongly expressed in IM of the stomach but not in GM, indicating that OC2 and ACSL5 are early‐stage biomarkers for GC. We also observed a high correlation between the levels of OC2 and ACSL5 mRNAs in the GENT database These results suggest that epigenetic alteration of OC2 upregulates its expression, which then activates ACSL5; thus, OC2 is induced in IM by epigenetic alteration and triggers ACSL5 expression, and thus OC2 and ACSL5 may cooperatively promote intestinal differentiation and GC progression. What's new? DNA hypomethylation can promote cancer development through activation of genes with oncogenic potential. Here, the authors found that CpGs in the promoter‐proximal DNA of ONECUT2 were hypomethylated in intestinal metaplasia and gastric cancers, and that hypomethylation was associated with ONECUT2 upregulation. Functional analysis demonstrated that ONECUT2 has oncogenic potential and could activate ACSL5, which is also expressed in intestinal metaplasia, suggesting that ONECUT2 and ACSL5 may cooperate to promote intestinal differentiation or development of gastric cancer. Taken together, the findings suggest that ONECUT2 and its downstream target ACSL5 could be used to develop early detection biomarkers and prevent gastric carcinogenesis.
Collapse
Affiliation(s)
- Eun-Hye Seo
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Hee-Jin Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Jong-Hwan Kim
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Byungho Lim
- Division of Drug Discovery Research, Research Center for Drug Discovery Technology, Korea Research Institute of Chemical Technology, Daejeon, Republic of Korea
| | - Jong-Lyul Park
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Seon-Young Kim
- Department of Functional Genomics, Korea University of Science and Technology, Daejeon, Republic of Korea.,Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Sang-Il Lee
- Department of Surgery, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Hyun-Yong Jeong
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Kyu-Sang Song
- Department of Pathology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Yong-Sung Kim
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.,Department of Functional Genomics, Korea University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
17
|
CBP and p300 coactivators contribute to the maintenance of Isl1 expression by the Onecut transcription factors in embryonic spinal motor neurons. Mol Cell Neurosci 2019; 101:103411. [PMID: 31648029 DOI: 10.1016/j.mcn.2019.103411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 08/14/2019] [Accepted: 09/11/2019] [Indexed: 11/24/2022] Open
Abstract
Onecut transcription factors are required to maintain Islet1 (Isl1) expression in developing spinal motor neurons (MNs), and this process is critical for proper MN differentiation. However, the mechanisms whereby OC stimulate Isl1 expression remain unknown. CREB-binding protein (CBP) and p300 paralogs are transcriptional coactivators that interact with OC proteins in hepatic cells. In the embryonic spinal cord, CBP and p300 play key roles in neurogenesis and MN differentiation. Here, using chromatin immunoprecipitation and in ovo electroporation in chicken spinal cord, we provide evidence that CBP and p300 contribute to the regulation of Isl1 expression by the OC factors in embryonic spinal MNs. CBP and p300 are detected on the CREST2 enhancer of Isl1 where OC factors are also bound. Inhibition of CBP and p300 activity inhibits activation of the CREST2 enhancer and prevents the stimulation of Isl1 expression by the OC factors. These observations suggest that CBP and p300 coactivators cooperate with OC factors to maintain Isl1 expression in postmitotic MNs.
Collapse
|
18
|
Feigin CY, Newton AH, Pask AJ. Widespread cis-regulatory convergence between the extinct Tasmanian tiger and gray wolf. Genome Res 2019; 29:1648-1658. [PMID: 31533979 PMCID: PMC6771401 DOI: 10.1101/gr.244251.118] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 08/19/2019] [Indexed: 12/18/2022]
Abstract
The extinct marsupial Tasmanian tiger, or thylacine, and the eutherian gray wolf are among the most widely recognized examples of convergent evolution in mammals. Despite being distantly related, these large predators independently evolved extremely similar craniofacial morphologies, and evidence suggests that they filled similar ecological niches. Previous analyses revealed little evidence of adaptive convergence between their protein-coding genes. Thus, the genetic basis of their convergence is still unclear. Here, we identified candidate craniofacial cis-regulatory elements across vertebrates and compared their evolutionary rates in the thylacine and wolf, revealing abundant signatures of convergent positive selection. Craniofacial thylacine-wolf accelerated regions were enriched near genes involved in TGF beta (TGFB) and BMP signaling, both of which are key morphological signaling pathways with critical roles in establishing the identities and boundaries between craniofacial tissues. Similarly, enhancers of genes involved in craniofacial nerve development showed convergent selection and involvement in these pathways. Taken together, these results suggest that adaptation in cis-regulators of TGF beta and BMP signaling may provide a mechanism to explain the coevolution of developmentally and functionally integrated craniofacial structures in these species. We also found that despite major structural differences in marsupial and eutherian brains, accelerated regions in both species were common near genes with roles in brain development. Our findings support the hypothesis that, relative to protein-coding genes, positive selection on cis-regulatory elements is likely to be an essential driver of adaptive convergent evolution and may underpin thylacine-wolf phenotypic similarities.
Collapse
Affiliation(s)
- Charles Y Feigin
- School of BioSciences, The University of Melbourne, Parkville, Victoria 3010, Australia.,Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Axel H Newton
- School of BioSciences, The University of Melbourne, Parkville, Victoria 3010, Australia.,Museums Victoria, Melbourne, Victoria 3053, Australia
| | - Andrew J Pask
- School of BioSciences, The University of Melbourne, Parkville, Victoria 3010, Australia.,Museums Victoria, Melbourne, Victoria 3053, Australia
| |
Collapse
|
19
|
Vuilleumier R, Lian T, Flibotte S, Khan ZN, Fuchs A, Pyrowolakis G, Allan DW. Retrograde BMP signaling activates neuronal gene expression through widespread deployment of a conserved BMP-responsive cis-regulatory activation element. Nucleic Acids Res 2019; 47:679-699. [PMID: 30476189 PMCID: PMC6344883 DOI: 10.1093/nar/gky1135] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 10/25/2018] [Indexed: 12/29/2022] Open
Abstract
Retrograde Bone Morphogenetic Protein (BMP) signaling in neurons is essential for the differentiation and synaptic function of many neuronal subtypes. BMP signaling regulates these processes via Smad transcription factor activity, yet the scope and nature of Smad-dependent gene regulation in neurons are mostly unknown. Here, we applied a computational approach to predict Smad-binding cis-regulatory BMP-Activating Elements (BMP-AEs) in Drosophila, followed by transgenic in vivo reporter analysis to test their neuronal subtype enhancer activity in the larval central nervous system (CNS). We identified 34 BMP-AE-containing genomic fragments that are responsive to BMP signaling in neurons, and showed that the embedded BMP-AEs are required for this activity. RNA-seq analysis identified BMP-responsive genes in the CNS and revealed that BMP-AEs selectively enrich near BMP-activated genes. These data suggest that functional BMP-AEs control nearby BMP-activated genes, which we validated experimentally. Finally, we demonstrated that the BMP-AE motif mediates a conserved Smad-responsive function in the Drosophila and vertebrate CNS. Our results provide evidence that BMP signaling controls neuronal function by directly coordinating the expression of a battery of genes through widespread deployment of a conserved Smad-responsive cis-regulatory motif.
Collapse
Affiliation(s)
- Robin Vuilleumier
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tianshun Lian
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephane Flibotte
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zaynah N Khan
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alisa Fuchs
- BIOSS, Centre for Biological Signaling Studies and Institute for Biology I, Faculty of Biology, Albert-Ludwigs University of Freiburg, Freiburg, Germany.,Max-Planck Institute for Molecular Genetics, Berlin, Germany
| | - George Pyrowolakis
- BIOSS, Centre for Biological Signaling Studies and Institute for Biology I, Faculty of Biology, Albert-Ludwigs University of Freiburg, Freiburg, Germany
| | - Douglas W Allan
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
20
|
Zhuang M, Li X, Zhu J, Zhang J, Niu F, Liang F, Chen M, Li D, Han P, Ji SJ. The m6A reader YTHDF1 regulates axon guidance through translational control of Robo3.1 expression. Nucleic Acids Res 2019; 47:4765-4777. [PMID: 30843071 PMCID: PMC6511866 DOI: 10.1093/nar/gkz157] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 02/25/2019] [Accepted: 02/27/2019] [Indexed: 12/02/2022] Open
Abstract
N 6-Methyladenosine (m6A) is a dynamic mRNA modification which regulates protein expression in various posttranscriptional levels. Functional studies of m6A in nervous system have focused on its writers and erasers so far, whether and how m6A readers mediate m6A functions through recognizing and binding their target mRNA remains poorly understood. Here, we find that the expression of axon guidance receptor Robo3.1 which plays important roles in midline crossing of spinal commissural axons is regulated precisely at translational level. The m6A reader YTHDF1 binds to and positively regulates translation of m6A-modified Robo3.1 mRNA. Either mutation of m6A sites in Robo3.1 mRNA or YTHDF1 knockdown or knockout leads to dramatic reduction of Robo3.1 protein without affecting Robo3.1 mRNA level. Specific ablation of Ythdf1 in spinal commissural neurons results in pre-crossing axon guidance defects. Our findings identify a mechanism that YTHDF1-mediated translation of m6A-modified Robo3.1 mRNA controls pre-crossing axon guidance in spinal cord.
Collapse
Affiliation(s)
- Mengru Zhuang
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- SUSTech-HKUST Joint PhD Program, Division of Life Science, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Xinbei Li
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Junda Zhu
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jian Zhang
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Fugui Niu
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- SUSTech-HIT Joint Graduate Program, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Fanghao Liang
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- SUSTech-HIT Joint Graduate Program, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Mengxian Chen
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Duo Li
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Peng Han
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Sheng-Jian Ji
- Department of Biology, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Institute of Neuroscience, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
21
|
Trejo JL. Cranial Nerves: Mind Your Head. Anat Rec (Hoboken) 2019; 302:374-377. [DOI: 10.1002/ar.24071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/20/2018] [Accepted: 01/28/2019] [Indexed: 12/20/2022]
Affiliation(s)
- José Luis Trejo
- Department of Translational Neuroscience, CSIC; Cajal Institute; Madrid Spain
| |
Collapse
|
22
|
Kitazawa T, Rijli FM. Barrelette map formation in the prenatal mouse brainstem. Curr Opin Neurobiol 2018; 53:210-219. [PMID: 30342228 DOI: 10.1016/j.conb.2018.09.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/03/2018] [Accepted: 09/24/2018] [Indexed: 12/30/2022]
Abstract
The rodent whiskers are topographically mapped in brainstem sensory nuclei as neuronal modules known as barrelettes. Little is known about how the facial whisker pattern is copied into a brainstem barrelette topographic pattern, which serves as a template for the establishment of thalamic barreloid and, in turn, cortical barrel maps, and how precisely is the whisker pattern mapped in the brainstem during prenatal development. Here, we review recent insights advancing our understanding of the intrinsic and extrinsic patterning mechanisms contributing to establish topographical equivalence between the facial whisker pattern and the mouse brainstem during prenatal development and their relative importance.
Collapse
Affiliation(s)
- Taro Kitazawa
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4051 Basel, Switzerland
| | - Filippo M Rijli
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4051 Basel, Switzerland; University of Basel, 4003 Basel, Switzerland.
| |
Collapse
|
23
|
BMP signaling downstream of the Highwire E3 ligase sensitizes nociceptors. PLoS Genet 2018; 14:e1007464. [PMID: 30001326 PMCID: PMC6042685 DOI: 10.1371/journal.pgen.1007464] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/01/2018] [Indexed: 01/18/2023] Open
Abstract
A comprehensive understanding of the molecular machinery important for nociception is essential to improving the treatment of pain. Here, we show that the BMP signaling pathway regulates nociception downstream of the E3 ubiquitin ligase highwire (hiw). hiw loss of function in nociceptors caused antagonistic and pleiotropic phenotypes with simultaneous insensitivity to noxious heat but sensitized responses to optogenetic activation of nociceptors. Thus, hiw functions to both positively and negatively regulate nociceptors. We find that a sensory reception-independent sensitization pathway was associated with BMP signaling. BMP signaling in nociceptors was up-regulated in hiw mutants, and nociceptor-specific expression of hiw rescued all nociception phenotypes including the increased BMP signaling. Blocking the transcriptional output of the BMP pathway with dominant negative Mad suppressed nociceptive hypersensitivity that was induced by interfering with hiw. The up-regulated BMP signaling phenotype in hiw genetic mutants could not be suppressed by mutation in wallenda suggesting that hiw regulates BMP in nociceptors via a wallenda independent pathway. In a newly established Ca2+ imaging preparation, we observed that up-regulated BMP signaling caused a significantly enhanced Ca2+ signal in the axon terminals of nociceptors that were stimulated by noxious heat. This response likely accounts for the nociceptive hypersensitivity induced by elevated BMP signaling in nociceptors. Finally, we showed that 24-hour activation of BMP signaling in nociceptors was sufficient to sensitize nociceptive responses to optogenetically-triggered nociceptor activation without altering nociceptor morphology. Overall, this study demonstrates the previously unrevealed roles of the Hiw-BMP pathway in the regulation of nociception and provides the first direct evidence that up-regulated BMP signaling physiologically sensitizes responses of nociceptors and nociception behaviors. Although pain is a universally experienced sensation that has a significant impact on human lives and society, the molecular mechanisms of pain remain poorly understood. Elucidating these mechanisms is particularly important to gaining insight into the clinical development of currently incurable chronic pain diseases. Taking an advantage of the powerful genetic model organism Drosophila melanogaster (fruit flies), we unveil the Highwire-BMP signaling pathway as a novel molecular pathway that regulates the sensitivity of nociceptive sensory neurons. Highwire and the molecular components of the BMP signaling pathway are known to be widely conserved among animal phyla, from nematode worms to humans. Since abnormal sensitivity of nociceptive sensory neurons can play a critical role in the development of chronic pain conditions, a deeper understanding of the regulation of nociceptor sensitivity has the potential to advance effective therapeutic strategies to treat difficult pain conditions.
Collapse
|
24
|
Iwasato T, Erzurumlu RS. Development of tactile sensory circuits in the CNS. Curr Opin Neurobiol 2018; 53:66-75. [PMID: 29908482 DOI: 10.1016/j.conb.2018.06.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/30/2018] [Accepted: 06/01/2018] [Indexed: 12/13/2022]
Abstract
Molecular identification of neuronal types and genetic and imaging approaches to characterize their properties reveal morphological, physiological and dynamic aspects of sensory circuit development. Here we focus on the mouse tactile sensory circuitry, with particular emphasis on the main trigeminal pathway that connects the whiskers, the major tactile organ in rodents, to the neocortex. At each level of this pathway, neurogenesis, axonal elongation, pathfinding, target recognition and circuit reorganization including dendritic refinement of cortical layer 4 neurons occur contemporaneously and a multitude of molecular signals are used in differing combinations. We highlight recent advances in development of tactile circuitry and note gaps in our understanding.
Collapse
Affiliation(s)
- Takuji Iwasato
- National Institute of Genetics, Mishima, Japan; Department of Genetics, SOKENDAI, Mishima, Japan
| | - Reha S Erzurumlu
- Department of Anatomy and Neurobiology, School of Medicine, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
25
|
Workman A, Zhu L, Keel BN, Smith TPL, Jones C. The Wnt Signaling Pathway Is Differentially Expressed during the Bovine Herpesvirus 1 Latency-Reactivation Cycle: Evidence That Two Protein Kinases Associated with Neuronal Survival, Akt3 and BMPR2, Are Expressed at Higher Levels during Latency. J Virol 2018; 92:e01937-17. [PMID: 29321317 PMCID: PMC5972910 DOI: 10.1128/jvi.01937-17] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 01/04/2018] [Indexed: 12/20/2022] Open
Abstract
Sensory neurons in trigeminal ganglia (TG) of calves latently infected with bovine herpesvirus 1 (BoHV-1) abundantly express latency-related (LR) gene products, including a protein (ORF2) and two micro-RNAs. Recent studies in mouse neuroblastoma cells (Neuro-2A) demonstrated ORF2 interacts with β-catenin and a β-catenin coactivator, high-mobility group AT-hook 1 (HMGA1) protein, which correlates with increased β-catenin-dependent transcription and cell survival. β-Catenin and HMGA1 are readily detected in a subset of latently infected TG neurons but not TG neurons from uninfected calves or reactivation from latency. Consequently, we hypothesized that the Wnt/β-catenin signaling pathway is differentially expressed during the latency and reactivation cycle and an active Wnt pathway promotes latency. RNA-sequencing studies revealed that 102 genes associated with the Wnt/β-catenin signaling pathway were differentially expressed in TG during the latency-reactivation cycle in calves. Wnt agonists were generally expressed at higher levels during latency, but these levels decreased during dexamethasone-induced reactivation. The Wnt agonist bone morphogenetic protein receptor 2 (BMPR2) was intriguing because it encodes a serine/threonine receptor kinase that promotes neuronal differentiation and inhibits cell death. Another differentially expressed gene encodes a protein kinase (Akt3), which is significant because Akt activity enhances cell survival and is linked to herpes simplex virus 1 latency and neuronal survival. Additional studies demonstrated ORF2 increased Akt3 steady-state protein levels and interacted with Akt3 in transfected Neuro-2A cells, which correlated with Akt3 activation. Conversely, expression of Wnt antagonists increased during reactivation from latency. Collectively, these studies suggest Wnt signaling cooperates with LR gene products, in particular ORF2, to promote latency.IMPORTANCE Lifelong BoHV-1 latency primarily occurs in sensory neurons. The synthetic corticosteroid dexamethasone consistently induces reactivation from latency in calves. RNA sequencing studies revealed 102 genes associated with the Wnt/β-catenin signaling pathway are differentially regulated during the latency-reactivation cycle. Two protein kinases associated with the Wnt pathway, Akt3 and BMPR2, were expressed at higher levels during latency but were repressed during reactivation. Furthermore, five genes encoding soluble Wnt antagonists and β-catenin-dependent transcription inhibitors were induced during reactivation from latency. These findings are important because Wnt, BMPR2, and Akt3 promote neurogenesis and cell survival, processes crucial for lifelong viral latency. In transfected neuroblastoma cells, a viral protein expressed during latency (ORF2) interacts with and enhances Akt3 protein kinase activity. These findings provide insight into how cellular factors associated with the Wnt signaling pathway cooperate with LR gene products to regulate the BoHV-1 latency-reactivation cycle.
Collapse
Affiliation(s)
- Aspen Workman
- United States Department of Agriculture, Agricultural Research Service, U.S. Meat Animal Research Center, Clay Center, Nebraska, USA
| | - Liqian Zhu
- Oklahoma State University Center for Veterinary Health Sciences, Department of Veterinary Pathobiology, Stillwater, Oklahoma, USA
- College of Veterinary Medicine and Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
| | - Brittney N Keel
- United States Department of Agriculture, Agricultural Research Service, U.S. Meat Animal Research Center, Clay Center, Nebraska, USA
| | - Timothy P L Smith
- United States Department of Agriculture, Agricultural Research Service, U.S. Meat Animal Research Center, Clay Center, Nebraska, USA
| | - Clinton Jones
- Oklahoma State University Center for Veterinary Health Sciences, Department of Veterinary Pathobiology, Stillwater, Oklahoma, USA
| |
Collapse
|
26
|
Hayashi Y, Mikawa S, Masumoto K, Katou F, Sato K. GDF11 expression in the adult rat central nervous system. J Chem Neuroanat 2018; 89:21-36. [PMID: 29448002 DOI: 10.1016/j.jchemneu.2018.02.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 11/16/2017] [Accepted: 02/10/2018] [Indexed: 01/12/2023]
Abstract
Growth differentiation factor 11 (GDF11), also known as bone morphogenetic protein 11 (BMP11), is a member of the transforming growth factor β (TGF-β) superfamily. Although GDF11 plays pivotal roles during development, including anterior/posterior patterning, formation of the kidney, stomach, spleen and endocrine pancreas, little information is available for GDF11 expression in the adult central nervous system (CNS). We, thus, investigated GDF11 expression in the adult rat CNS using immunohistochemistry. GDF11 was intensely expressed in most neurons and their axons. Furthermore, we found that astrocytes and ependymal cells also express GDF11 protein. These data indicate that GDF11 is widely expressed throughout the adult CNS, and its abundant expression in the adult brain strongly supports the idea that GDF11 plays important roles in the adult brain.
Collapse
Affiliation(s)
- Yutaro Hayashi
- Department of Dentistry and Oral and Maxillofacial Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan; Department of Organ & Tissue Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Sumiko Mikawa
- Department of Organ & Tissue Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Kazuma Masumoto
- Department of Dentistry and Oral and Maxillofacial Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Fuminori Katou
- Department of Dentistry and Oral and Maxillofacial Surgery, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Kohji Sato
- Department of Organ & Tissue Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka, 431-3192, Japan.
| |
Collapse
|
27
|
Lopes DM, Denk F, McMahon SB. The Molecular Fingerprint of Dorsal Root and Trigeminal Ganglion Neurons. Front Mol Neurosci 2017; 10:304. [PMID: 29018326 PMCID: PMC5623188 DOI: 10.3389/fnmol.2017.00304] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/11/2017] [Indexed: 12/15/2022] Open
Abstract
The dorsal root ganglia (DRG) and trigeminal ganglia (TG) are clusters of cell bodies of highly specialized sensory neurons which are responsible for relaying information about our environment to the central nervous system. Despite previous efforts to characterize sensory neurons at the molecular level, it is still unknown whether those present in DRG and TG have distinct expression profiles and therefore a unique molecular fingerprint. To address this question, we isolated lumbar DRG and TG neurons using fluorescence-activated cell sorting from Advillin-GFP transgenic mice and performed RNA sequencing. Our transcriptome analyses showed that, despite being overwhelmingly similar, a number of genes are differentially expressed in DRG and TG neurons. Importantly, we identified 24 genes which were uniquely expressed in either ganglia, including an arginine vasopressin receptor and several homeobox genes, giving each population a distinct molecular fingerprint. We compared our findings with published studies to reveal that many genes previously reported to be present in neurons are in fact likely to originate from other cell types in the ganglia. Additionally, our neuron-specific results aligned well with a dataset examining whole human TG and DRG. We propose that the data can both improve our understanding of primary afferent biology and help contribute to the development of drug treatments and gene therapies which seek targets with unique or restricted expression patterns.
Collapse
Affiliation(s)
- Douglas M Lopes
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Franziska Denk
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| | - Stephen B McMahon
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, London, United Kingdom
| |
Collapse
|
28
|
Abstract
Bone morphogenetic protein-7 (BMP7), a member of the transforming growth factor-β (TGF-β) superfamily, has various effects in many biological events. However, there is little information on BMP7 expression in the adult central nervous system (CNS). Therefore, we investigated BMP7 levels in the adult rat CNS using immunohistochemistry. Abundant BMP7 expression was seen in astrocytes throughout the CNS and strong BMP7 expression was also observed in neuropils of the gray matter. Furthermore, BMP7 expression was observed in several kinds of neurons, including oxytocin, dopaminergic and noradrenergic neurons. These data suggest that BMP7 is widely expressed throughout the adult CNS, and support the idea that BMP7 plays pivotal roles in the adult brain, as well as in the developing brain. BMP7 is expressed throughout the adult CNS, and abundantly expressed in astrocytes. BMP7 is also expressed in some kinds of neurons and axons.
Collapse
Key Words
- Astrocyte
- BMP, bone morphogenetic protein
- BMPR, bone morphogenetic protein receptor
- BSA, bovine serum albumin
- CNS, central nervous system
- CSPGs, chondroitin sulfate proteoglycans
- GFAP, glial fibrillary acidic protein
- IHC, immunohistochemistry
- IR, immunoreactivity
- Immunohistochemistry
- Neuron
- PB, phosphate buffer
- RT, room temperature
- SVZ, subventricular zone
- TGF-β, transforming growth factor β
- TTBS, Tris-buffered saline containing 0.05% Tween-20
Collapse
|
29
|
Chordin and noggin expression in the adult rat trigeminal nuclei. J Chem Neuroanat 2016; 78:36-41. [PMID: 27546891 DOI: 10.1016/j.jchemneu.2016.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 08/07/2016] [Indexed: 01/27/2023]
Abstract
Bone morphogenetic proteins (BMP) exert its biological functions by interacting with membrane bound receptors. However, functions of BMPs are also regulated in the extracellular space by secreted antagonistic regulators, such as chordin and noggin. Although the deep involvement of BMP signaling in the development and functions of the trigeminal nuclei has been postulated, little information is available for its expression in the trigeminal nuclei. We, thus, investigated chordin and noggin expression in the adult rat trigeminal nuclei using immunohistochemistry. Chordin and noggin were intensely expressed throughout the trigeminal nuclei. In addition, interesting differences are observed between chordin expression and noggin expression. For example, chordin prefers dendritic expression than noggin, suggesting that chordin is involved in the regulation of dendritic morphology and synaptic homeostasis. Furthermore, chordin and noggin were differentially expressed in the neuropil of the trigeminal nuclei. Since BMP signaling is known to play a pivotal role to make precise neural network, theses differences might be important to keep precise interneuronal connections by regulating local BMP signaling intensity in each region. Interestingly, we also detected chordin and noggin expression in axons of the trigeminal nerves. These data indicate that chordin and noggin play pivotal roles also in the adult trigeminal system.
Collapse
|
30
|
Lo FS, Erzurumlu RS. Sensory Activity-Dependent and Sensory Activity-Independent Properties of the Developing Rodent Trigeminal Principal Nucleus. Dev Neurosci 2016; 38:163-170. [PMID: 27287019 DOI: 10.1159/000446395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 04/24/2016] [Indexed: 11/19/2022] Open
Abstract
The whisker-sensory trigeminal central pathway of rodents is an established model for studies of activity-dependent neural plasticity. The first relay station of the pathway is the trigeminal principal nucleus (PrV), the ventral part of which receives sensory inputs mainly from the infraorbital branch of the maxillary trigeminal nerve (ION). Whisker-sensory afferents play an important role in the development of the morphological and physiological properties of PrV neurons. In neonates, deafferentation by ION transection leads to the disruption of whisker-related neural patterns (barrelettes) and cell death within a specific time window (critical period), as revealed by morphological studies. Whisker-sensory inputs control synaptic elimination, postsynaptic AMPA receptor trafficking, astrocyte-mediated synaptogenesis, and receptive-field characteristics of PrV cells, without a postnatal critical period. Sensory activity-dependent synaptic plasticity requires the activation of NMDA receptors and involves the participation of glia. However, the basic physiological properties of PrV neurons, such as cell type-specific ion channels, presynaptic terminal function, postsynaptic NMDA receptor subunit composition, and formation of the inhibitory circuitry, are independent of sensory inputs. Therefore, the first relay station of the whisker sensation is largely mature-like and functional at birth. Delineation of activity-dependent and activity-independent features of the postnatal PrV is important for understanding the development and functional characteristics of downstream trigeminal stations in the thalamus and neocortex. This mini review focuses on such features of the developing rodent PrV.
Collapse
Affiliation(s)
- Fu-Sun Lo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Md., USA
| | | |
Collapse
|
31
|
Yamashita K, Mikawa S, Sato K. BMP3 expression in the adult rat CNS. Brain Res 2016; 1643:35-50. [PMID: 27130896 DOI: 10.1016/j.brainres.2016.04.057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 03/14/2016] [Accepted: 04/25/2016] [Indexed: 12/13/2022]
Abstract
Bone morphogenetic protein-3 (BMP3) is a very unique member of the TGF-β superfamily, because it functions as an antagonist to both the canonical BMP and activin pathways and plays important roles in multiple biological events. Although BMP3 expression has been described in the early development of the kidney, intestine and bone, little information is available for BMP3 expression in the central nervous system (CNS). We, thus, investigated BMP3 expression in the adult rat CNS using immunohistochemistry. BMP3 was intensely expressed in most neurons and their axons. Furthermore, we found that astrocytes and ependymal cells also express BMP3 protein. These data indicate that BMP3 is widely expressed throughout the adult CNS, and its abundant expression in the adult brain strongly supports the idea that BMP3 plays important roles in the adult brain.
Collapse
Affiliation(s)
- Kanna Yamashita
- Department of Basic Nursing, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Sumiko Mikawa
- Department of Anatomy & Neuroscience, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka 431-3192, Japan
| | - Kohji Sato
- Department of Anatomy & Neuroscience, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka 431-3192, Japan.
| |
Collapse
|
32
|
Karpinski BA, Bryan CA, Paronett EM, Baker JL, Fernandez A, Horvath A, Maynard TM, Moody SA, LaMantia AS. A cellular and molecular mosaic establishes growth and differentiation states for cranial sensory neurons. Dev Biol 2016; 415:228-241. [PMID: 26988119 DOI: 10.1016/j.ydbio.2016.03.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 02/02/2016] [Accepted: 03/13/2016] [Indexed: 02/06/2023]
Abstract
We compared apparent origins, cellular diversity and regulation of initial axon growth for differentiating cranial sensory neurons. We assessed the molecular and cellular composition of the developing olfactory and otic placodes, and cranial sensory ganglia to evaluate contributions of ectodermal placode versus neural crest at each site. Special sensory neuron populations-the olfactory and otic placodes, as well as those in vestibulo-acoustic ganglion- are entirely populated with cells expressing cranial placode-associated, rather than neural crest-associated markers. The remaining cranial sensory ganglia are a mosaic of cells that express placode-associated as well as neural crest-associated markers. We found two distinct populations of neural crest in the cranial ganglia: the first, as expected, is labeled by Wnt1:Cre mediated recombination. The second is not labeled by Wnt1:Cre recombination, and expresses both Sox10 and FoxD3. These populations-Wnt1:Cre recombined, and Sox10/Foxd3-expressing- are proliferatively distinct from one another. Together, the two neural crest-associated populations are substantially more proliferative than their placode-associated counterparts. Nevertheless, the apparently placode- and neural crest-associated populations are similarly sensitive to altered signaling that compromises cranial morphogenesis and differentiation. Acute disruption of either Fibroblast growth factor (Fgf) or Retinoic acid (RA) signaling alters axon growth and cell death, but does not preferentially target any of the three distinct populations. Apparently, mosaic derivation and diversity of precursors and early differentiating neurons, modulated uniformly by local signals, supports early cranial sensory neuron differentiation and growth.
Collapse
Affiliation(s)
- Beverly A Karpinski
- Department of Pharmacology and Physiology, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA; Department of Anatomy and Regenerative Biology, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA; The GW Institute for Neuroscience, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA.
| | - Corey A Bryan
- Department of Pharmacology and Physiology, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA; The GW Institute for Neuroscience, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA.
| | - Elizabeth M Paronett
- Department of Pharmacology and Physiology, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA; The GW Institute for Neuroscience, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA.
| | - Jennifer L Baker
- Center for the Advanced Study of Human Paleobiology, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA.
| | - Alejandra Fernandez
- Department of Pharmacology and Physiology, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA; The GW Institute for Neuroscience, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA.
| | - Anelia Horvath
- Department of Pharmacology and Physiology, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA; The GW Institute for Neuroscience, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA.
| | - Thomas M Maynard
- Department of Pharmacology and Physiology, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA; The GW Institute for Neuroscience, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA.
| | - Sally A Moody
- Department of Anatomy and Regenerative Biology, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA; The GW Institute for Neuroscience, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA.
| | - Anthony-S LaMantia
- Department of Pharmacology and Physiology, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA; The GW Institute for Neuroscience, The George Washington University, School of Medicine and Health Sciences, Washington DC, USA.
| |
Collapse
|
33
|
Adameyko I, Fried K. The Nervous System Orchestrates and Integrates Craniofacial Development: A Review. Front Physiol 2016; 7:49. [PMID: 26924989 PMCID: PMC4759458 DOI: 10.3389/fphys.2016.00049] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/02/2016] [Indexed: 01/14/2023] Open
Abstract
Development of a head is a dazzlingly complex process: a number of distinct cellular sources including cranial ecto- and endoderm, mesoderm and neural crest contribute to facial and other structures. In the head, an extremely fine-tuned developmental coordination of CNS, peripheral neural components, sensory organs and a musculo-skeletal apparatus occurs, which provides protection and functional integration. The face can to a large extent be considered as an assembly of sensory systems encased and functionally fused with appendages represented by jaws. Here we review how the developing brain, neurogenic placodes and peripheral nerves influence the morphogenesis of surrounding tissues as a part of various general integrative processes in the head. The mechanisms of this impact, as we understand it now, span from the targeted release of the morphogens necessary for shaping to providing a niche for cellular sources required in later development. In this review we also discuss the most recent findings and ideas related to how peripheral nerves and nerve-associated cells contribute to craniofacial development, including teeth, during the post- neural crest period and potentially in regeneration.
Collapse
Affiliation(s)
- Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska InstitutetStockholm, Sweden; Department of Molecular Neurosciences, Center of Brain Research, Medical University of ViennaVienna, Austria
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institutet Stockholm, Sweden
| |
Collapse
|
34
|
O'Keeffe GW, Gutierrez H, Howard L, Laurie CW, Osorio C, Gavaldà N, Wyatt SL, Davies AM. Region-specific role of growth differentiation factor-5 in the establishment of sympathetic innervation. Neural Dev 2016; 11:4. [PMID: 26878848 PMCID: PMC4755026 DOI: 10.1186/s13064-016-0060-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/08/2016] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Nerve growth factor (NGF) is the prototypical target-derived neurotrophic factor required for sympathetic neuron survival and for the growth and ramification of sympathetic axons within most but not all sympathetic targets. This implies the operation of additional target-derived factors for regulating terminal sympathetic axon growth and branching. RESULTS Here report that growth differentiation factor 5 (GDF5), a widely expressed member of the transforming growth factor beta (TGFβ) superfamily required for limb development, promoted axon growth from mouse superior cervical ganglion (SCG) neurons independently of NGF and enhanced axon growth in combination with NGF. GDF5 had no effect on neuronal survival and influenced axon growth during a narrow window of postnatal development when sympathetic axons are ramifying extensively in their targets in vivo. SCG neurons expressed all receptors capable of participating in GDF5 signaling at this stage of development. Using compartment cultures, we demonstrated that GDF5 exerted its growth promoting effect by acting directly on axons and by initiating retrograde canonical Smad signalling to the nucleus. GDF5 is synthesized in sympathetic targets, and examination of several anatomically circumscribed tissues in Gdf5 null mice revealed regional deficits in sympathetic innervation. There was a marked, highly significant reduction in the sympathetic innervation density of the iris, a smaller though significant reduction in the trachea, but no reduction in the submandibular salivary gland. There was no reduction in the number of neurons in the SCG. CONCLUSIONS These findings show that GDF5 is a novel target-derived factor that promotes sympathetic axon growth and branching and makes a distinctive regional contribution to the establishment of sympathetic innervation, but unlike NGF, plays no role in regulating sympathetic neuron survival.
Collapse
Affiliation(s)
- Gerard W O'Keeffe
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
- Dept. Anatomy/Neuroscience and Biosciences Institute, UCC, Cork, Ireland
| | - Humberto Gutierrez
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
- Current address, School of Life Sciences, University of Lincoln, Brayford Pool, Lincoln, LN6 7TS, UK
| | - Laura Howard
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | | | - Catarina Osorio
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
- Current address, MRC Centre for Developmental Neurobiology, King's College London, New Hunt's House, 4th Floor, Guy's Hospital Campus, London, SE1 1UL, UK
| | - Núria Gavaldà
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
- Current address, SOM Innovation Biotech SL, c/Baldiri Reixac 4, 08028, Barcelona, Spain
| | - Sean L Wyatt
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK
| | - Alun M Davies
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AT, UK.
| |
Collapse
|
35
|
Berndt AJE, Tang JCY, Ridyard MS, Lian T, Keatings K, Allan DW. Gene Regulatory Mechanisms Underlying the Spatial and Temporal Regulation of Target-Dependent Gene Expression in Drosophila Neurons. PLoS Genet 2015; 11:e1005754. [PMID: 26713626 PMCID: PMC4694770 DOI: 10.1371/journal.pgen.1005754] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 11/30/2015] [Indexed: 11/18/2022] Open
Abstract
Neuronal differentiation often requires target-derived signals from the cells they innervate. These signals typically activate neural subtype-specific genes, but the gene regulatory mechanisms remain largely unknown. Highly restricted expression of the FMRFa neuropeptide in Drosophila Tv4 neurons requires target-derived BMP signaling and a transcription factor code that includes Apterous. Using integrase transgenesis of enhancer reporters, we functionally dissected the Tv4-enhancer of FMRFa within its native cellular context. We identified two essential but discrete cis-elements, a BMP-response element (BMP-RE) that binds BMP-activated pMad, and a homeodomain-response element (HD-RE) that binds Apterous. These cis-elements have low activity and must be combined for Tv4-enhancer activity. Such combinatorial activity is often a mechanism for restricting expression to the intersection of cis-element spatiotemporal activities. However, concatemers of the HD-RE and BMP-RE cis-elements were found to independently generate the same spatiotemporal expression as the Tv4-enhancer. Thus, the Tv4-enhancer atypically combines two low-activity cis-elements that confer the same output from distinct inputs. The activation of target-dependent genes is assumed to 'wait' for target contact. We tested this directly, and unexpectedly found that premature BMP activity could not induce early FMRFa expression; also, we show that the BMP-insensitive HD-RE cis-element is activated at the time of target contact. This led us to uncover a role for the nuclear receptor, seven up (svp), as a repressor of FMRFa induction prior to target contact. Svp is normally downregulated immediately prior to target contact, and we found that maintaining Svp expression prevents cis-element activation, whereas reducing svp gene dosage prematurely activates cis-element activity. We conclude that the target-dependent FMRFa gene is repressed prior to target contact, and that target-derived BMP signaling directly activates FMRFa gene expression through an atypical gene regulatory mechanism.
Collapse
Affiliation(s)
- Anthony J. E. Berndt
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jonathan C. Y. Tang
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States America
| | - Marc S. Ridyard
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tianshun Lian
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kathleen Keatings
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Douglas W. Allan
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
36
|
Bechara A, Laumonnerie C, Vilain N, Kratochwil CF, Cankovic V, Maiorano NA, Kirschmann MA, Ducret S, Rijli FM. Hoxa2 Selects Barrelette Neuron Identity and Connectivity in the Mouse Somatosensory Brainstem. Cell Rep 2015; 13:783-797. [PMID: 26489473 DOI: 10.1016/j.celrep.2015.09.031] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 07/19/2015] [Accepted: 09/09/2015] [Indexed: 10/22/2022] Open
Abstract
Mouse whiskers are somatotopically mapped in brainstem trigeminal nuclei as neuronal modules known as barrelettes. Whisker-related afferents form barrelettes in ventral principal sensory (vPrV) nucleus, whereas mandibular input targets dorsal PrV (dPrV). How barrelette neuron identity and circuitry is established is poorly understood. We found that ectopic Hoxa2 expression in dPrV neurons is sufficient to attract whisker-related afferents, induce asymmetrical dendrite arbors, and allow ectopic barrelette map formation. Moreover, the thalamic area forming whisker-related barreloids is prenatally targeted by both vPrV and dPrV axons followed by perinatal large-scale pruning of dPrV axons and refinement of vPrV barrelette input. Ectopic Hoxa2 expression allows topographically directed targeting and refinement of dPrV axons with vPrV axons into a single whisker-related barreloid map. Thus, a single HOX transcription factor is sufficient to switch dPrV into a vPrV barrelette neuron program and coordinate input-output topographic connectivity of a dermatome-specific circuit module.
Collapse
Affiliation(s)
- Ahmad Bechara
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Christophe Laumonnerie
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Nathalie Vilain
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Claudius F Kratochwil
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Vanja Cankovic
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Nicola A Maiorano
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Moritz A Kirschmann
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Sebastien Ducret
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Filippo M Rijli
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland; University of Basel, 4056 Basel, Switzerland.
| |
Collapse
|
37
|
Laumonnerie C, Bechara A, Vilain N, Kurihara Y, Kurihara H, Rijli FM. Facial whisker pattern is not sufficient to instruct a whisker-related topographic map in the mouse somatosensory brainstem. Development 2015; 142:3704-12. [PMID: 26417040 DOI: 10.1242/dev.128736] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/15/2015] [Indexed: 11/20/2022]
Abstract
Facial somatosensory input is relayed by trigeminal ganglion (TG) neurons and serially wired to brainstem, thalamus and cortex. Spatially ordered sets of target neurons generate central topographic maps reproducing the spatial arrangement of peripheral facial receptors. Facial pattern provides a necessary template for map formation, but may be insufficient to impose a brain somatotopic pattern. In mice, lower jaw sensory information is relayed by the trigeminal nerve mandibular branch, whose axons target the brainstem dorsal principal sensory trigeminal nucleus (dPrV). Input from mystacial whiskers is relayed by the maxillary branch and forms a topographic representation of rows and whiskers in the ventral PrV (vPrV). To investigate peripheral organisation in imposing a brain topographic pattern, we analysed Edn1(-/-) mice, which present ectopic whisker rows on the lower jaw. We found that these whiskers were innervated by mandibular TG neurons which initially targeted dPrV. Unlike maxillary TG neurons, the ectopic whisker-innervating mandibular neuron cell bodies and pre-target central axons did not segregate into a row-specific pattern nor target the dPrV with a topographic pattern. Following periphery-driven molecular repatterning to a maxillary-like identity, mandibular neurons partially redirected their central projections from dPrV to vPrV. Thus, while able to induce maxillary-like molecular features resulting in vPrV final targeting, a spatially ordered lower jaw ectopic whisker pattern is insufficient to impose row-specific pre-target organisation of the central mandibular tract or a whisker-related matching pattern of afferents in dPrV. These results provide novel insights into periphery-dependent versus periphery-independent mechanisms of trigeminal ganglion and brainstem patterning in matching whisker topography.
Collapse
Affiliation(s)
- Christophe Laumonnerie
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel 4058, Switzerland
| | - Ahmad Bechara
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel 4058, Switzerland
| | - Nathalie Vilain
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel 4058, Switzerland
| | - Yukiko Kurihara
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8654, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo, 102-0075, Japan
| | - Hiroki Kurihara
- Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8654, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo, 102-0075, Japan
| | - Filippo M Rijli
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel 4058, Switzerland University of Basel, Basel 4056, Switzerland
| |
Collapse
|
38
|
Onecut1 and Onecut2 transcription factors operate downstream of Pax6 to regulate horizontal cell development. Dev Biol 2015; 402:48-60. [PMID: 25794677 DOI: 10.1016/j.ydbio.2015.02.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 02/18/2015] [Accepted: 02/22/2015] [Indexed: 11/22/2022]
Abstract
Genetic studies of the last decades strongly indicated that generation of particular retinal cell types is governed by gene regulatory networks of transcription factors and their target genes. The paired and homeodomain transcription factor Pax6 plays a pivotal role in retinal development as its inactivation in the retinal progenitor cell population leads to abolished differentiation of all retinal cell types. However, until now, only a few transcription factors operating downstream of Pax6 responsible for generation of individual retinal cell types have been identified. In this study, we identified two transcription factors of the Onecut family, Onecut1 and Onecut2, as Pax6 downstream-acting factors. Onecut1 and Onecut2 were previously shown to be expressed in developing horizontal cells, retinal ganglion cells and cone photoreceptors; however, their role in differentiation of these cell types is poorly understood. In this study, we show that the horizontal cell genesis is severely disturbed in Onecut-deficient retinae. In single Onecut1 and Onecut2 mutants, the number of horizontal cells is dramatically reduced while horizontal cells are completely missing in the Onecut1/Onecut2 compound mutant. Analysis of genes involved in the horizontal cell genesis such as Foxn4, Ptf1a, Prox1 and Lim1 showed that although horizontal cells are initially formed, they are not maintained in Onecut-deficient retinae. Taken together, this study suggests the model in which Pax6 regulates the maintenance of horizontal cells through the activation of Onecut1 and Onecut2 transcription factors.
Collapse
|
39
|
Interaction between Oc-1 and Lmx1a promotes ventral midbrain dopamine neural stem cells differentiation into dopamine neurons. Brain Res 2015; 1608:40-50. [PMID: 25747864 DOI: 10.1016/j.brainres.2015.02.046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 12/20/2022]
Abstract
Recent studies have shown that Onecut (Oc) transcription factors may be involved in the early development of midbrain dopaminergic neurons (mdDA). The expression profile of Oc factors matches that of Lmx1a, an important intrinsic transcription factor in the development of mDA neuron. Moreover, the Wnt1-Lmx1a pathway controls the mdDA differentiation. However, their expression dynamics and molecular mechanisms remain to be determined. To address these issues, we hypothesize that cross-talk between Oc-1 and Lmx1a regulates the mdDA specification and differentiation through the canonical Wnt-β-catenin pathway. We found that Oc-1 and Lmx1a displayed a very similar expression profile from embryonic to adult ventral midbrain (VM) tissues. Oc-1 regulated the proliferation and differentiation of ventral midbrain neural stem cells (vmNSCs). Downregulation of Oc-1 decreased both transcript and protein level of Lmx1a. Oc-1 interacted with lmx1a in vmNSCs in vitro and in VM tissues in vivo. Knockdown of Lmx1a reduced the expression of Oc-1 and Wnt1 in vmNSCs. Inhibiting Wnt1 signaling in vmNSCs provoked similar responses. Our data suggested that Oc-1 interacts with Lmx1a to promote vmNSCs differentiation into dopamine neuron through Wnt1-Lmx1a pathway.
Collapse
|
40
|
BMP5 expression in the adult rat brain. Neuroscience 2014; 284:972-987. [PMID: 25110111 DOI: 10.1016/j.neuroscience.2014.07.057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 07/23/2014] [Accepted: 07/24/2014] [Indexed: 01/27/2023]
Abstract
Bone morphogenetic protein-5 (BMP5), a member of the transforming growth factor-β (TGF-β) superfamily, has many effects in several biological events. Although BMP5 expression has been well reported in the early development of the central nervous system (CNS), there is little information about its expression in the adult CNS. Thus, we analyzed BMP5 expression in the adult rat CNS by immunohistochemistry. Abundant BMP5 expression was observed in most neurons, and their dendrites and axons. Furthermore, strong BMP5 expression was also detected in the neuropil of the gray matters with high plasticity, such as the molecular layer of the cerebellum, locus coeruleus, and nucleus of the solitary tract. In addition, we showed BMP5 expression also in astrocytes, ependymal cells and meninges. Our data suggest that BMP5 is widely expressed throughout the adult CNS, and this abundant expression in the adult brain strongly supports the idea that BMP5 plays important roles not only in the developing brain but also in the adult brain.
Collapse
|
41
|
Zhong J, Zou H. BMP signaling in axon regeneration. Curr Opin Neurobiol 2014; 27:127-34. [PMID: 24713578 PMCID: PMC4122622 DOI: 10.1016/j.conb.2014.03.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Revised: 03/13/2014] [Accepted: 03/14/2014] [Indexed: 11/17/2022]
Abstract
Neuronal competence to re-extend axons and a permissive environment that allows growth cone navigation are two major determinants for successful axon regeneration. Here, we review the roles of bone morphogenetic protein (BMP) signaling in mediating both neuronal and glial injury responses after CNS injury. BMPs can activate a pro-regenerative transcription program in neurons through Smad-mediated canonical pathway, or act locally on cytoskeleton assembly at distal axons via non-canonical pathways. Emerging evidence implicates retrograde BMP signalosomes in connecting the cytoskeletal and nuclear responses. In addition, BMP/Smad signaling modulates neurotrophin-mediated axonal outgrowth, and interacts with the epigenetic machinery to initiate epigenetic reprogramming for axon regeneration. Besides their influences on neurons, BMPs also regulate astrogliosis, inflammatory processes, and neural progenitor cell differentiation at the injury site, all of which can either positively or negatively modify the injury microenvironment. Lastly, an increasing number of BMP signaling partners, sensitizers, and downstream effectors collectively fine-tune the signaling intensity and spatiotemporal dynamics of BMP activity in an integrated signaling network during axon regeneration.
Collapse
Affiliation(s)
- Jian Zhong
- Burke Medical Research Institute, 785 Mamaroneck Ave., White Plains, NY 10605, United States; Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, United States
| | - Hongyan Zou
- Fishberg Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States; Department of Neurosurgery, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States.
| |
Collapse
|
42
|
Modrell MS, Hockman D, Uy B, Buckley D, Sauka-Spengler T, Bronner ME, Baker CVH. A fate-map for cranial sensory ganglia in the sea lamprey. Dev Biol 2014; 385:405-16. [PMID: 24513489 PMCID: PMC3928997 DOI: 10.1016/j.ydbio.2013.10.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 10/17/2013] [Accepted: 10/21/2013] [Indexed: 11/30/2022]
Abstract
Cranial neurogenic placodes and the neural crest make essential contributions to key adult characteristics of all vertebrates, including the paired peripheral sense organs and craniofacial skeleton. Neurogenic placode development has been extensively characterized in representative jawed vertebrates (gnathostomes) but not in jawless fishes (agnathans). Here, we use in vivo lineage tracing with DiI, together with neuronal differentiation markers, to establish the first detailed fate-map for placode-derived sensory neurons in a jawless fish, the sea lamprey Petromyzon marinus, and to confirm that neural crest cells in the lamprey contribute to the cranial sensory ganglia. We also show that a pan-Pax3/7 antibody labels ophthalmic trigeminal (opV, profundal) placode-derived but not maxillomandibular trigeminal (mmV) placode-derived neurons, mirroring the expression of gnathostome Pax3 and suggesting that Pax3 (and its single Pax3/7 lamprey ortholog) is a pan-vertebrate marker for opV placode-derived neurons. Unexpectedly, however, our data reveal that mmV neuron precursors are located in two separate domains at neurula stages, with opV neuron precursors sandwiched between them. The different branches of the mmV nerve are not comparable between lampreys and gnatho-stomes, and spatial segregation of mmV neuron precursor territories may be a derived feature of lampreys. Nevertheless, maxillary and mandibular neurons are spatially segregated within gnathostome mmV ganglia, suggesting that a more detailed investigation of gnathostome mmV placode development would be worthwhile. Overall, however, our results highlight the conservation of cranial peripheral sensory nervous system development across vertebrates, yielding insight into ancestral vertebrate traits. The first detailed fate-map for placode-derived sensory neurons in a jawless fish. Pax3 is a pan-vertebrate marker for ophthalmic trigeminal placode-derived neurons. Maxillomandibular trigeminal neuron precursors are located in two separate domains. Confirmation that lamprey neural crest cells contribute to cranial sensory ganglia. Results overall highlight conservation of cranial sensory nervous system development.
Collapse
|
43
|
Francius C, Clotman F. Generating spinal motor neuron diversity: a long quest for neuronal identity. Cell Mol Life Sci 2014; 71:813-29. [PMID: 23765105 PMCID: PMC11113339 DOI: 10.1007/s00018-013-1398-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 05/30/2013] [Accepted: 05/31/2013] [Indexed: 03/26/2023]
Abstract
Understanding how thousands of different neuronal types are generated in the CNS constitutes a major challenge for developmental neurobiologists and is a prerequisite before considering cell or gene therapies of nervous lesions or pathologies. During embryonic development, spinal motor neurons (MNs) segregate into distinct subpopulations that display specific characteristics and properties including molecular identity, migration pattern, allocation to specific motor columns, and innervation of defined target. Because of the facility to correlate these different characteristics, the diversification of spinal MNs has become the model of choice for studying the molecular and cellular mechanisms underlying the generation of multiple neuronal populations in the developing CNS. Therefore, how spinal motor neuron subpopulations are produced during development has been extensively studied during the last two decades. In this review article, we will provide a comprehensive overview of the genetic and molecular mechanisms that contribute to the diversification of spinal MNs.
Collapse
Affiliation(s)
- Cédric Francius
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Neural Differentiation, 55 Avenue Hippocrate, Box (B1.55.11), 1200 Brussels, Belgium
| | - Frédéric Clotman
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Neural Differentiation, 55 Avenue Hippocrate, Box (B1.55.11), 1200 Brussels, Belgium
| |
Collapse
|
44
|
Lykissas MG, Aichmair A, Sama AA, Hughes AP, Lebl DR, Cammisa FP, Girardi FP. Nerve injury and recovery after lateral lumbar interbody fusion with and without bone morphogenetic protein-2 augmentation: a cohort-controlled study. Spine J 2014; 14:217-24. [PMID: 24269858 DOI: 10.1016/j.spinee.2013.06.109] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 06/29/2013] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Despite common use of intraoperative electrophysiologic neuromonitoring, injuries to the lumbar plexus during lateral lumbar interbody fusion (LLIF) have been reported. Emerging data suggest that recombinant human bone morphogenetic protein-2 (rhBMP-2) use during an anterior or transforaminal lumbar interbody fusion may be associated with an increased risk of neurological deficit. Clinical data on the sequelae of rhBMP-2 implantation in close proximity to the lumbosacral plexus during LLIF remains to be understood. PURPOSE The purpose of this study was to compare the incidence of neurologic deficits and pain in patients undergoing LLIF with and without rhBMP-2. STUDY DESIGN/SETTING Retrospective outcome analysis in controlled cohorts undergoing the lateral exposure technique for LLIF with and without rhBMP-2. METHODS The electronic medical records of patients undergoing LLIF with and without supplemental posterior fusion for degenerative spinal conditions were retrospectively reviewed over a 6-year period. Patients with previous lumbar spine surgery or follow-up of less than 6 months were excluded. Patients were divided into 2 groups, Group 1 (rhBMP-2 use; n=72) and Group 2 (autograft/allograft use; n=72), and were matched according to the age at the time of surgery, gender, weight, body mass index, side of approach, total number of treated spinal segments, use of supplemental posterior fusion, and length of follow-up. RESULTS Immediately after surgery, a sensory deficit was recorded in 33 patients in Group 1 and 35 patients in Group 2 (odds ratio [OR] 0.895; 90% confidence interval [CI] 0.516-1.550; p=.739). At last follow-up, a persistent sensory deficit was identified in 29 patients whose LLIF procedure was supplemented by rhBMP-2 and 20 patients in whom autograft/allograft was used (OR 1.754; 90% CI 0.976-3.151; p=.115). A motor deficit was recorded in 37 patients immediately after the rhBMP-2 procedure and 28 patients treated with autograft/allograft (OR 1.661; 90% CI 0.953-2.895; p=.133). A persistent motor deficit was recorded in 35 and 17 patients in Groups 1 and 2, respectively, at last follow-up (OR 3.060; 90% CI 1.681-5.571; p=.002). During the first postoperative examination, 37 patients in Group 1 and 25 patients in Group 2 complained of anterior thigh or groin pain (OR 1.987; 90% CI 1.133-3.488; p=.045). At last follow-up, there was a significantly higher number of patients in Group 1 who complained of persistent anterior thigh or groin pain than Group 2 (8 vs. 0 patients) (OR 16.470; 90% CI 1.477-183.700; p=.006). CONCLUSIONS Our results provide evidence of an increased rate of postoperative neurologic deficit and anterior thigh/groin pain after LLIF using rhBMP-2, when compared with matched controls without rhBMP-2 exposure. This study suggests a potential direct deleterious effect of rhBMP-2 on the lumbosacral plexus.
Collapse
Affiliation(s)
- Marios G Lykissas
- Department of Orthopedic Surgery, Spine and Scoliosis Service, Hospital for Special Surgery, Weill Cornell Medical College, 535 E. 70th St, New York, NY 10021, USA.
| | - Alexander Aichmair
- Department of Orthopedic Surgery, Spine and Scoliosis Service, Hospital for Special Surgery, Weill Cornell Medical College, 535 E. 70th St, New York, NY 10021, USA
| | - Andrew A Sama
- Department of Orthopedic Surgery, Spine and Scoliosis Service, Hospital for Special Surgery, Weill Cornell Medical College, 535 E. 70th St, New York, NY 10021, USA
| | - Alexander P Hughes
- Department of Orthopedic Surgery, Spine and Scoliosis Service, Hospital for Special Surgery, Weill Cornell Medical College, 535 E. 70th St, New York, NY 10021, USA
| | - Darren R Lebl
- Department of Orthopedic Surgery, Spine and Scoliosis Service, Hospital for Special Surgery, Weill Cornell Medical College, 535 E. 70th St, New York, NY 10021, USA
| | - Frank P Cammisa
- Department of Orthopedic Surgery, Spine and Scoliosis Service, Hospital for Special Surgery, Weill Cornell Medical College, 535 E. 70th St, New York, NY 10021, USA
| | - Federico P Girardi
- Department of Orthopedic Surgery, Spine and Scoliosis Service, Hospital for Special Surgery, Weill Cornell Medical College, 535 E. 70th St, New York, NY 10021, USA
| |
Collapse
|
45
|
Dendrite complexity of sympathetic neurons is controlled during postnatal development by BMP signaling. J Neurosci 2013; 33:15132-44. [PMID: 24048844 DOI: 10.1523/jneurosci.4748-12.2013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Dendrite development is controlled by the interplay of intrinsic and extrinsic signals affecting initiation, growth, and maintenance of complex dendrites. Bone morphogenetic proteins (BMPs) stimulate dendrite growth in cultures of sympathetic, cortical, and hippocampal neurons but it was unclear whether BMPs control dendrite morphology in vivo. Using a conditional knock-out strategy to eliminate Bmpr1a and Smad4 in immature noradrenergic sympathetic neurons we now show that dendrite length, complexity, and neuron cell body size are reduced in adult mice deficient of Bmpr1a. The combined deletion of Bmpr1a and Bmpr1b causes no further decrease in dendritic features. Sympathetic neurons devoid of Bmpr1a/1b display normal Smad1/5/8 phosphorylation, which suggests that Smad-independent signaling paths are involved in dendritic growth control downstream of BMPR1A/B. Indeed, in the Smad4 conditional knock-out dendrite and cell body size are not affected and dendrite complexity and number are increased. Together, these results demonstrate an in vivo function for BMPs in the generation of mature sympathetic neuron dendrites. BMPR1 signaling controls dendrite complexity postnatally during the major dendritic growth period of sympathetic neurons.
Collapse
|
46
|
Mikawa S, Sato K. Chordin expression in the adult rat brain. Neuroscience 2013; 258:16-33. [PMID: 24231736 DOI: 10.1016/j.neuroscience.2013.11.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Revised: 10/11/2013] [Accepted: 11/03/2013] [Indexed: 11/25/2022]
Abstract
Bone morphogenetic proteins (BMPs) exert its biological functions by interacting with membrane bound receptors. However, functions of BMPs are also regulated in the extracellular space by secreted antagonistic regulators. Chordin is an extracellular BMP antagonist that binds BMP-2, 4, and 7 with high affinity and thus interferes with binding to BMP receptors. Although chordin expression has been well described in the early development of the CNS, little information is available for its expression in the adult CNS. We, thus, investigated chordin expression in the adult rat CNS using immunohistochemistry. Chordin was intensely expressed in most neurons, and their dendrites and axons. In addition, abundant chordin expression was also observed in the neuropil of the gray matters where high plasticity is reported, such as the molecular layer of the cerebellum and the superficial layer of the superior colliculus. Furthermore, we found that astrocytes and ependymal cells also express chordin protein. These data indicate that chordin is more widely expressed throughout the adult CNS than previously reported, and its continued abundant expression in the adult brain strongly supports the idea that chordin plays pivotal roles also in the adult brain.
Collapse
Affiliation(s)
- S Mikawa
- Department of Anatomy & Neuroscience, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka 431-3192, Japan
| | - K Sato
- Department of Anatomy & Neuroscience, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashiku, Hamamatsu, Shizuoka 431-3192, Japan.
| |
Collapse
|
47
|
Ji SJ, Jaffrey SR. Axonal transcription factors: novel regulators of growth cone-to-nucleus signaling. Dev Neurobiol 2013; 74:245-58. [PMID: 23897628 DOI: 10.1002/dneu.22112] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 07/11/2013] [Accepted: 07/15/2013] [Indexed: 02/03/2023]
Abstract
Developing axons contain transcripts that are locally translated to influence the axonal proteome. Recent studies have shown that axonal transcripts include mRNAs encoding transcription factors. These mRNAs are translated to produce transcription factors that can be retrogradely trafficked back to the nucleus, where they regulate gene expression programs. These findings point to a novel mechanism of growth cone-to-nucleus signaling that occurs when growth cones encounter extracellular signaling molecules that stimulate local translation of these transcription factors, thereby influencing gene transcription. Here we summarize recent findings on local translation of transcription factors in axons and their roles in different neuronal processes such as neuronal specification, survival, and axon regeneration. Comprehensive axonal transcriptome studies have revealed transcripts that encode many more transcription factors and cofactors, suggesting a potentially broad role for this type of signaling. We review the progress on the approaches and tools that have been developed to study local translation and retrograde trafficking of transcription factors. We also highlight the challenges in the field and discuss the potential routes to resolving them.
Collapse
Affiliation(s)
- Sheng-Jian Ji
- Department of Pharmacology, Weill Medical College, Cornell University, New York, New York, 10065
| | | |
Collapse
|
48
|
Engelhard C, Sarsfield S, Merte J, Wang Q, Li P, Beppu H, Kolodkin AL, Sucov HM, Ginty DD. MEGF8 is a modifier of BMP signaling in trigeminal sensory neurons. eLife 2013; 2:e01160. [PMID: 24052814 PMCID: PMC3776557 DOI: 10.7554/elife.01160] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 08/16/2013] [Indexed: 01/15/2023] Open
Abstract
Bone morphogenetic protein (BMP) signaling has emerged as an important regulator of sensory neuron development. Using a three-generation forward genetic screen in mice we have identified Megf8 as a novel modifier of BMP4 signaling in trigeminal ganglion (TG) neurons. Loss of Megf8 disrupts axon guidance in the peripheral nervous system and leads to defects in development of the limb, heart, and left-right patterning, defects that resemble those observed in Bmp4 loss-of-function mice. Bmp4 is expressed in a pattern that defines the permissive field for the peripheral projections of TG axons and mice lacking BMP signaling in sensory neurons exhibit TG axon defects that resemble those observed in Megf8−/− embryos. Furthermore, TG axon growth is robustly inhibited by BMP4 and this inhibition is dependent on Megf8. Thus, our data suggest that Megf8 is involved in mediating BMP4 signaling and guidance of developing TG axons. DOI:http://dx.doi.org/10.7554/eLife.01160.001 The peripheral nervous system relays information between the brain and spinal cord (the central nervous system) and the rest of the body. During development, neurons of the peripheral nervous system must extend processes (axons) long distances to reach the cells that they will eventually form connections with. Signaling molecules tell neuronal processes which direction to move in, and also tell them when they have reached their intended destination. One group of molecules involved in the extension and guidance of neuronal processes are growth factors known as bone morphogenetic proteins (BMPs). These proteins contribute to a range of developmental processes, including the formation of the limbs and the skeleton, as well as various organs. They also help to establish the correct left-right patterning of the embryo, and direct the migration of sensory neurons. Now, Engelhard et al. have used a genetic screen to identify additional signaling molecules involved in the development of the peripheral nervous system. They screened mice with a range of mutations, and found that animals with a mutant form of the gene that codes for a protein called MEGF8 closely resembled mice that lacked a member of the BMP family, BMP4. These mutants showed abnormal development of the skeleton and heart, and had six or seven digits on each limb (polydactyly). Given the similarities between mice that lacked the gene for BMP4 and those that lacked the gene for MEGF8, Engelhard et al. explored these parallels further, and the results of a series of experiments were consistent with the two proteins being part of the same signaling cascade. In addition to identifying a novel signaling molecule that is involved in the formation of the peripheral nervous system, Engelhard et al. have provided new insights into the mechanisms by which one of the best known developmental signaling cascades is regulated. DOI:http://dx.doi.org/10.7554/eLife.01160.002
Collapse
Affiliation(s)
- Caitlin Engelhard
- The Solomon H Snyder Department of Neuroscience , Howard Hughes Medical Institute, Johns Hopkins University School of Medicine , Baltimore , United States
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Audouard E, Schakman O, Ginion A, Bertrand L, Gailly P, Clotman F. The Onecut transcription factor HNF-6 contributes to proper reorganization of Purkinje cells during postnatal cerebellum development. Mol Cell Neurosci 2013; 56:159-68. [PMID: 23669529 DOI: 10.1016/j.mcn.2013.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/18/2013] [Accepted: 05/02/2013] [Indexed: 01/05/2023] Open
Abstract
The Onecut (OC) family of transcription factors comprises three members in mammals, namely HNF-6 (or OC-1), OC-2 and OC-3. During embryonic development, these transcriptional activators control cell differentiation in pancreas, in liver and in the nervous system. Adult Hnf6 mutant mice exhibit locomotion defects characterized by hindlimb muscle weakness, abnormal gait and defective balance and coordination. Indeed, HNF-6 is required in spinal motor neurons for proper formation of the hindlimb neuromuscular junctions, which likely explain muscle weakness observed in corresponding mutant animals. The goal of the present study was to determine the cause of the balance and coordination defects in Hnf6 mutant mice. Coordination and balance deficits were quantified by rotarod and runway tests. Hnf6 mutant animals showed an increase in the fall frequency from the beam and were unable to stay on the rotarod even at low speed, indicating a severe balance and coordination deficit. To identify the origin of this abnormality, we assessed whether the development of the main CNS structure involved in the control of balance and coordination, namely the cerebellum, was affected by the absence of HNF-6. Firstly, we observed that Hnf6 was expressed transiently during the first week after birth in the Purkinje cells of wild type newborn mice. Secondly, we showed that, in Hnf6-/- mice, the organization of Purkinje cells became abnormal during a second phase of their development. Indeed, Purkinje cells were produced normally but part of them failed to reorganize as a regular continuous monolayer at the interface between the molecular and the granular layer of the cerebellum. Thus, the Onecut factor HNF-6 contributes to the reorganization of Purkinje cells during a late phase of cerebellar development.
Collapse
Affiliation(s)
- Emilie Audouard
- Université catholique de Louvain, Institute of Neuroscience, Laboratory of Neural Differentiation, Brussels, Belgium
| | | | | | | | | | | |
Collapse
|
50
|
Kelly CE, Thymiakou E, Dixon JE, Tanaka S, Godwin J, Episkopou V. Rnf165/Ark2C enhances BMP-Smad signaling to mediate motor axon extension. PLoS Biol 2013; 11:e1001538. [PMID: 23610558 PMCID: PMC3627648 DOI: 10.1371/journal.pbio.1001538] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 03/07/2013] [Indexed: 12/21/2022] Open
Abstract
Little is known about extrinsic signals required for the advancement of motor neuron (MN) axons, which extend over long distances in the periphery to form precise connections with target muscles. Here we present that Rnf165 (Arkadia-like; Arkadia2; Ark2C) is expressed specifically in the nervous system and that its loss in mice causes motor innervation defects that originate during development and lead to wasting and death before weaning. The defects range from severe reduction of motor axon extension as observed in the dorsal forelimb to shortening of presynaptic branches of the phrenic nerve, as observed in the diaphragm. Molecular functional analysis showed that in the context of the spinal cord Ark2C enhances transcriptional responses of the Smad1/5/8 effectors, which are activated (phosphorylated) downstream of Bone Morphogenetic Protein (BMP) signals. Consistent with Ark2C-modulated BMP signaling influencing motor axons, motor pools in the spinal cord were found to harbor phosphorylated Smad1/5/8 (pSmad) and treatment of primary MN with BMP inhibitor diminished axon length. In addition, genetic reduction of BMP-Smad signaling in Ark2C (+/-) mice caused the emergence of Ark2C (-/-) -like dorsal forelimb innervation deficits confirming that enhancement of BMP-Smad responses by Ark2C mediates efficient innervation. Together the above data reveal an involvement of BMP-Smad signaling in motor axon advancement.
Collapse
Affiliation(s)
- Claire E. Kelly
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Efstathia Thymiakou
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - James E. Dixon
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Shinya Tanaka
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Jonathan Godwin
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Vasso Episkopou
- Division of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|