1
|
Reiner A, Medina L, Abellan A, Deng Y, Toledo CA, Luksch H, Vega-Zuniga T, Riley NB, Hodos W, Karten HJ. Neurochemistry and circuit organization of the lateral spiriform nucleus of birds: A uniquely nonmammalian direct pathway component of the basal ganglia. J Comp Neurol 2024; 532:e25620. [PMID: 38733146 PMCID: PMC11090467 DOI: 10.1002/cne.25620] [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: 08/30/2023] [Revised: 03/24/2024] [Accepted: 04/16/2024] [Indexed: 05/13/2024]
Abstract
We used diverse methods to characterize the role of avian lateral spiriform nucleus (SpL) in basal ganglia motor function. Connectivity analysis showed that SpL receives input from globus pallidus (GP), and the intrapeduncular nucleus (INP) located ventromedial to GP, whose neurons express numerous striatal markers. SpL-projecting GP neurons were large and aspiny, while SpL-projecting INP neurons were medium sized and spiny. Connectivity analysis further showed that SpL receives inputs from subthalamic nucleus (STN) and substantia nigra pars reticulata (SNr), and that the SNr also receives inputs from GP, INP, and STN. Neurochemical analysis showed that SpL neurons express ENK, GAD, and a variety of pallidal neuron markers, and receive GABAergic terminals, some of which also contain DARPP32, consistent with GP pallidal and INP striatal inputs. Connectivity and neurochemical analysis showed that the SpL input to tectum prominently ends on GABAA receptor-enriched tectobulbar neurons. Behavioral studies showed that lesions of SpL impair visuomotor behaviors involving tracking and pecking moving targets. Our results suggest that SpL modulates brainstem-projecting tectobulbar neurons in a manner comparable to the demonstrated influence of GP internus on motor thalamus and of SNr on tectobulbar neurons in mammals. Given published data in amphibians and reptiles, it seems likely the SpL circuit represents a major direct pathway-type circuit by which the basal ganglia exerts its motor influence in nonmammalian tetrapods. The present studies also show that avian striatum is divided into three spatially segregated territories with differing connectivity, a medial striato-nigral territory, a dorsolateral striato-GP territory, and the ventrolateral INP motor territory.
Collapse
Affiliation(s)
- Anton Reiner
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163
| | - Loreta Medina
- Department of Experimental Medicine, Universitat de Lleida, Lleida, Spain
- Laboratory of Evolutionary and Developmental Neurobiology, Lleida’s Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Lleida, Catalonia, Spain
| | - Antonio Abellan
- Department of Experimental Medicine, Universitat de Lleida, Lleida, Spain
- Laboratory of Evolutionary and Developmental Neurobiology, Lleida’s Institute for Biomedical Research-Dr. Pifarré Foundation (IRBLleida), Lleida, Catalonia, Spain
| | - Yunping Deng
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Center, Memphis, TN 38163
| | - Claudio A.B. Toledo
- Neuroscience Research Nucleus, Universidade Cidade de Sao Paulo, Sao Paulo 65057-420, Brazil
| | - Harald Luksch
- School of Life Sciences, Technische Universität München, Freising-Weihenstephan, Germany
| | - Tomas Vega-Zuniga
- School of Life Sciences, Technische Universität München, Freising-Weihenstephan, Germany
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Nell B. Riley
- Department of Psychology, University of Maryland College Park 20742-4411
| | - William Hodos
- Department of Psychology, University of Maryland College Park 20742-4411
| | - Harvey J. Karten
- Department of Neurosciences, University of California San Diego, San Diego, CA 92093-0608
| |
Collapse
|
2
|
Herzog H, Glöckler S, Flamm J, Ladel S, Maigler F, Pitzer C, Schindowski K. Intranasal Nose-to-Brain Drug Delivery via the Olfactory Region in Mice: Two In-Depth Protocols for Region-Specific Intranasal Application of Antibodies and for Expression Analysis of Fc Receptors via In Situ Hybridization in the Nasal Mucosa. Methods Mol Biol 2024; 2754:387-410. [PMID: 38512678 DOI: 10.1007/978-1-0716-3629-9_21] [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] [Indexed: 03/23/2024]
Abstract
A region-specific catheter-based intranasal administration method was successfully developed, established, and validated as reported previously. By using this method, drugs can be applicated specifically to the olfactory region. Thereby, intranasally administered drugs could be delivered via neuronal connections to the central nervous system. Here, we present a detailed protocol with a step-by-step procedure for nose-to-brain delivery via the olfactory mucosa.Fc receptors such as the neonatal Fc receptor (FcRn) and potentially Fcγ receptor IIb (FcγRIIb) are involved in the uptake and transport of antibodies via the olfactory nasal mucosa. To better characterize their expression levels and their role in CNS drug delivery via the nose, an in situ hybridization (ISH) protocol was adapted for nasal mucosa samples and described in abundant details.
Collapse
Affiliation(s)
- Helena Herzog
- Institute of Applied Biotechnology, University of Applied Science Biberach, Biberach, Germany
- Faculty of Natural Science, University of Ulm, Ulm, Germany
| | - Sara Glöckler
- Institute of Applied Biotechnology, University of Applied Science Biberach, Biberach, Germany
- Faculty of Natural Science, University of Ulm, Ulm, Germany
| | - Johannes Flamm
- Institute of Applied Biotechnology, University of Applied Science Biberach, Biberach, Germany
- Faculty of Natural Science, University of Ulm, Ulm, Germany
| | - Simone Ladel
- Institute of Applied Biotechnology, University of Applied Science Biberach, Biberach, Germany
- Faculty of Natural Science, University of Ulm, Ulm, Germany
| | - Frank Maigler
- Institute of Applied Biotechnology, University of Applied Science Biberach, Biberach, Germany
- Faculty of Natural Science, University of Ulm, Ulm, Germany
| | - Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Heidelberg University, Heidelberg, Germany
| | - Katharina Schindowski
- Institute of Applied Biotechnology, University of Applied Science Biberach, Biberach, Germany.
| |
Collapse
|
3
|
Schindowski K. Differential Regulation of Neurotrophic Factors During Pathogenic Tau-Aggregation in a Tau Transgenic Mouse Model for Alzheimer's Disease: A Protocol for Double-Labeling mRNA by In Situ Hybridization and Protein Epitopes by Immunohistochemistry. Methods Mol Biol 2024; 2754:361-385. [PMID: 38512677 DOI: 10.1007/978-1-0716-3629-9_20] [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] [Indexed: 03/23/2024]
Abstract
Alzheimer's disease (AD), most tauopathies, and other neurodegenerative diseases are highly associated to impaired neurotrophin regulation and imbalanced neurotrophin transport and distribution. Neurotrophins are crucial for the survival and maintenance of distinct neuronal population therefore their supply is essential for a healthy brain. Tau phosphorylation occurs at different sites of the tau protein and some phospho-epitopes are highly associated to AD (e.g., abnormally phosphorylated tau at Thr212/Ser214). Though the importance of neurotrophins is well known, their analysis in tissue is not trivial and needs careful consideration. Here a detailed protocol is presented, which combines in situ hybridization (ISH) with immunohistochemistry (IHC) to analyze neurotrophin mRNA expression during tau neuropathology and the results were confirmed by immunological methods.With this protocol, it was demonstrated that Brain-Derived Neurotrophic Factor (BDNF) and its receptor Tropomyosin receptor kinase B (TrkB) were significantly decreased in tau-transgenic mice compared to their age-matched littermates. Neurotrophin-3 (NT-3) and its receptor TrkC were not altered with statistical significance, but a tendency for decreased NT-3 and slightly increased TrkC expression was observed in tau transgenic mice. The loss of BDNF-ISH signal was predominantly observed in hippocampus (CA1 and CA3) and cortex (layer II-VI) and verified by BDNF-immunoreactivity. Decreased BDNF and TrkB mRNA was negatively correlated with abnormal tau phosphorylation at Thr212/Ser214 in cortical neurons in transgenic mice. Strikingly, no correlation was observed with age-related phospho-epitopes such as Ser202/Thr205. Interestingly, both, the mRNA and protein levels of Nerve Growth Factor (NGF) were significantly increased in hippocampal neurons in the tau models as demonstrated by ISH, immunofluorescence, and Western Blotting. Here, some co-localization of NGF mRNA and phospho-tau (Thr212/Ser214) was observed but was a rare event. Since there is growing evidence for the relevance of neurotrophic factor distribution in the pathogenesis of neurodegeneration, this technique is a useful tool to investigate the underlying mechanisms and potential therapeutic intervention.
Collapse
Affiliation(s)
- Katharina Schindowski
- Institute of Applied Biotechnology, University of Applied Science Biberach, Biberach, Germany.
| |
Collapse
|
4
|
Masliukov PM, Emanuilov AI, Budnik AF. Sympathetic innervation of the development, maturity, and aging of the gastrointestinal tract. Anat Rec (Hoboken) 2023; 306:2249-2263. [PMID: 35762574 DOI: 10.1002/ar.25015] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 04/21/2022] [Accepted: 05/24/2022] [Indexed: 11/10/2022]
Abstract
The sympathetic nervous system inhibits gut motility, secretion, and blood flow in the gut microvasculature and can modulate gastrointestinal inflammation. Sympathetic neurons signal via catecholamines, neuropeptides, and gas mediators. In the current review, we summarize the current understanding of the mature sympathetic innervation of the gastrointestinal tract with a focus mainly on the prevertebral sympathetic ganglia as the main output to the gut. We also highlight recent work regarding the developmental processes of sympathetic innervation. The anatomy, neurochemistry, and connections of the sympathetic prevertebral ganglia with different parts of the gut are considered in adult organisms during prenatal and postnatal development and aging. The processes and mechanisms that control the development of sympathetic neurons, including their migratory pathways, neuronal differentiation, and aging, are reviewed.
Collapse
Affiliation(s)
- Petr M Masliukov
- Department of Normal Physiology, Yaroslavl State Medical University, Yaroslavl, Russia
| | - Andrey I Emanuilov
- Department of Human Anatomy, Yaroslavl State Medical University, Yaroslavl, Russia
| | - Antonina F Budnik
- Department of Normal and Pathological Anatomy, Kabardino-Balkarian State University named after H.M. Berbekov, Nalchik, Russia
| |
Collapse
|
5
|
Ernsberger U, Deller T, Rohrer H. The sympathies of the body: functional organization and neuronal differentiation in the peripheral sympathetic nervous system. Cell Tissue Res 2021; 386:455-475. [PMID: 34757495 PMCID: PMC8595186 DOI: 10.1007/s00441-021-03548-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023]
Abstract
During the last 30 years, our understanding of the development and diversification of postganglionic sympathetic neurons has dramatically increased. In parallel, the list of target structures has been critically extended from the cardiovascular system and selected glandular structures to metabolically relevant tissues such as white and brown adipose tissue, lymphoid tissues, bone, and bone marrow. A critical question now emerges for the integration of the diverse sympathetic neuron classes into neural circuits specific for these different target tissues to achieve the homeostatic regulation of the physiological ends affected.
Collapse
Affiliation(s)
- Uwe Ernsberger
- Institute for Clinical Neuroanatomy, Goethe University, Frankfurt/Main, Germany.
| | - Thomas Deller
- Institute for Clinical Neuroanatomy, Goethe University, Frankfurt/Main, Germany
| | - Hermann Rohrer
- Institute for Clinical Neuroanatomy, Goethe University, Frankfurt/Main, Germany.
| |
Collapse
|
6
|
The diversity of neuronal phenotypes in rodent and human autonomic ganglia. Cell Tissue Res 2020; 382:201-231. [PMID: 32930881 PMCID: PMC7584561 DOI: 10.1007/s00441-020-03279-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 08/10/2020] [Indexed: 12/29/2022]
Abstract
Selective sympathetic and parasympathetic pathways that act on target organs represent the terminal actors in the neurobiology of homeostasis and often become compromised during a range of neurodegenerative and traumatic disorders. Here, we delineate several neurotransmitter and neuromodulator phenotypes found in diverse parasympathetic and sympathetic ganglia in humans and rodent species. The comparative approach reveals evolutionarily conserved and non-conserved phenotypic marker constellations. A developmental analysis examining the acquisition of selected neurotransmitter properties has provided a detailed, but still incomplete, understanding of the origins of a set of noradrenergic and cholinergic sympathetic neuron populations, found in the cervical and trunk region. A corresponding analysis examining cholinergic and nitrergic parasympathetic neurons in the head, and a range of pelvic neuron populations, with noradrenergic, cholinergic, nitrergic, and mixed transmitter phenotypes, remains open. Of particular interest are the molecular mechanisms and nuclear processes that are responsible for the correlated expression of the various genes required to achieve the noradrenergic phenotype, the segregation of cholinergic locus gene expression, and the regulation of genes that are necessary to generate a nitrergic phenotype. Unraveling the neuron population-specific expression of adhesion molecules, which are involved in axonal outgrowth, pathway selection, and synaptic organization, will advance the study of target-selective autonomic pathway generation.
Collapse
|
7
|
Selective Induction of Human Autonomic Neurons Enables Precise Control of Cardiomyocyte Beating. Sci Rep 2020; 10:9464. [PMID: 32528170 PMCID: PMC7289887 DOI: 10.1038/s41598-020-66303-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022] Open
Abstract
The autonomic nervous system (ANS) regulates tissue homeostasis and remodelling through antagonistic effects of noradrenergic sympathetic and cholinergic parasympathetic signalling. Despite numerous reports on the induction of sympathetic neurons from human pluripotent stem cells (hPSCs), no induction methods have effectively derived cholinergic parasympathetic neurons from hPSCs. Considering the antagonistic effects of noradrenergic and cholinergic inputs on target organs, both sympathetic and parasympathetic neurons are expected to be induced. This study aimed to develop a stepwise chemical induction method to induce sympathetic-like and parasympathetic-like ANS neurons. Autonomic specification was achieved through restricting signals inducing sensory or enteric neurogenesis and activating bone morphogenetic protein (BMP) signals. Global mRNA expression analyses after stepwise induction, including single-cell RNA-seq analysis of induced neurons and functional assays revealed that each induced sympathetic-like or parasympathetic-like neuron acquired pharmacological and electrophysiological functional properties with distinct marker expression. Further, we identified selective induction methods using appropriate seeding cell densities and neurotrophic factor concentrations. Neurons were individually induced, facilitating the regulation of the beating rates of hiPSC-derived cardiomyocytes in an antagonistic manner. The induction methods yield specific neuron types, and their influence on various tissues can be studied by co-cultured assays.
Collapse
|
8
|
Ernsberger U, Rohrer H. Sympathetic tales: subdivisons of the autonomic nervous system and the impact of developmental studies. Neural Dev 2018; 13:20. [PMID: 30213267 PMCID: PMC6137933 DOI: 10.1186/s13064-018-0117-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 08/12/2018] [Indexed: 02/06/2023] Open
Abstract
Remarkable progress in a range of biomedical disciplines has promoted the understanding of the cellular components of the autonomic nervous system and their differentiation during development to a critical level. Characterization of the gene expression fingerprints of individual neurons and identification of the key regulators of autonomic neuron differentiation enables us to comprehend the development of different sets of autonomic neurons. Their individual functional properties emerge as a consequence of differential gene expression initiated by the action of specific developmental regulators. In this review, we delineate the anatomical and physiological observations that led to the subdivision into sympathetic and parasympathetic domains and analyze how the recent molecular insights melt into and challenge the classical description of the autonomic nervous system.
Collapse
Affiliation(s)
- Uwe Ernsberger
- Institute for Clinical Neuroanatomy, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany
| | - Hermann Rohrer
- Institute for Clinical Neuroanatomy, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt/Main, Germany
| |
Collapse
|
9
|
El Faitwri T, Huber K. Expression pattern of delta-like 1 homolog in developing sympathetic neurons and chromaffin cells. Gene Expr Patterns 2018; 30:49-54. [PMID: 30144579 DOI: 10.1016/j.gep.2018.08.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 08/21/2018] [Accepted: 08/21/2018] [Indexed: 02/03/2023]
Abstract
Delta-like 1 homolog (DLK1) is a member of the epidermal growth factor (EGF)-like family and an atypical notch ligand that is widely expressed during early mammalian development with putative functions in the regulation of cell differentiation and proliferation. During later stages of development, DLK1 is downregulated and becomes increasingly restricted to specific cell types, including several types of endocrine cells. DLK1 has been linked to various tumors and associated with tumor stem cell features. Sympathoadrenal precursors are neural crest derived cells that give rise to either sympathetic neurons of the autonomic nervous system or the endocrine chromaffin cells located in the adrenal medulla or extraadrenal positions. As these cells are the putative cellular origin of neuroblastoma, one of the most common malignant tumors in early childhood, their molecular characterization is of high clinical importance. In this study we have examined the precise spatiotemporal expression of DLK1 in developing sympathoadrenal cells. We show that DLK1 mRNA is highly expressed in early sympathetic neuron progenitors and that its expression depends on the presence of Phox2B. DLK1 expression becomes quickly restricted to a small subpopulation of cells in sympathetic ganglia, while virtually all chromaffin cells in the adrenal medulla and the Organ of Zuckerkandl still express high levels of DLK1 at late gestational stages.
Collapse
Affiliation(s)
- Tehani El Faitwri
- Institute of Anatomy & Cell Biology, Albert-Ludwigs-University Freiburg, Albert-Str. 17, 79104, Freiburg, Germany; Department of Histology and Anatomy, Faculty of Medicine, Benghazi University, Benghazi, Libya
| | - Katrin Huber
- Institute of Anatomy & Cell Biology, Albert-Ludwigs-University Freiburg, Albert-Str. 17, 79104, Freiburg, Germany; Department of Medicine, University of Fribourg, Route Albert-Gockel 1, 1700, Fribourg, Switzerland.
| |
Collapse
|
10
|
Koszinowski S, La Padula V, Edlich F, Krieglstein K, Busch H, Boerries M. Bid Expression Network Controls Neuronal Cell Fate During Avian Ciliary Ganglion Development. Front Physiol 2018; 9:797. [PMID: 30008673 PMCID: PMC6034111 DOI: 10.3389/fphys.2018.00797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 06/07/2018] [Indexed: 11/13/2022] Open
Abstract
Avian ciliary ganglion (CG) development involves a transient execution phase of apoptosis controlling the final number of neurons, but the time-dependent molecular mechanisms for neuronal cell fate are largely unknown. To elucidate the molecular networks regulating important aspects of parasympathetic neuronal development, a genome-wide expression analysis was performed during multiple stages of avian CG development between embryonic days E6 and E14. The transcriptome data showed a well-defined sequence of events, starting from neuronal migration via neuronal fate cell determination, synaptic transmission, and regulation of synaptic plasticity to growth factor associated signaling. In particular, we extracted a neuronal apoptosis network that characterized the cell death execution phase at E8/E9 and apoptotic cell clearance at E14 by combining the gene time series analysis with network synthesis from the chicken interactome. Network analysis identified TP53 as key regulator and predicted involvement of the BH3 interacting domain death agonist (BID). A virus-based RNAi knockdown approach in vivo showed a crucial impact of BID expression on the execution of ontogenetic programmed cell death (PCD). In contrast, Bcl-XL expression did not impact PCD. Therefore, BID-mediated apoptosis represents a novel cue essential for timing within CG maturation.
Collapse
Affiliation(s)
- Sophie Koszinowski
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Veronica La Padula
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Frank Edlich
- Institute for Biochemistry and Molecular Biology, and Centre for Biological Signalling Studies BIOSS, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Kerstin Krieglstein
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Hauke Busch
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,Luebeck Institute for Experimental Dermatology, University of Lübeck, Lübeck, Germany.,Institute for Cardiogenetics, University of Lübeck, Lübeck, Germany
| | - Melanie Boerries
- Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
The dystrophin isoform Dp71e Δ71 is involved in neurite outgrowth and neuronal differentiation of PC12 cells. J Proteomics 2018; 191:80-87. [PMID: 29625189 DOI: 10.1016/j.jprot.2018.03.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/19/2018] [Accepted: 03/25/2018] [Indexed: 11/22/2022]
Abstract
The Dp71 protein is the most abundant dystrophin in the central nervous system (CNS). Several dystrophin Dp71 isoforms have been described and are classified into three groups, each with a different C-terminal end. However, the functions of Dp71 isoforms remain unknown. In the present study, we analysed the effect of Dp71eΔ71 overexpression on neuronal differentiation of PC12 Tet-On cells. Overexpression of dystrophin Dp71eΔ71 stimulates neuronal differentiation, increasing the percentage of cells with neurites and neurite length. According to 2-DE analysis, Dp71eΔ71 overexpression modified the protein expression profile of rat pheochromocytoma PC12 Tet-On cells that had been treated with neuronal growth factor (NGF) for nine days. Interestingly, all differentially expressed proteins were up-regulated compared to the control. The proteomic analysis showed that Dp71eΔ71 increases the expression of proteins with important roles in the differentiation process, such as HspB1, S100A6, and K8 proteins involved in the cytoskeletal structure and HCNP protein involved in neurotransmitter synthesis. The expression of neuronal marker TH was also up-regulated. Mass spectrometry data are available via ProteomeXchange with identifier PXD009114. SIGNIFICANCE: This study is the first to explore the role of the specific isoform Dp71eΔ71. The results obtained here support the hypothesis that the dystrophin Dp71eΔ71 isoform has an important role in the neurite outgrowth by regulating the levels of proteins involved in the cytoskeletal structure, such as HspB1, S100A6, and K8, and in neurotransmitter synthesis, such as HCNP and TH, biological processes required to stimulate neuronal differentiation.
Collapse
|
12
|
Regulation of Neurotrophic Factors During Pathogenic Tau-Aggregation: A Detailed Protocol for Double-Labeling mRNA by In Situ Hybridization and Protein Epitopes by Immunohistochemistry. Methods Mol Biol 2018. [PMID: 27975267 DOI: 10.1007/978-1-4939-6598-4_27] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025]
Abstract
Alzheimer's disease (AD), most tauopathies, and other neurodegenerative diseases are highly associated with impaired neurotrophin regulation and imbalanced neutrophin distribution. Tau phosphorylation occurs at different sites of the tau protein and some phospho-epitopes are associated with normal ageing (like tau phosphorylated at Ser202/Thr205 detected by the antibody clone AT8) and others are highly associated with AD (abnormally phosphorylated tau at Thr212/Ser214 detected by the antibody clone AT100). Neurotrophins are crucial for the survival and maintenance of distinct neuronal population; therefore, their supply is essential for a healthy brain. Though their importance is well known, their analysis in tissue is not trivial and needs careful consideration. Here, a detailed a protocol is presented, how to combine in situ hybridization (ISH) with immunohistochemistry (IHC) to analyze neurotrophins during tau neuropathology and the results were confirmed by immunological methods. In addition, the preparation of the riboprobes is presented step-by-step. Since there are growing evidences for the relevance of neurotrophic factor distribution in the pathogenesis of AD, this technique is one useful tool to investigate the underlying mechanisms and therapeutic intervention.
Collapse
|
13
|
Long-term impact of intrauterine neuroinflammation and treatment with magnesium sulphate and betamethasone: Sex-specific differences in a preterm labor murine model. Sci Rep 2017; 7:17883. [PMID: 29263436 PMCID: PMC5738437 DOI: 10.1038/s41598-017-18197-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/06/2017] [Indexed: 11/17/2022] Open
Abstract
Preterm infants are at significantly increased risk for lifelong neurodevelopmental disability with male offspring disproportionately affected. Corticosteroids (such as betamethasone) and magnesium sulphate (MgSO4) are administered to women in preterm labor to reduce neurologic morbidity. Despite widespread use of MgSO4 in clinical practice, its effects on adult offspring are not well known nor have sex-specific differences in therapeutic response been explored. The objective of our study was to examine the long-term effects of perinatal neuroinflammation and the effectiveness of prenatal MgSO4/betamethasone treatments between males and females in a murine model via histologic and expression analyses. Our results demonstrate that male but not female offspring exposed to intrauterine inflammation demonstrated impaired performance in neurodevelopmental testing in early life assessed via negative geotaxis, while those exposed to injury plus treatment fared better. Histologic analysis of adult male brains identified a significant reduction in hippocampal neural density in the injured group compared to controls. Evaluation of key neural markers via qRT-PCR demonstrated more profound differences in gene expression in adult males exposed to injury and treatment compared to female offspring, which largely showed resistance to injury. Prenatal treatment with MgSO4/betamethasone confers long-term benefits beyond cerebral palsy prevention with sex-specific differences in response.
Collapse
|
14
|
Massa MG, Gisevius B, Hirschberg S, Hinz L, Schmidt M, Gold R, Prochnow N, Haghikia A. Multiple Sclerosis Patient-Specific Primary Neurons Differentiated from Urinary Renal Epithelial Cells via Induced Pluripotent Stem Cells. PLoS One 2016; 11:e0155274. [PMID: 27158987 PMCID: PMC4861271 DOI: 10.1371/journal.pone.0155274] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 04/26/2016] [Indexed: 12/11/2022] Open
Abstract
As multiple sclerosis research progresses, it is pertinent to continue to develop suitable paradigms to allow for ever more sophisticated investigations. Animal models of multiple sclerosis, despite their continuing contributions to the field, may not be the most prudent for every experiment. Indeed, such may be either insufficient to reflect the functional impact of human genetic variations or unsuitable for drug screenings. Thus, we have established a cell- and patient-specific paradigm to provide an in vitro model within which to perform future genetic investigations. Renal proximal tubule epithelial cells were isolated from multiple sclerosis patients’ urine and transfected with pluripotency-inducing episomal factors. Subsequent induced pluripotent stem cells were formed into embryoid bodies selective for ectodermal lineage, resulting in neural tube-like rosettes and eventually neural progenitor cells. Differentiation of these precursors into primary neurons was achieved through a regimen of neurotrophic and other factors. These patient-specific primary neurons displayed typical morphology and functionality, also staining positive for mature neuronal markers. The development of such a non-invasive procedure devoid of permanent genetic manipulation during the course of differentiation, in the context of multiple sclerosis, provides an avenue for studies with a greater cell- and human-specific focus, specifically in the context of genetic contributions to neurodegeneration and drug discovery.
Collapse
Affiliation(s)
- Megan G Massa
- Neurologische Klinik der Ruhr-Universität Bochum, St. Josef-Hospital, Bochum, Germany
| | - Barbara Gisevius
- Neurologische Klinik der Ruhr-Universität Bochum, St. Josef-Hospital, Bochum, Germany
| | - Sarah Hirschberg
- Neurologische Klinik der Ruhr-Universität Bochum, St. Josef-Hospital, Bochum, Germany
| | - Lisa Hinz
- Neurologische Klinik der Ruhr-Universität Bochum, St. Josef-Hospital, Bochum, Germany
| | - Matthias Schmidt
- Department of Neuroanatomy, Ruhr-Universität Bochum, Bochum, Germany
| | - Ralf Gold
- Neurologische Klinik der Ruhr-Universität Bochum, St. Josef-Hospital, Bochum, Germany
| | - Nora Prochnow
- Department of Neuroanatomy, Ruhr-Universität Bochum, Bochum, Germany
| | - Aiden Haghikia
- Neurologische Klinik der Ruhr-Universität Bochum, St. Josef-Hospital, Bochum, Germany
| |
Collapse
|
15
|
MiR-124 is differentially expressed in derivatives of the sympathoadrenal cell lineage and promotes neurite elongation in chromaffin cells. Cell Tissue Res 2016; 365:225-32. [DOI: 10.1007/s00441-016-2395-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 03/09/2016] [Indexed: 01/02/2023]
|
16
|
Abstract
UNLABELLED The RNA binding protein Lin28B is expressed in developing tissues and sustains stem and progenitor cell identity as a negative regulator of the Let-7 family of microRNAs, which induces differentiation. Lin28B is activated in neuroblastoma (NB), a childhood tumor in sympathetic ganglia and adrenal medulla. Forced expression of Lin28B in embryonic mouse sympathoadrenal neuroblasts elicits postnatal NB formation. However, the normal function of Lin28B in the development of sympathetic neurons and chromaffin cells and the mechanisms involved in Lin28B-induced tumor formation are unclear. Here, we demonstrate a mirror-image expression of Lin28B and Let-7a in developing chick sympathetic ganglia. Lin28B expression is not restricted to undifferentiated progenitor cells but, is observed in proliferating noradrenergic neuroblasts. Lin28 knockdown in cultured sympathetic neuroblasts decreases proliferation, whereas Let-7 inhibition increases the proportion of neuroblasts in the cell cycle. Lin28B overexpression enhances proliferation, but only during a short developmental period, and it does not reduce Let-7a. Effects of in vivo Lin28B overexpression were analyzed in the LSL-Lin28B(DBHiCre) mouse line. Sympathetic ganglion and adrenal medulla volume and the expression level of Let-7a were not altered, although Lin28B expression increased by 12- to 17-fold. In contrast, Let-7a expression was strongly reduced in LSL-Lin28B(DbhiCre) NB tumor tissue. These data demonstrate essential functions for endogenous Lin28 and Let-7 in neuroblast proliferation. However, Lin28B overexpression neither sustains neuroblast proliferation nor affects let-7 expression. Thus, in contrast to other pediatric tumors, Lin28B-induced NB is not due to expansion of proliferating embryonic neuroblasts, and Let-7-independent functions are implicated during initial NB development. SIGNIFICANCE STATEMENT Lin28A/B proteins are highly expressed in early development and maintain progenitor cells by blocking the biogenesis and differentiation function of Let-7 microRNAs. Lin28B is aberrantly upregulated in the childhood tumor neuroblastoma (NB). NB develops in sympathetic ganglia and adrenal medulla and is elicited by forced Lin28B expression. We demonstrate that Lin28A/B and Let-7 are essential for sympathetic neuroblast proliferation during normal development. Unexpectedly, Lin28B upregulation in a mouse model does not affect neuroblast proliferation, ganglion size, and Let-7 expression during early postnatal development. Lin28B-induced NB, in contrast to other pediatric cancers, does not evolve from neuroblasts that continue to divide and involves Let-7-independent functions during initial development.
Collapse
|
17
|
Stanzel S, Stubbusch J, Pataskar A, Howard MJ, Deller T, Ernsberger U, Tiwari VK, Rohrer H, Tsarovina K. Distinct roles of hand2 in developing and adult autonomic neurons. Dev Neurobiol 2016; 76:1111-24. [PMID: 26818017 DOI: 10.1002/dneu.22378] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Revised: 01/05/2016] [Accepted: 01/07/2016] [Indexed: 11/08/2022]
Abstract
The bHLH transcription factor Hand2 is essential for the acquisition and maintenance of noradrenergic properties of embryonic sympathetic neurons and controls neuroblast proliferation. Hand2 is also expressed in embryonic and postnatal parasympathetic ganglia and remains expressed in sympathetic neurons up to the adult stage. Here, we address its function in developing parasympathetic and adult sympathetic neurons. We conditionally deleted Hand2 in the parasympathetic sphenopalatine ganglion by crossing a line of floxed Hand2 mice with DbhiCre transgenic mice, taking advantage of the transient Dbh expression in parasympathetic ganglia. Hand2 elimination does not affect Dbh expression and sphenopalatine ganglion size at E12.5 and E16.5, in contrast to sympathetic ganglia. These findings demonstrate different functions for Hand2 in the parasympathetic and sympathetic lineage. Our previous Hand2 knockdown in postmitotic, differentiated chick sympathetic neurons resulted in decreased expression of noradrenergic marker genes but it was unclear whether Hand2 is required for maintaining noradrenergic neuron identity in adult animals. We now show that Hand2 elimination in adult Dbh-expressing sympathetic neurons does not decrease the expression of Th and Dbh, in contrast to the situation during development. However, gene expression profiling of adult sympathetic neurons identified 75 Hand2-dependent target genes. Interestingly, a notable proportion of down-regulated genes (15%) encode for proteins with synaptic and neurotransmission functions. These results demonstrate a change in Hand2 target genes during maturation of sympathetic neurons. Whereas Hand2 controls genes regulating noradrenergic differentiation during development, Hand2 seems to be involved in the regulation of genes controlling neurotransmission in adult sympathetic neurons. © 2016 Wiley Periodicals, Inc. Develop Neurobiol 76: 1111-1124, 2016.
Collapse
Affiliation(s)
- Sabine Stanzel
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany
| | - Jutta Stubbusch
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany
| | - Abhijeet Pataskar
- Institute of Molecular Biology (IMB) Boehringer Ingelheim Foundation, Ackermannweg 4, Mainz, 55128, Germany
| | - Marthe J Howard
- Department of Neurosciences and Program in Neurosciences and Neurological Disorders, University of Toledo Health Sciences Campus, Toledo, Ohio, 43614
| | - Thomas Deller
- Institute of Clinical Neuroanatomy, Goethe University Frankfurt/M, Theodor-Stern-Kai 7, Frankfurt/M, 60590, Germany
| | - Uwe Ernsberger
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany.,Institute of Clinical Neuroanatomy, Goethe University Frankfurt/M, Theodor-Stern-Kai 7, Frankfurt/M, 60590, Germany.,Ernst-Strüngmann-Institute, Deutschordenstr. 46, Frankfurt/M, 60528, Germany
| | - Vijay K Tiwari
- Institute of Molecular Biology (IMB) Boehringer Ingelheim Foundation, Ackermannweg 4, Mainz, 55128, Germany
| | - Hermann Rohrer
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany.,Institute of Clinical Neuroanatomy, Goethe University Frankfurt/M, Theodor-Stern-Kai 7, Frankfurt/M, 60590, Germany.,Ernst-Strüngmann-Institute, Deutschordenstr. 46, Frankfurt/M, 60528, Germany
| | - Konstantina Tsarovina
- Developmental Neurobiology, Max-Planck-Institute for Brain Research, Max-von-Laue-Str. 4, Frankfurt/M, 60438, Germany
| |
Collapse
|
18
|
Koszinowski S, Boerries M, Busch H, Krieglstein K. RARβ regulates neuronal cell death and differentiation in the avian ciliary ganglion. Dev Neurobiol 2015; 75:1204-18. [PMID: 25663354 PMCID: PMC4832352 DOI: 10.1002/dneu.22278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 02/06/2023]
Abstract
Programmed cell death during chicken ciliary ganglion (CG) development is mostly discussed as an extrinsically regulated process, guided either by the establishment of a functional balance between preganglionic and postganglionic activity or the availability of target‐derived neurotrophic factors. We found that the expression of the gene coding for the nuclear retinoic acid receptor β (RARB) is transiently upregulated prior to and during the execution phase of cell death in the CG. Using retroviral vectors, the expression of RARB was knocked down during embryonic development in ovo. The knockdown led to a significant increase in CG neuron number after the cell death phase. BrdU injections and active caspase‐3 staining revealed that this increase in neuron number was due to an inhibition of apoptosis during the normal cell death phase. Furthermore, apoptotic neuron numbers were significantly increased at a stage when cell death is normally completed. While the cholinergic phenotype of the neurons remained unchanged after RARB knockdown, the expression of the proneural gene Cash1 was increased, but somatostatin‐like immunoreactivity, a hallmark of the mature choroid neuron population, was decreased. Taken together, these results point toward a delay in neuronal differentiation as well as cell death. The availability of nuclear retinoic acid receptor β (RARβ) and RARβ‐induced transcription of genes could therefore be a new intrinsic cue for the maturation of CG neurons and their predisposition to undergo cell death. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 75: 1204–1218, 2015
Collapse
Affiliation(s)
- Sophie Koszinowski
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg (ALU), Freiburg, Germany.,University of Freiburg, Faculty of Biology, Schaenzlestrasse 1, D-79104, Freiburg, Germany
| | - Melanie Boerries
- Institute of Molecular Medicine and Cell Research, Centre for Biochemistry und Molecular Cell Research (ZBMZ), University of Freiburg, ALU, Stefan-Meier-Str.17, Freiburg, Germany.,German Cancer Consortium (DKTK), Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hauke Busch
- Institute of Molecular Medicine and Cell Research, Centre for Biochemistry und Molecular Cell Research (ZBMZ), University of Freiburg, ALU, Stefan-Meier-Str.17, Freiburg, Germany.,German Cancer Consortium (DKTK), Freiburg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kerstin Krieglstein
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg (ALU), Freiburg, Germany
| |
Collapse
|
19
|
Stubbusch J, Narasimhan P, Hennchen M, Huber K, Unsicker K, Ernsberger U, Rohrer H. Lineage and stage specific requirement for Dicer1 in sympathetic ganglia and adrenal medulla formation and maintenance. Dev Biol 2015; 400:210-23. [PMID: 25661788 DOI: 10.1016/j.ydbio.2015.01.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 01/23/2015] [Accepted: 01/24/2015] [Indexed: 10/24/2022]
Abstract
The development of sympathetic neurons and chromaffin cells is differentially controlled at distinct stages by various extrinsic and intrinsic signals. Here we use conditional deletion of Dicer1 in neural crest cells and noradrenergic neuroblasts to identify stage specific functions in sympathoadrenal lineages. Conditional Dicer1 knockout in neural crest cells of Dicer1(Wnt1Cre) mice results in a rapid reduction in the size of developing sympathetic ganglia and adrenal medulla. In contrast, Dicer1 elimination in noradrenergic neuroblasts of Dicer1(DbhiCre) animals affects sympathetic neuron survival starting at late embryonic stages and chromaffin cells persist at least until postnatal week 1. A differential function of Dicer1 signaling for the development of embryonic noradrenergic and cholinergic sympathetic neurons is demonstrated by the selective increase in the expression of Tlx3 and the cholinergic marker genes VAChT and ChAT at E16.5. The number of Dbh, Th and TrkA expressing noradrenergic neurons is strongly decreased in Dicer1-deficient sympathetic ganglia at birth, whereas Tlx3(+)/ Ret(+) cholinergic neurons cells are spared from cell death. The postnatal death of chromaffin cells is preceded by the loss of Ascl1, mir-375 and Pnmt and an increase in the markers Ret and NF-M, which suggests that Dicer1 is required for the maintenance of chromaffin cell differentiation and survival. Taken together, these findings demonstrate distinct stage and lineage specific functions of Dicer1 signaling in differentiation and survival of sympathetic neurons and adrenal chromaffin cells.
Collapse
Affiliation(s)
- Jutta Stubbusch
- Max-Planck-Institute for Brain Research, Research Group Developmental Neurobiology, Max-von-Laue-Street 4, 60438 Frankfurt/Main, Germany
| | - Priyanka Narasimhan
- Albert-Ludwigs-University Freiburg, Institute of Anatomy& Cell Biology, Albert-Street 17, 79104 Freiburg, Germany
| | - Melanie Hennchen
- Max-Planck-Institute for Brain Research, Research Group Developmental Neurobiology, Max-von-Laue-Street 4, 60438 Frankfurt/Main, Germany
| | - Katrin Huber
- Albert-Ludwigs-University Freiburg, Institute of Anatomy& Cell Biology, Albert-Street 17, 79104 Freiburg, Germany
| | - Klaus Unsicker
- Albert-Ludwigs-University Freiburg, Institute of Anatomy& Cell Biology, Albert-Street 17, 79104 Freiburg, Germany
| | - Uwe Ernsberger
- Max-Planck-Institute for Brain Research, Research Group Developmental Neurobiology, Max-von-Laue-Street 4, 60438 Frankfurt/Main, Germany; Institute of Clinical Neuroanatomy, Goethe-University Frankfurt, Theodor-Stern-Kai 7, Frankfurt/Main, Germany
| | - Hermann Rohrer
- Max-Planck-Institute for Brain Research, Research Group Developmental Neurobiology, Max-von-Laue-Street 4, 60438 Frankfurt/Main, Germany; Institute of Clinical Neuroanatomy, Goethe-University Frankfurt, Theodor-Stern-Kai 7, Frankfurt/Main, Germany.
| |
Collapse
|
20
|
Koszinowski S, Buss K, Kaehlcke K, Krieglstein K. Signaling via the transcriptionally regulated activin receptor 2B is a novel mediator of neuronal cell death during chicken ciliary ganglion development. Int J Dev Neurosci 2015; 41:98-104. [PMID: 25660516 DOI: 10.1016/j.ijdevneu.2015.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 01/30/2015] [Accepted: 01/31/2015] [Indexed: 11/25/2022] Open
Abstract
The TGF-β ligand superfamily members activin A and BMP control important aspects of embryonic neuronal development and differentiation. Both are known to bind to activin receptor subtypes IIA (ActRIIA) and IIB, while in the avian ciliary ganglion (CG), so far only ActRIIA-expression has been described. We show that the expression of ACVR2B, coding for the ActRIIB, is tightly regulated during CG development and the knockdown of ACVR2B expression leads to a deregulation in the execution of neuronal apoptosis and therefore affects ontogenetic programmed cell death in vivo. While the differentiation of choroid neurons was impeded in the knockdown, pointing toward a reduction in activin A-mediated neural differentiation signaling, naturally occurring neuronal cell death in the CG was not prevented by follistatin treatment. Systemic injections of the BMP antagonist noggin, on the other hand, reduced the number of apoptotic neurons to a similar extent as ACVR2B knockdown. We therefore propose a novel pathway in the regulation of CG neuron ontogenetic programmed cell death, which could be mediated by BMP and signals via the ActRIIB.
Collapse
Affiliation(s)
- S Koszinowski
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, D-79104 Freiburg, Germany; Faculty of Biology, Albert-Ludwigs-University Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany.
| | - K Buss
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, D-79104 Freiburg, Germany
| | - K Kaehlcke
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, D-79104 Freiburg, Germany
| | - K Krieglstein
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology, Albert-Ludwigs-University Freiburg, D-79104 Freiburg, Germany.
| |
Collapse
|
21
|
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: 1.7] [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
|
22
|
Tlx3 controls cholinergic transmitter and Peptide phenotypes in a subset of prenatal sympathetic neurons. J Neurosci 2013; 33:10667-75. [PMID: 23804090 DOI: 10.1523/jneurosci.0192-13.2013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The embryonic sympathetic nervous system consists of predominantly noradrenergic neurons and a very small population of cholinergic neurons. Postnatal development further allows target-dependent switch of a subset of noradrenergic neurons into cholinergic phenotype. How embryonic cholinergic neurons are specified at the prenatal stages remains largely unknown. In this study, we found that the expression of transcription factor Tlx3 was progressively restricted to a small population of embryonic sympathetic neurons in mice. Immunostaining for vesicular acetylcholine transporter (VAChT) showed that Tlx3 was highly expressed in cholinergic neurons at the late embryonic stage E18.5. Deletion of Tlx3 resulted in the loss of Vacht expression at E18.5 but not E12.5. By contrast, Tlx3 was required for expression of the cholinergic peptide vasoactive intestinal polypeptide (VIP), and somatostatin (SOM) at both E12.5 and E18.5. Furthermore, we found that, at E18.5 these putative cholinergic neurons expressed glial cell line-derived neurotrophic factor family coreceptor Ret but not tyrosine hydroxylase (Ret(+)/TH(-)). Deletion of Tlx3 also resulted in disappearance of high-level Ret expression. Last, unlike Tlx3, Ret was required for the expression of VIP and SOM at E18.5 but not E12.5. Together, these results indicate that transcription factor Tlx3 is required for the acquisition of cholinergic phenotype at the late embryonic stage as well as the expression and maintenance of cholinergic peptides VIP and SOM throughout prenatal development of mouse sympathetic neurons.
Collapse
|
23
|
Stubbusch J, Narasimhan P, Huber K, Unsicker K, Rohrer H, Ernsberger U. Synaptic protein and pan-neuronal gene expression and their regulation by Dicer-dependent mechanisms differ between neurons and neuroendocrine cells. Neural Dev 2013; 8:16. [PMID: 23961995 PMCID: PMC3766641 DOI: 10.1186/1749-8104-8-16] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 07/19/2013] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Neurons in sympathetic ganglia and neuroendocrine cells in the adrenal medulla share not only their embryonic origin from sympathoadrenal precursors in the neural crest but also a range of functional features. These include the capacity for noradrenaline biosynthesis, vesicular storage and regulated release. Yet the regulation of neuronal properties in early neuroendocrine differentiation is a matter of debate and the developmental expression of the vesicle fusion machinery, which includes components found in both neurons and neuroendocrine cells, is not resolved. RESULTS Analysis of synaptic protein and pan-neuronal marker mRNA expression during mouse development uncovers profound differences between sympathetic neurons and adrenal chromaffin cells, which result in qualitatively similar but quantitatively divergent transcript profiles. In sympathetic neurons embryonic upregulation of synaptic protein mRNA follows early and persistent induction of pan-neuronal marker transcripts. In adrenal chromaffin cells pan-neuronal marker expression occurs only transiently and synaptic protein messages remain at distinctly low levels throughout embryogenesis. Embryonic induction of synaptotagmin I (Syt1) in sympathetic ganglia and postnatal upregulation of synaptotagmin VII (Syt7) in adrenal medulla results in a cell type-specific difference in isoform prevalence. Dicer 1 inactivation in catecholaminergic cells reduces high neuronal synaptic protein mRNA levels but not their neuroendocrine low level expression. Pan-neuronal marker mRNAs are induced in chromaffin cells to yield a more neuron-like transcript pattern, while ultrastructure is not altered. CONCLUSIONS Our study demonstrates that remarkably different gene regulatory programs govern the expression of synaptic proteins in the neuronal and neuroendocrine branch of the sympathoadrenal system. They result in overlapping but quantitatively divergent transcript profiles. Dicer 1-dependent regulation is required to establish high neuronal mRNA levels for synaptic proteins and to maintain repression of neurofilament messages in neuroendocrine cells.
Collapse
Affiliation(s)
- Jutta Stubbusch
- Max Planck Institute for Brain Research, Deutschordenstrasse 46 D-60528, Frankfurt, Germany.
| | | | | | | | | | | |
Collapse
|
24
|
Krück S, Nesemann J, Scaal M. Development of somites, muscle, and skeleton is independent of signals from the wolffian duct. Dev Dyn 2013; 242:941-8. [DOI: 10.1002/dvdy.23986] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 04/16/2013] [Accepted: 05/01/2013] [Indexed: 11/09/2022] Open
Affiliation(s)
| | - Johanna Nesemann
- Institute of Anatomy and Cell Biology; Department of Molecular Embryology; University of Freiburg; Freiburg; Germany
| | | |
Collapse
|
25
|
Huber K, Narasimhan P, Shtukmaster S, Pfeifer D, Evans SM, Sun Y. The LIM-Homeodomain transcription factor Islet-1 is required for the development of sympathetic neurons and adrenal chromaffin cells. Dev Biol 2013; 380:286-98. [PMID: 23648511 PMCID: PMC5544970 DOI: 10.1016/j.ydbio.2013.04.027] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 04/04/2013] [Accepted: 04/17/2013] [Indexed: 12/25/2022]
Abstract
Islet-1 is a LIM-Homeodomain transcription factor with important functions for the development of distinct neuronal and non-neuronal cell populations. We show here that Islet-1 acts genetically downstream of Phox2B in cells of the sympathoadrenal cell lineage and that the development of sympathetic neurons and chromaffin cells is impaired in mouse embryos with a conditional deletion of Islet-1 controlled by the wnt1 promotor. Islet-1 is not essential for the initial differentiation of sympathoadrenal cells, as indicated by the correct expression of pan-neuronal and catecholaminergic subtype specific genes in primary sympathetic ganglia of Islet-1 deficient mouse embryos. However, our data indicate that the subsequent survival of sympathetic neuron precursors and their differentiation towards TrkA expressing neurons depends on Islet-1 function. In contrast to spinal sensory neurons, sympathetic neurons of Islet-1 deficient mice did not display ectopic expression of genes normally present in the CNS. In Islet-1 deficient mouse embryos the numbers of chromaffin cells were only mildly reduced, in contrast to that of sympathetic neurons, but the initiation of the adrenaline synthesizing enzyme PNMT was abrogated and the expression level of chromogranin A was diminished. Microarray analysis revealed that developing chromaffin cells of Islet-1 deficient mice displayed normal expression levels of TH, DBH and the transcription factors Phox2B, Mash-1, Hand2, Gata3 and Insm1, but the expression levels of the transcription factors Gata2 and Hand1, and AP-2β were significantly reduced. Together our data indicate that Islet-1 is not essentially required for the initial differentiation of sympathoadrenal cells, but has an important function for the correct subsequent development of sympathetic neurons and chromaffin cells.
Collapse
Affiliation(s)
- Katrin Huber
- Institute for Anatomy and Cell Biology, Department of Molecular Embryology, Albert-Ludwigs-University, Freiburg, Germany.
| | | | | | | | | | | |
Collapse
|
26
|
Schober A, Parlato R, Huber K, Kinscherf R, Hartleben B, Huber TB, Schütz G, Unsicker K. Cell loss and autophagy in the extra-adrenal chromaffin organ of Zuckerkandl are regulated by glucocorticoid signalling. J Neuroendocrinol 2013; 25:34-47. [PMID: 23078542 PMCID: PMC3564403 DOI: 10.1111/j.1365-2826.2012.02367.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2012] [Accepted: 07/27/2012] [Indexed: 12/20/2022]
Abstract
Neuroendocrine chromaffin cells exist in both intra- and extra-adrenal locations; the organ of Zuckerkandl (OZ) constitutes the largest accumulation of extra-adrenal chromaffin tissue in mammals. The OZ disappears postnatally by modes that are still enigmatic but can be maintained by treatment with glucocorticoids (GC). Whether the response to GC reflects a pharmacological or a physiological role of GC has not been clarified. Using mice with a conditional deletion of the GC-receptor (GR) gene restricted to cells expressing the dopamine β-hydroxylase (DBH) gene [GR(fl/fl) ; DBHCre abbreviated (GR(DBHCre) )], we now present the first evidence for a physiological role of GC signalling in the postnatal maintenance of the OZ: postnatal losses of OZ chromaffin cells in GR(DBHCre) mice are doubled compared to wild-type littermates. We find that postnatal cell loss in the OZ starts at birth and is accompanied by autophagy. Electron microscopy reveals autophagic vacuoles and autophagolysosomes in chromaffin cells. Autophagy in OZ extra-adrenal chromaffin cells is confirmed by showing accumulation of p62 protein, which occurs, when autophagy is blocked by deleting the Atg5 gene (Atg5(DBHCre) mice). Cathepsin-D, a lysosomal marker, is expressed in cells that surround chromaffin cells and are positive for the macrophage marker BM8. Macrophages are relatively more abundant in mice lacking the GR, indicating more robust elimination of degenerating chromaffin cells in GR(DBHCre) mice than in wild-type littermates. In summary, our results indicate that extra-adrenal chromaffin cells in the OZ show signs of autophagy, which accompany their postnatal numerical decline, a process that is controlled by GR signalling.
Collapse
Affiliation(s)
- Andreas Schober
- Department of Molecular Embryology, Institute of Anatomy and Cell Biology II, Albert-Ludwigs-University Freiburg, Freiburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Reiff T, Huber L, Kramer M, Delattre O, Janoueix-Lerosey I, Rohrer H. Midkine and Alk signaling in sympathetic neuron proliferation and neuroblastoma predisposition. Development 2011; 138:4699-708. [PMID: 21989914 DOI: 10.1242/dev.072157] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor in childhood and arises from cells of the developing sympathoadrenergic lineage. Activating mutations in the gene encoding the ALK tyrosine kinase receptor predispose for NB. Here, we focus on the normal function of Alk signaling in the control of sympathetic neuron proliferation, as well as on the effects of mutant ALK. Forced expression of wild-type ALK and NB-related constitutively active ALK mutants in cultures of proliferating immature sympathetic neurons results in a strong proliferation increase, whereas Alk knockdown and pharmacological inhibition of Alk activity decrease proliferation. Alk activation upregulates NMyc and trkB and maintains Alk expression by an autoregulatory mechanism involving Hand2. The Alk-ligand Midkine (Mk) is expressed in immature sympathetic neurons and in vivo inhibition of Alk signaling by virus-mediated shRNA knockdown of Alk and Mk leads to strongly reduced sympathetic neuron proliferation. Taken together, these results demonstrate that the extent and timing of sympathetic neurogenesis is controlled by Mk/Alk signaling. The predisposition for NB caused by activating ALK mutations may thus be explained by aberrations of normal neurogenesis, i.e. elevated and sustained Alk signaling and increased NMyc expression.
Collapse
Affiliation(s)
- Tobias Reiff
- Research Group Developmental Neurobiology, Max Planck Institute for Brain Research, Deutschordenstr. 46, 60528, Frankfurt/M, Germany
| | | | | | | | | | | |
Collapse
|
28
|
Huber L, Ferdin M, Holzmann J, Stubbusch J, Rohrer H. HoxB8 in noradrenergic specification and differentiation of the autonomic nervous system. Dev Biol 2011; 363:219-33. [PMID: 22236961 DOI: 10.1016/j.ydbio.2011.12.026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 12/14/2011] [Accepted: 12/15/2011] [Indexed: 10/25/2022]
Abstract
Different prespecification of mesencephalic and trunk neural crest cells determines their response to environmental differentiation signals and contributes to the generation of different autonomic neuron subtypes, parasympathetic ciliary neurons in the head and trunk noradrenergic sympathetic neurons. The differentiation of ciliary and sympathetic neurons shares many features, including the initial BMP-induced expression of noradrenergic characteristics that is, however, subsequently lost in ciliary but maintained in sympathetic neurons. The molecular basis of specific prespecification and differentiation patterns has remained unclear. We show here that HoxB gene expression in trunk neural crest is maintained in sympathetic neurons. Ectopic expression of a single HoxB gene, HoxB8, in mesencephalic neural crest results in a strongly increased expression of sympathetic neuron characteristics like the transcription factor Hand2, tyrosine hydroxylase (TH) and dopamine-beta-hydroxylase (DBH) in ciliary neurons. Other subtype-specific properties like RGS4 and RCad are not induced. HoxB8 has only minor effects in postmitotic ciliary neurons and is unable to induce TH and DBH in the enteric nervous system. Thus, we conclude that HoxB8 acts by maintaining noradrenergic properties transiently expressed in ciliary neuron progenitors during normal development. HoxC8, HoxB9, HoxB1 and HoxD10 elicit either small and transient or no effects on noradrenergic differentiation, suggesting a selective effect of HoxB8. These results implicate that Hox genes contribute to the differential development of autonomic neuron precursors by maintaining noradrenergic properties in the trunk sympathetic neuron lineage.
Collapse
Affiliation(s)
- Leslie Huber
- Research Group Developmental Neurobiology, Max Planck Institute for Brain Research, Frankfurt/Main, Germany
| | | | | | | | | |
Collapse
|
29
|
Schmidt M, Huber L, Majdazari A, Schütz G, Williams T, Rohrer H. The transcription factors AP-2β and AP-2α are required for survival of sympathetic progenitors and differentiated sympathetic neurons. Dev Biol 2011; 355:89-100. [DOI: 10.1016/j.ydbio.2011.04.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2011] [Revised: 04/13/2011] [Accepted: 04/14/2011] [Indexed: 11/26/2022]
|
30
|
Distinct and conserved prominin-1/CD133-positive retinal cell populations identified across species. PLoS One 2011; 6:e17590. [PMID: 21407811 PMCID: PMC3047580 DOI: 10.1371/journal.pone.0017590] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 01/28/2011] [Indexed: 02/08/2023] Open
Abstract
Besides being a marker of various somatic stem cells in mammals, prominin-1 (CD133) plays a role in maintaining the photoreceptor integrity since mutations in the PROM1 gene are linked with retinal degeneration. In spite of that, little information is available regarding its distribution in eyes of non-mammalian vertebrates endowed with high regenerative abilities. To address this subject, prominin-1 cognates were isolated from axolotl, zebrafish and chicken, and their retinal compartmentalization was investigated and compared to that of their mammalian orthologue. Interestingly, prominin-1 transcripts--except for the axolotl--were not strictly restricted to the outer nuclear layer (i.e., photoreceptor cells), but they also marked distinct subdivisions of the inner nuclear layer (INL). In zebrafish, where the prominin-1 gene is duplicated (i.e., prominin-1a and prominin-1b), a differential expression was noted for both paralogues within the INL being localized either to its vitreal or scleral subdivision, respectively. Interestingly, expression of prominin-1a within the former domain coincided with Pax-6-positive cells that are known to act as progenitors upon injury-induced retino-neurogenesis. A similar, but minute population of prominin-1-positive cells located at the vitreal side of the INL was also detected in developing and adult mice. In chicken, however, prominin-1-positive cells appeared to be aligned along the scleral side of the INL reminiscent of zebrafish prominin-1b. Taken together our data indicate that in addition to conserved expression of prominin-1 in photoreceptors, significant prominin-1-expressing non-photoreceptor retinal cell populations are present in the vertebrate eye that might represent potential sources of stem/progenitor cells for regenerative therapies.
Collapse
|
31
|
Peterziel H, Sackmann T, Strelau J, Kuhn PH, Lichtenthaler SF, Marom K, Klar A, Unsicker K. F-spondin regulates neuronal survival through activation of disabled-1 in the chicken ciliary ganglion. Mol Cell Neurosci 2010; 46:483-97. [PMID: 21145970 DOI: 10.1016/j.mcn.2010.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Revised: 11/19/2010] [Accepted: 12/02/2010] [Indexed: 01/06/2023] Open
Abstract
The extracellular membrane-associated protein F-spondin has been implicated in cell-matrix and cell-cell adhesion and plays an important role in axonal pathfinding. We report here that F-spondin is expressed in non-neuronal cells in the embryonic chicken ciliary ganglion (CG) and robustly promotes survival of cultured CG neurons. Using deletion constructs of F-spondin we found that the amino-terminal Reelin/Spondin domain cooperates with thrombospondin type 1 repeat (TSR) 6, a functional TGFβ-activation domain. In ovo treatment with blocking antibodies raised against the Reelin/Spondin domain or the TSR-domains caused increased apoptosis of CG neurons during the phase of programmed cell death and loss of about 30% of the neurons compared to controls. The Reelin/Spondin domain receptor - APP and its downstream signalling molecule disabled-1 are expressed in CG neurons. F-spondin induced rapid phosphorylation of disabled-1. Moreover, both blocking the central APP domain and interference with disabled-1 signalling disrupted the survival promoting effect of F-spondin. Taken together, our data suggest that F-spondin can promote neuron survival by a mechanism involving the Reelin/Spondin and the TSR domains.
Collapse
Affiliation(s)
- H Peterziel
- Neuroanatomy & Interdisciplinary Center for Neurosiences (IZN), University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Schindowski K, von Bohlen und Halbach O, Strelau J, Ridder DA, Herrmann O, Schober A, Schwaninger M, Unsicker K. Regulation of GDF-15, a distant TGF-β superfamily member, in a mouse model of cerebral ischemia. Cell Tissue Res 2010; 343:399-409. [PMID: 21128084 PMCID: PMC3032194 DOI: 10.1007/s00441-010-1090-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2010] [Accepted: 11/04/2010] [Indexed: 11/20/2022]
Abstract
GDF-15 is a novel distant member of the TGF-β superfamily and is widely distributed in the brain and peripheral nervous system. We have previously reported that GDF-15 is a potent neurotrophic factor for lesioned dopaminergic neurons in the substantia nigra, and that GDF-15-deficient mice show progressive postnatal losses of motor and sensory neurons. We have now investigated the regulation of GDF-15 mRNA and immunoreactivity in the murine hippocampal formation and selected cortical areas following an ischemic lesion by occlusion of the middle cerebral artery (MCAO). MCAO prominently upregulates GDF-15 mRNA in the hippocampus and parietal cortex at 3 h and 24 h after lesion. GDF-15 immunoreactivity, which is hardly detectable in the unlesioned brain, is drastically upregulated in neurons identified by double-staining with NeuN. NeuN staining reveals that most, if not all, neurons in the granular layer of the dentate gyrus and pyramidal layers of the cornu ammonis become GDF-15-immunoreactive. Moderate induction of GDF-15 immunoreactivity has been observed in a small number of microglial cells identified by labeling with tomato lectin, whereas astroglial cells remain GDF-15-negative after MCAO. Comparative analysis of the size of the infarcted area after MCAO in GDF-15 wild-type and knockout mice has failed to reveal significant differences. Together, our data substantiate the notion that GDF-15 is prominently upregulated in the lesioned brain and might be involved in orchestrating post-lesional responses other than the trophic support of neurons.
Collapse
Affiliation(s)
- Katharina Schindowski
- Institute for Pharmaceutical Biotechnology, University of Applied Science Biberach, Biberach/Riss, Germany
| | | | | | | | | | | | | | | |
Collapse
|
33
|
The Gata3 transcription factor is required for the survival of embryonic and adult sympathetic neurons. J Neurosci 2010; 30:10833-43. [PMID: 20702712 DOI: 10.1523/jneurosci.0175-10.2010] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The transcription factor Gata3 is essential for the development of sympathetic neurons and adrenal chromaffin cells. As Gata3 expression is maintained up to the adult stage, we addressed its function in differentiated sympathoadrenal cells at embryonic and adult stages by conditional Gata3 elimination. Inactivation of Gata3 in embryonic DBH-expressing neurons elicits a strong reduction in neuron numbers due to apoptotic cell death and reduced proliferation. No selective effect on noradrenergic gene expression (TH and DBH) was observed. Interestingly, Gata3 elimination in DBH-expressing neurons of adult animals also results in a virtually complete loss of sympathetic neurons. In the Gata3-deficient population, the expression of anti-apoptotic genes (Bcl-2, Bcl-xL, and NFkappaB) is diminished, whereas the expression of pro-apoptotic genes (Bik, Bok, and Bmf) was increased. The expression of noradrenergic genes (TH and DBH) is not affected. These results demonstrate that Gata3 is continuously required for maintaining survival but not differentiation in the sympathetic neuron lineage up to mature neurons of adult animals.
Collapse
|
34
|
Apostolova G, Dechant G. Development of neurotransmitter phenotypes in sympathetic neurons. Auton Neurosci 2009; 151:30-8. [DOI: 10.1016/j.autneu.2009.08.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
35
|
Generating diversity: Mechanisms regulating the differentiation of autonomic neuron phenotypes. Auton Neurosci 2009; 151:17-29. [PMID: 19819195 DOI: 10.1016/j.autneu.2009.08.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Sympathetic and parasympathetic postganglionic neurons innervate a wide range of target tissues. The subpopulation of neurons innervating each target tissue can express unique combinations of neurotransmitters, neuropeptides, ion channels and receptors, which together comprise the chemical phenotype of the neurons. The target-specific chemical phenotype shown by autonomic postganglionic neurons arises during development. In this review, we examine the different mechanisms that generate such a diversity of neuronal phenotypes from the pool of apparently homogenous neural crest progenitor cells that form the sympathetic ganglia. There is evidence that the final chemical phenotype of autonomic postganglionic neurons is generated by both signals at the level of the cell body that trigger cell-autonomous programs, as well as signals from the target tissues they innervate.
Collapse
|
36
|
Abellán A, Medina L. Subdivisions and derivatives of the chicken subpallium based on expression of LIM and other regulatory genes and markers of neuron subpopulations during development. J Comp Neurol 2009; 515:465-501. [DOI: 10.1002/cne.22083] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
37
|
Schmidt M, Lin S, Pape M, Ernsberger U, Stanke M, Kobayashi K, Howard MJ, Rohrer H. The bHLH transcription factor Hand2 is essential for the maintenance of noradrenergic properties in differentiated sympathetic neurons. Dev Biol 2009; 329:191-200. [PMID: 19254708 DOI: 10.1016/j.ydbio.2009.02.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Revised: 02/17/2009] [Accepted: 02/18/2009] [Indexed: 01/01/2023]
Abstract
The basic helix-loop-helix transcription factor Hand2 is essential for the proliferation and noradrenergic differentiation of sympathetic neuron precursors during development. Here we address the function of Hand2 in postmitotic, differentiated sympathetic neurons. Knockdown of endogenous Hand2 in cultured E12 chick sympathetic neurons by siRNA results in a significant (about 60%) decrease in the expression of the noradrenergic marker genes dopamine-beta-hydroxylase (DBH) and tyrosine hydroxylase (TH). In contrast, expression of the pan-neuronal genes TuJ1, HuC and SCG10 was not affected. To analyze the in vivo role of Hand2 in differentiated sympathetic neurons we used mice harboring a conditional Hand2-null allele and excised the gene by expression of Cre recombinase under control of the DBH promotor. Mouse embryos homozygous for Hand2 gene deletion showed decreased sympathetic neuron number and TH expression was strongly reduced in the residual neuron population. The in vitro Hand2 knockdown also enhances the CNTF-induced expression of the cholinergic marker genes vesicular acetylcholine transporter (VAChT) and choline acetyltransferase (ChAT). Taken together, these findings demonstrate that the Hand2 transcription factor plays a key role in maintaining noradrenergic properties in differentiated neurons.
Collapse
Affiliation(s)
- Mirko Schmidt
- RG Developmental Neurobiology, Department of Neurochemistry, MPI for Brain Research, Deutschordenstr. 46, 60528 Frankfurt/M, Germany
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Huber K, Franke A, Brühl B, Krispin S, Ernsberger U, Schober A, von Bohlen und Halbach O, Rohrer H, Kalcheim C, Unsicker K. Persistent expression of BMP-4 in embryonic chick adrenal cortical cells and its role in chromaffin cell development. Neural Dev 2008; 3:28. [PMID: 18945349 PMCID: PMC2582231 DOI: 10.1186/1749-8104-3-28] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2008] [Accepted: 10/22/2008] [Indexed: 11/29/2022] Open
Abstract
Background Adrenal chromaffin cells and sympathetic neurons both originate from the neural crest, yet signals that trigger chromaffin development remain elusive. Bone morphogenetic proteins (BMPs) emanating from the dorsal aorta are important signals for the induction of a sympathoadrenal catecholaminergic cell fate. Results We report here that BMP-4 is also expressed by adrenal cortical cells throughout chick embryonic development, suggesting a putative role in chromaffin cell development. Moreover, bone morphogenetic protein receptor IA is expressed by both cortical and chromaffin cells. Inhibiting BMP-4 with noggin prevents the increase in the number of tyrosine hydroxylase positive cells in adrenal explants without affecting cell proliferation. Hence, adrenal BMP-4 is likely to induce tyrosine hydroxylase in sympathoadrenal progenitors. To investigate whether persistent BMP-4 exposure is able to induce chromaffin traits in sympathetic ganglia, we locally grafted BMP-4 overexpressing cells next to sympathetic ganglia. Embryonic day 8 chick sympathetic ganglia, in addition to principal neurons, contain about 25% chromaffin-like cells. Ectopic BMP-4 did not increase this proportion, yet numbers and sizes of 'chromaffin' granules were significantly increased. Conclusion BMP-4 may serve to promote specific chromaffin traits, but is not sufficient to convert sympathetic neurons into a chromaffin phenotype.
Collapse
Affiliation(s)
- Katrin Huber
- Neuroanatomy, Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Kapeller J, Houghton LA, Mönnikes H, Walstab J, Möller D, Bönisch H, Burwinkel B, Autschbach F, Funke B, Lasitschka F, Gassler N, Fischer C, Whorwell PJ, Atkinson W, Fell C, Büchner KJ, Schmidtmann M, van der Voort I, Wisser AS, Berg T, Rappold G, Niesler B. First evidence for an association of a functional variant in the microRNA-510 target site of the serotonin receptor-type 3E gene with diarrhea predominant irritable bowel syndrome. Hum Mol Genet 2008; 17:2967-2977. [PMID: 18614545 DOI: 10.1093/hmg/ddn195] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Diarrhea predominant irritable bowel syndrome (IBS-D) is a complex disorder related to dysfunctions in the serotonergic system. As cis-regulatory variants can play a role in the etiology of complex conditions, we investigated the untranslated regions (UTRs) of the serotonin receptor type 3 subunit genes HTR3A and HTR3E. Mutation analysis was carried out in a pilot sample of 200 IBS patients and 100 healthy controls from the UK. The novel HTR3E 3'-UTR variant c.*76G>A (rs62625044) was associated with female IBS-D (P = 0.033, OR = 8.53). This association was confirmed in a replication study, including 119 IBS-D patients and 195 controls from Germany (P = 0.0046, OR = 4.92). Pooled analysis resulted in a highly significant association of c.*76G>A with female IBS-D (P = 0.0002, OR = 5.39). In a reporter assay, c.*76G>A affected binding of miR-510 to the HTR3E 3'-UTR and caused elevated luciferase expression. HTR3E and miR-510 co-localize in enterocytes of the gut epithelium as shown by in situ hybridization and RT-PCR. This is the first example indicating micro RNA-related expression regulation of a serotonin receptor gene with a cis-regulatory variant affecting this regulation and appearing to be associated with female IBS-D.
Collapse
Affiliation(s)
- Johannes Kapeller
- Department of Human Molecular Genetics, University of Heidelberg, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Pape M, Doxakis E, Reiff T, Duong CV, Davies A, Geissen M, Rohrer H. A function for the calponin family member NP25 in neurite outgrowth. Dev Biol 2008; 321:434-43. [PMID: 18652818 DOI: 10.1016/j.ydbio.2008.07.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2007] [Revised: 06/30/2008] [Accepted: 07/01/2008] [Indexed: 11/19/2022]
Abstract
The neuronal protein 25 (NP25), a member of the calponin (CaP) protein family, has previously been identified as neuron-specific protein in the adult rat brain. Here, we show an early onset of NP25 expression in the chick embryo neural tube. NP25 represents, together with NeuroM, one of the earliest markers for postmitotic neurons. To elucidate its function in the developing nervous system, NP25 was overexpressed in E5 and E9 sensory neurons, E7 sympathetic neurons and PC12 cells that show different endogenous NP25 expression levels. Whereas E5 and E9 sensory neurons and PC12 cells, which express low endogenous levels of NP25, responded by enhanced neurite outgrowth, a reduction of neurite length was observed in sympathetic neurons, which already express high endogenous levels of NP25. Knockdown of NP25 in sensory neurons using NP25 siRNA resulted in shorter neurites, whereas reduction of NP25 expression in sympathetic neurons led to increased neurite length. These results suggest a dynamic function for NP25 in the regulation of neurite growth, with an optimal level of NP25 required for maximal growth.
Collapse
Affiliation(s)
- Manuela Pape
- Max-Planck-Institute for Brain Research, Deutschordenstr. 46, 60528 Frankfurt/M, Germany
| | | | | | | | | | | | | |
Collapse
|
41
|
Ernsberger U. The role of GDNF family ligand signalling in the differentiation of sympathetic and dorsal root ganglion neurons. Cell Tissue Res 2008; 333:353-71. [PMID: 18629541 PMCID: PMC2516536 DOI: 10.1007/s00441-008-0634-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2008] [Accepted: 05/05/2008] [Indexed: 03/24/2023]
Abstract
The diversity of neurons in sympathetic ganglia and dorsal root ganglia (DRG) provides intriguing systems for the analysis of neuronal differentiation. Cell surface receptors for the GDNF family ligands (GFLs) glial cell-line-derived neurotrophic factor (GDNF), neurturin and artemin, are expressed in subpopulations of these neurons prompting the question regarding their involvement in neuronal subtype specification. Mutational analysis in mice has demonstrated the requirement for GFL signalling during embryonic development of cholinergic sympathetic neurons as shown by the loss of expression from the cholinergic gene locus in ganglia from mice deficient for ret, the signal transducing subunit of the GFL receptor complex. Analysis in mutant animals and transgenic mice overexpressing GFLs demonstrates an effect on sensitivity to thermal and mechanical stimuli in DRG neurons correlating at least partially with the altered expression of transient receptor potential ion channels and acid-sensitive cation channels. Persistence of targeted cells in mutant ganglia suggests that the alterations are caused by differentiation effects and not by cell loss. Because of the massive effect of GFLs on neurite outgrowth, it remains to be determined whether GFL signalling acts directly on neuronal specification or indirectly via altered target innervation and access to other growth factors. The data show that GFL signalling is required for the specification of subpopulations of sensory and autonomic neurons. In order to comprehend this process fully, the role of individual GFLs, the transduction of the GFL signals, and the interplay of GFL signalling with other regulatory pathways need to be deciphered.
Collapse
Affiliation(s)
- Uwe Ernsberger
- Interdisciplinary Center for Neurosciences (IZN), University of Heidelberg, Heidelberg, Germany.
| |
Collapse
|
42
|
Tsarovina K, Schellenberger J, Schneider C, Rohrer H. Progenitor cell maintenance and neurogenesis in sympathetic ganglia involves Notch signaling. Mol Cell Neurosci 2008; 37:20-31. [DOI: 10.1016/j.mcn.2007.08.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2007] [Revised: 08/10/2007] [Accepted: 08/16/2007] [Indexed: 02/02/2023] Open
|
43
|
Apostolova G, Dorn R, Ka S, Hallböök F, Lundeberg J, Liser K, Hakim V, Brodski C, Michaelidis TM, Dechant G. Neurotransmitter phenotype-specific expression changes in developing sympathetic neurons. Mol Cell Neurosci 2007; 35:397-408. [PMID: 17513123 DOI: 10.1016/j.mcn.2007.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Revised: 03/27/2007] [Accepted: 03/28/2007] [Indexed: 01/08/2023] Open
Abstract
During late developmental phases individual sympathetic neurons undergo a switch from noradrenergic to cholinergic neurotransmission. This phenomenon of plasticity depends on target-derived signals in vivo and is triggered by neurotrophic factors in neuronal cultures. To analyze genome-wide expression differences between the two transmitter phenotypes we employed DNA microarrays. RNA expression profiles were obtained from chick paravertebral sympathetic ganglia, treated with neurotrophin 3, glial cell line-derived neurotrophic factor or ciliary neurotrophic factor, all of which stimulate cholinergic differentiation. Results were compared with the effect of nerve growth factor, which functions as a pro-noradrenergic stimulus. The gene set common to all three comparisons defined the noradrenergic and cholinergic synexpression groups. Several functional categories, such as signal transduction, G-protein-coupled signaling, cation transport, neurogenesis and synaptic transmission, were enriched in these groups. Experiments based on the prediction that some of the identified genes play a role in the neurotransmitter switch identified bone morphogenetic protein signaling as an inhibitor of cholinergic differentiation.
Collapse
Affiliation(s)
- Galina Apostolova
- Institute for Neuroscience, Innsbruck Medical University, MZA, Anichstrasse 35, 6020 Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mosher JT, Yeager KJ, Kruger GM, Joseph NM, Hutchin ME, Dlugosz AA, Morrison SJ. Intrinsic differences among spatially distinct neural crest stem cells in terms of migratory properties, fate determination, and ability to colonize the enteric nervous system. Dev Biol 2006; 303:1-15. [PMID: 17113577 PMCID: PMC1910607 DOI: 10.1016/j.ydbio.2006.10.026] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2006] [Revised: 10/12/2006] [Accepted: 10/16/2006] [Indexed: 01/21/2023]
Abstract
We have systematically examined the developmental potential of neural crest stem cells from the enteric nervous system (gut NCSCs) in vivo to evaluate their potential use in cellular therapy for Hirschsprung disease and to assess differences in the properties of postmigratory NCSCs from different regions of the developing peripheral nervous system (PNS). When transplanted into developing chicks, flow-cytometrically purified gut NCSCs and sciatic nerve NCSCs exhibited intrinsic differences in migratory potential and neurogenic capacity throughout the developing PNS. Most strikingly, gut NCSCs migrated into the developing gut and formed enteric neurons, while sciatic nerve NCSCs failed to migrate into the gut or to make enteric neurons, even when transplanted into the gut wall. Enteric potential is therefore not a general property of NCSCs. Gut NCSCs also formed cholinergic neurons in parasympathetic ganglia, but rarely formed noradrenergic sympathetic neurons or sensory neurons. Supporting the potential for autologous transplants in Hirschsprung disease, we observed that Endothelin receptor B (Ednrb)-deficient gut NCSCs engrafted and formed neurons as efficiently in the Ednrb-deficient hindgut as did wild-type NCSCs. These results demonstrate intrinsic differences in the migratory properties and developmental potentials of regionally distinct NCSCs, indicating that it is critical to match the physiological properties of neural stem cells to the goals of proposed cell therapies.
Collapse
Affiliation(s)
- Jack T. Mosher
- Howard Hughes Medical Institute, Life Sciences Institute, Department of Internal Medicine, Center for Stem Cell Biology, University of Michigan, Ann Arbor, Michigan, 48109-2216
| | - Kelly J. Yeager
- Howard Hughes Medical Institute, Life Sciences Institute, Department of Internal Medicine, Center for Stem Cell Biology, University of Michigan, Ann Arbor, Michigan, 48109-2216
| | - Genevieve M. Kruger
- Howard Hughes Medical Institute, Life Sciences Institute, Department of Internal Medicine, Center for Stem Cell Biology, University of Michigan, Ann Arbor, Michigan, 48109-2216
| | - Nancy M. Joseph
- Howard Hughes Medical Institute, Life Sciences Institute, Department of Internal Medicine, Center for Stem Cell Biology, University of Michigan, Ann Arbor, Michigan, 48109-2216
| | - Mark E. Hutchin
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, 48109-2216
| | - Andrzej A. Dlugosz
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, 48109-2216
| | - Sean J. Morrison
- Howard Hughes Medical Institute, Life Sciences Institute, Department of Internal Medicine, Center for Stem Cell Biology, University of Michigan, Ann Arbor, Michigan, 48109-2216
- *Author for correspondence: 5435 Life Sciences Institute, 210 Washtenaw Ave., Ann Arbor, Michigan, 48109-2216; phone 734-647-6261; fax 734-615-8133; email
| |
Collapse
|
45
|
Politis PK, Rohrer H, Matsas R. Expression pattern of BM88 in the developing nervous system of the chick and mouse embryo. Gene Expr Patterns 2006; 7:165-77. [PMID: 16949349 DOI: 10.1016/j.modgep.2006.06.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2006] [Revised: 06/28/2006] [Accepted: 06/29/2006] [Indexed: 11/21/2022]
Abstract
We isolated a chick homologue of BM88 (cBM88), a cell-intrinsic nervous system-specific protein and examined the expression of BM88 mRNA and protein in the developing brain, spinal cord and peripheral nervous system of the chick embryo by in situ hybridization and immunohistochemistry. cBM88 is widely expressed in the developing central nervous system, both in the ventricular and mantle zones where precursor and differentiated cells lie, respectively. In the spinal cord, particularly strong cBM88 expression is detected ventrally in the motor neuron area. cBM88 is also expressed in the dorsal root ganglia and sympathetic ganglia. In the early neural tube, cBM88 is first detected at HH stage 15 and its expression increases with embryonic age. At early stages, cBM88 expression is weaker in the ventricular zone (VZ) and higher in the mantle zone. At later stages, when gliogenesis persists instead of neurogenesis, BM88 expression is abolished in the VZ and cBM88 is restricted in the neuron-containing mantle zone of the neural tube. Association of cBM88 expression with cells of the neuronal lineage in the chick spinal cord was demonstrated using a combination of markers characteristic of neuronal or glial precursors, as well as markers of differentiated neuronal, oligodendroglial and astroglial cells. In addition to the spinal cord, cBM88 is expressed in the HH stage 45 (embryonic day 19) brain, including the telencephalon, diencephalon, mesencephalon, optic tectum and cerebellum. BM88 is also widely expressed in the mouse embryonic CNS and PNS, in both nestin-positive neuroepithelial cells and post-mitotic betaIII-tubulin positive neurons.
Collapse
Affiliation(s)
- Panagiotis K Politis
- Laboratory of Cellular and Molecular Neurobiology, Hellenic Pasteur Institute, 127 Vas. Sofias Avenue, Athens 11521, Greece
| | | | | |
Collapse
|
46
|
Lohr J, Gut P, Karch N, Unsicker K, Huber K. Development of adrenal chromaffin cells in Sf1 heterozygous mice. Cell Tissue Res 2006; 325:437-44. [PMID: 16685530 DOI: 10.1007/s00441-006-0213-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Accepted: 04/06/2006] [Indexed: 10/24/2022]
Abstract
Adrenal medullary chromaffin cells are derivatives of the neural crest and are widely believed to share a common sympathoadrenal (SA) progenitor with sympathetic neurons. For decades, the adrenal cortical environment was assumed to be essential for channelling SA progenitors towards an endocrine chromaffin cell fate. Our recent analysis of steroidogenic factor 1(Sf1) -/- mice, which lack an adrenal cortex, has challenged this view: in Sf1 -/- mice chromaffin cells migrate to the correct "adrenal" location and undergo largely normal differentiation. In contrast to Sf1 homozygous mutants, heterozygous animals have an adrenal cortex, which, however, is smaller than in wildtype littermates. We show here that the Sf1 +/- adrenal cortical anlagen attract normal numbers of chromaffin progenitor cells into their vicinity by embryonic day 13.5 (E13.5). Two days later, however, only a few scattered cells with highly immature features have immigrated into the adrenal cortex, whereas the remainder form a coherent cell assembly ectopically located at the medial surface of the gland. These cells appear more mature than the scattered intracortical chromaffin progenitors and express the adrenaline synthesizing enzyme PNMT with a delay of 1 day in comparison with wildtype littermates. Nevertheless, chromaffin progenitor cells undergo a numerical reduction of approximately 30% by E17.5. Together, our data suggest that normal adrenocortical development is critical for the correct immigration of chromaffin progenitors into the cortical anlagen, for the timing of PNMT expression and for the regulation of chromaffin cell numbers.
Collapse
Affiliation(s)
- Jennifer Lohr
- Neuroanatomy and Interdisciplinary Centre for Neurosciences (IZN), University of Heidelberg, Im Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | | | | | | | | |
Collapse
|
47
|
Oberle S, Schober A, Meyer V, Holtmann B, Henderson C, Sendtner M, Unsicker K. Loss of leukemia inhibitory factor receptor beta or cardiotrophin-1 causes similar deficits in preganglionic sympathetic neurons and adrenal medulla. J Neurosci 2006; 26:1823-32. [PMID: 16467531 PMCID: PMC6793615 DOI: 10.1523/jneurosci.4127-05.2006] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Leukemia inhibitory factor (LIF) receptor beta (LIFRbeta) is a receptor for a variety of neurotrophic cytokines, including LIF, ciliary neurotrophic factor (CNTF), and cardiotrophin-1 (CT-1). These cytokines play an essential role for the survival and maintenance of developing and postnatal somatic motoneurons. CNTF may also serve the maintenance of autonomic, preganglionic sympathetic neurons (PSNs) in the spinal cord, as suggested by its capacity to prevent their death after destruction of one of their major targets, the adrenal medulla. Although somatic motoneurons and PSNs share a common embryonic origin, they are distinct in several respects, including responses to lesions. We have studied PSNs in mice with targeted deletions of the LIFRbeta or CT-1 genes, respectively. We show that LIF, CNTF, and CT-1 are synthesized in embryonic adrenal gland and spinal cord and that PSNs express LIFRbeta. In embryonic day 18.5 LIFRbeta (-/-) and CT-1 (-/-) mice, PSNs were reduced by approximately 20%. PSNs projecting to the adrenal medulla were more severely affected (-55%). Although LIFRbeta (-/-) mice revealed normal numbers of adrenal chromaffin cells and axons terminating on chromaffin cells, levels of adrenaline and numbers of adrenaline-synthesizing cells were significantly reduced. We conclude that activation of LIFRbeta is required for normal development of PSNs and one of their prominent targets, the adrenal medulla. Thus, both somatic motoneurons and PSNs in the spinal cord depend on LIFRbeta signaling for their development and maintenance, although PSNs seem to be overall less affected than somatic motoneurons by LIFRbeta deprivation.
Collapse
|
48
|
Huber K, Ernsberger U. Cholinergic differentiation occurs early in mouse sympathetic neurons and requires Phox2b. Gene Expr 2006; 13:133-9. [PMID: 17017126 PMCID: PMC6032475 DOI: 10.3727/000000006783991854] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The generation of neurotransmitter identity in the autonomic nervous system is a classical model system to study the development of neuronal diversity. Analysis of the expression of genes coding for enzymes of noradrenaline biosynthesis in the sympathoadrenal system allowed the characterization of factors involved in the differentiation of the noradrenergic transmitter phenotype. The development of cholinergic properties in the autonomic system is less well understood. Here we show that expression of mRNAs for choline acetyltransferase (ChAT) and the vesicular acetylcholine transporter (VAChT), both encoded by the cholinergic gene locus, is induced in mouse sympathetic ganglia at embryonic day 11 (E11). Positive cells amount to more than 50% of Phox2b-positive sympathetic cells at cervical levels. The proportion declines caudally, decreasing to approximately 20% of Phox2b-positive cells at lower thoracic levels. In the adrenal anlage, ChAT and VAChT mRNA are largely undetectable at E11 and E13. In mice homozygous for a mutational inactivation of the transcription factor Phox2b, ChAT and VAChT mRNA expression is absent from sympathetic ganglia. The data show that expression from the cholinergic gene locus is regulated differently in sympathetic neurons and adrenal chromaffin cells. Phox2b is required for development of cholinergic neurons but does not suffice to support cholinergic properties in chromaffin cells.
Collapse
Affiliation(s)
- K Huber
- Institut für Anatomie und Zellbiologie III, Interdisziplindäres Zentrum für Neurowissenschaften, Ruprecht-Karls-Universität Heidelberg, Imn Neuenheimer Feld 307, 69120 Heidelberg, Germany
| | | |
Collapse
|
49
|
Stanke M, Duong CV, Pape M, Geissen M, Burbach G, Deller T, Gascan H, Otto C, Parlato R, Schütz G, Rohrer H. Target-dependent specification of the neurotransmitter phenotype: cholinergic differentiation of sympathetic neurons is mediated in vivo by gp 130 signaling. Development 2005; 133:141-50. [PMID: 16319110 DOI: 10.1242/dev.02189] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Sympathetic neurons are generated through a succession of differentiation steps that initially lead to noradrenergic neurons innervating different peripheral target tissues. Specific targets, like sweat glands in rodent footpads, induce a change from noradrenergic to cholinergic transmitter phenotype. Here, we show that cytokines acting through the gp 130 receptor are present in sweat glands. Selective elimination of the gp 130 receptor in sympathetic neurons prevents the acquisition of cholinergic and peptidergic features (VAChT, ChT1, VIP) without affecting other properties of sweat gland innervation. The vast majority of cholinergic neurons in the stellate ganglion, generated postnatally, are absent in gp 130-deficient mice. These results demonstrate an essential role of gp 130-signaling in the target-dependent specification of the cholinergic neurotransmitter phenotype.
Collapse
Affiliation(s)
- Matthias Stanke
- Research Group Developmental Neurobiology, Max-Planck-Institute for Brain Research, Deutschordenstrasse 46, 60528 Frankfurt/M, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Badde A, Bumsted-O'Brien KM, Schulte D. Chick receptor protein tyrosine phosphatase lambda/psi (cRPTPlambda/cRPTPpsi) is dynamically expressed at the midbrain-hindbrain boundary and in the embryonic neural retina. Gene Expr Patterns 2005; 5:786-91. [PMID: 15922674 DOI: 10.1016/j.modgep.2005.04.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2004] [Revised: 03/31/2005] [Accepted: 04/01/2005] [Indexed: 11/17/2022]
Abstract
The isthmic organizer, located near the boundary between the developing midbrain and hindbrain, controls the patterning of adjacent brain regions. Here we describe the spatial and temporal expression of chick receptor-like protein tyrosine phosphatase lambda (cRPTPlambda, also known as cRPTPpsi) during the development of this structure. After an initial widespread expression throughout the caudal forebrain and midbrain region, expression of cRPTPlambda is confined to the ventral midline of the neural tube, the future neural retina and lens, and a sharp ring at the isthmic constriction, overlying the molecular mid-hindbrain boundary (MHB). MHB expression of cRPTPlambda borders at the caudal limit of the expression domain of the transcription factor Otx2, appears to partially overlap with that of the secreted protein Wnt1 and is similar to that of the Ig-CAM CEPU-1. In the neural retina, expression is restricted to the ventricular zone, where the cell bodies of retinal progenitor cells reside.
Collapse
Affiliation(s)
- Anja Badde
- Department of Neuroanatomy, Max-Planck-Institute for Brain Research, Deutschordenstr. 46, 60528 Frankfurt/M., Germany
| | | | | |
Collapse
|