Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
2.
Cell Tissue Res ; 386(3): 455-475, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34757495

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.


Subject(s)
Neural Pathways/physiology , Neurons/physiology , Sympathetic Nervous System/physiology , Animals , Cell Differentiation , Humans
3.
Cell Tissue Res ; 382(2): 201-231, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32930881

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.


Subject(s)
Autonomic Nervous System/physiology , Ganglia, Sympathetic/physiology , Animals , Humans , Neurons , Phenotype , Rodentia
4.
Cell Tissue Res ; 372(2): 325-337, 2018 05.
Article in English | MEDLINE | ID: mdl-29374774

ABSTRACT

The ALK gene encodes a tyrosine kinase receptor characterized by an expression pattern mainly restricted to the developing central and peripheral nervous systems. In 2008, the discovery of ALK activating mutations in neuroblastoma, a tumor of the sympathetic nervous system, represented a breakthrough in the understanding of the pathogenesis of this pediatric cancer and established mutated ALK as a tractable therapeutic target for precision medicine. Subsequent studies addressed the identity of ALK ligands, as well as its physiological function in the sympathoadrenal lineage, its role in neuroblastoma development and the signaling pathways triggered by mutated ALK. This review focuses on these different aspects of the ALK biology and summarizes the various therapeutic strategies relying on ALK inhibition in neuroblastoma, either as monotherapies or combinatory treatments.


Subject(s)
Anaplastic Lymphoma Kinase/metabolism , Neuroblastoma/enzymology , Neuroblastoma/pathology , Neurons/pathology , Sympathetic Nervous System/pathology , Anaplastic Lymphoma Kinase/chemistry , Anaplastic Lymphoma Kinase/genetics , Animals , Humans , Mutation , Neurogenesis , Neurons/metabolism
5.
J Neurosci ; 36(40): 10425-10439, 2016 10 05.
Article in English | MEDLINE | ID: mdl-27707976

ABSTRACT

Neuroblastoma (NB) is a childhood tumor that arises from the sympathoadrenal lineage. MYCN amplification is the most reliable marker for poor prognosis and MYCN overexpression in embryonic mouse sympathetic ganglia results in NB-like tumors. MYCN cooperates with mutational activation of anaplastic lymphoma kinase (ALK), which promotes progression to NB, but the role of MYCN and ALK in tumorigenesis is still poorly understood. Here, we use chick sympathetic neuroblasts to examine the normal function of MYCN and MYC in the control of neuroblast proliferation, as well as effects of overexpression of MYCN, MYC, and activated ALK, alone and in combination. We demonstrate that MYC is more strongly expressed than MYCN during neurogenesis and is important for in vitro neuroblast proliferation. MYC and MYCN overexpression elicits increased proliferation but does not sustain neuroblast survival. Unexpectedly, long-term expression of activated ALKF1174L leads to cell-cycle arrest and promotes differentiation and survival of postmitotic neurons. ALKF1174L induces NEFM, RET, and VACHT and results in decreased expression of proapototic (BMF, BIM), adrenergic (TH), and cell-cycle genes (e.g., CDC25A, CDK1). In contrast, neuroblast proliferation is maintained when MYCN and ALKF1174L are coexpressed. Proliferating MYCN/ALKF1174L neuroblasts display a differentiated phenotype but differ from ALK-expressing neurons by the upregulation of SKP2, CCNA2, E2F8, and DKC1 Inhibition of the ubiquitin ligase SKP2 (S-phase kinase-associated protein 2), which targets the CDK inhibitor p27 for degradation, reduces neuroblast proliferation, implicating SKP2 in the maintained proliferation of MYCN/ALKF1174L neuroblasts. Together, our results characterize MYCN/ALK cooperation leading to neuroblast proliferation and survival that may represent initial steps toward NB development. SIGNIFICANCE STATEMENT: MYCN overexpression combined with activated anaplastic lymphoma kinase (ALK) is sufficient to induce neuroblastoma (NB) in mouse sympathoadrenal cells. To address cellular and molecular effects elicited by MYCN/ALK cooperation, we used cultures of chick sympathetic neuroblasts. We demonstrate that MYCN increases proliferation but not survival, whereas long-term expression of ALKF1174L elicits cell-cycle exit, differentiation, and survival of postmitotic neurons. Combined MYCN/ALKF1174L expression allows long-term proliferation and survival of neuroblasts with differentiated characteristics. In the presence of ALKF1174L signaling, MYCN induces the expression of the ubiquitin ligase SKP2 (S-phase kinase-associated protein 2), which targets p27 for degradation and is also upregulated in high-risk NB. SKP2 inhibition supports a function for SKP2 in the maintained neuroblast proliferation downstream of MYCN/ALK, which may represent an early step toward tumorigenesis.


Subject(s)
N-Myc Proto-Oncogene Protein/genetics , Neural Stem Cells , Neuroblastoma/pathology , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics , Anaplastic Lymphoma Kinase , Animals , Apoptosis/genetics , Cell Cycle Checkpoints , Cell Line, Tumor , Cell Proliferation , Cell Survival , Chick Embryo , Gene Expression Regulation, Neoplastic , Humans , Mice , Neurons/pathology , Proto-Oncogene Proteins c-myc/genetics
6.
J Neurosci ; 35(50): 16531-44, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26674877

ABSTRACT

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.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , DNA-Binding Proteins/genetics , MicroRNAs/genetics , Neuroblastoma/genetics , Neuroblastoma/pathology , Sympathetic Nervous System/growth & development , Adrenal Glands/metabolism , Animals , Cell Proliferation , Chick Embryo , DNA-Binding Proteins/physiology , Ganglia, Sympathetic/pathology , Mice , Mice, 129 Strain , MicroRNAs/physiology , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , RNA-Binding Proteins , Stem Cells/metabolism , Sympathetic Nervous System/physiology
7.
Dev Biol ; 400(2): 210-23, 2015 Apr 15.
Article in English | MEDLINE | ID: mdl-25661788

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.


Subject(s)
Adrenal Medulla/cytology , Chromaffin Cells/cytology , DEAD-box RNA Helicases/metabolism , Ganglia, Sympathetic/cytology , Ribonuclease III/metabolism , Adrenal Medulla/embryology , Adrenal Medulla/innervation , Adrenal Medulla/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Survival , Chromaffin Cells/metabolism , Ganglia, Sympathetic/embryology , Ganglia, Sympathetic/metabolism , Mice , Neural Crest/metabolism , Stem Cells/metabolism
8.
Cell Tissue Res ; 365(2): 225-32, 2016 08.
Article in English | MEDLINE | ID: mdl-27094431

ABSTRACT

The neural-crest-derived sympathoadrenal cell lineage gives rise to sympathetic neurons and to endocrine chromaffin cells of the adrenal medulla. Both cell types express a largely overlapping set of genes, including those coding for the molecular machinery related to the synthesis and exocytotic release of catecholamines. During their early development, sympathetic neurons and chromaffin cells rely on a shared transcription factor network that controls the establishment of these common features. Despite many similarities, mature sympathetic neurons and chromaffin cells significantly differ regarding their morphology and function. Most prominently, sympathetic neurons possess axons that are absent in mammalian adrenal chromaffin cells. The molecular mechanism underlying the divergent development of sympathoadrenal cells into neuronal and endocrine cells remains elusive. Mutational inactivation of the ribonuclease dicer hints at the importance of microRNAs in this diversification. We show here that miR-124 is detectable in developing sympathetic neurons but absent in chromaffin cell precursors. We further demonstrate that miR-124 promotes neurite elongation when transfected into cultured chromaffin cells indicating its capability to support the establishment of a neuronal morphology in non-neuronal sympathoadrenal cells. Our results also show that treatment of PC12 cells with the neurotrophin nerve growth factor leads to an upregulation of miR-124 expression and that inhibition of miR-124 reduces nerve-growth-factor-induced neurite outgrowth in PC12 cells. Thus, our data indicate that miR-124 contributes to the establishment of specific neuronal features in developing sympathoadrenal cells.


Subject(s)
Adrenal Medulla/cytology , Cell Lineage/genetics , Chromaffin Cells/metabolism , Gene Expression Profiling , MicroRNAs/metabolism , Neurites/metabolism , Sympathetic Nervous System/cytology , Amides/pharmacology , Animals , Cell Lineage/drug effects , Chromaffin Cells/drug effects , In Situ Hybridization , Mice , MicroRNAs/genetics , Nerve Growth Factors/pharmacology , Neurites/drug effects , PC12 Cells , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Rats , Up-Regulation/drug effects , Up-Regulation/genetics
9.
Stem Cells ; 33(2): 574-88, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25331182

ABSTRACT

Neural crest-derived stem cells (NCSCs) from the embryonic peripheral nervous system (PNS) can be reprogrammed in neurosphere (NS) culture to rNCSCs that produce central nervous system (CNS) progeny, including myelinating oligodendrocytes. Using global gene expression analysis we now demonstrate that rNCSCs completely lose their previous PNS characteristics and acquire the identity of neural stem cells derived from embryonic spinal cord. Reprogramming proceeds rapidly and results in a homogenous population of Olig2-, Sox3-, and Lex-positive CNS stem cells. Low-level expression of pluripotency inducing genes Oct4, Nanog, and Klf4 argues against a transient pluripotent state during reprogramming. The acquisition of CNS properties is prevented in the presence of BMP4 (BMP NCSCs) as shown by marker gene expression and the potential to produce PNS neurons and glia. In addition, genes characteristic for mesenchymal and perivascular progenitors are expressed, which suggests that BMP NCSCs are directed toward a pericyte progenitor/mesenchymal stem cell (MSC) fate. Adult NCSCs from mouse palate, an easily accessible source of adult NCSCs, display strikingly similar properties. They do not generate cells with CNS characteristics but lose the neural crest markers Sox10 and p75 and produce MSC-like cells. These findings show that embryonic NCSCs acquire a full CNS identity in NS culture. In contrast, MSC-like cells are generated from BMP NCSCs and pNCSCs, which reveals that postmigratory NCSCs are a source for MSC-like cells up to the adult stage.


Subject(s)
Antigens, Differentiation/metabolism , Embryo, Mammalian/metabolism , Neural Crest/metabolism , Neural Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Spinal Cord/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Kruppel-Like Factor 4 , Mice , Neural Crest/cytology , Neural Crest/embryology , Neural Stem Cells/cytology , Pluripotent Stem Cells/cytology , Spinal Cord/cytology , Spinal Cord/embryology
10.
J Neurosci ; 34(47): 15816-31, 2014 Nov 19.
Article in English | MEDLINE | ID: mdl-25411508

ABSTRACT

Specification of spinal cord neurons depends on gene regulation networks that impose distinct fates in neural progenitor cells (NPCs). Olig2 is a key transcription factor in these networks by inducing motor neuron (MN) specification and inhibiting interneuron identity. Despite the critical role of Olig2 in nervous system development and cancer progression, the upstream molecular mechanisms that control Olig2 gene transcription are not well understood. Here we demonstrate that Prox1, a transcription repressor and downstream target of proneural genes, suppresses Olig2 expression and therefore controls ventral spinal cord patterning. In particular, Prox1 is strongly expressed in V2 interneuron progenitors and largely excluded from Olig2+ MN progenitors (pMN). Gain- and loss-of-function studies in mouse NPCs and chick neural tube show that Prox1 is sufficient and necessary for the suppression of Olig2 expression and proper control of MN versus V2 interneuron identity. Mechanistically, Prox1 interacts with the regulatory elements of Olig2 gene locus in vivo and it is critical for proper Olig2 transcription regulation. Specifically, chromatin immunoprecipitation analysis in the mouse neural tube showed that endogenous Prox1 directly binds to the proximal promoter of the Olig2 gene locus, as well as to the K23 enhancer, which drives Olig2 expression in the pMN domain. Moreover, plasmid-based transcriptional assays in mouse NPCs suggest that Prox1 suppresses the activity of Olig2 gene promoter and K23 enhancer. These observations indicate that Prox1 controls binary fate decisions between MNs and V2 interneurons in NPCs via direct repression of Olig2 gene regulatory elements.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Homeodomain Proteins/physiology , Nerve Tissue Proteins/biosynthesis , Neurons/physiology , Spinal Cord/physiology , Tumor Suppressor Proteins/physiology , Animals , Chick Embryo , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Interneurons/physiology , Mice , Mice, Transgenic , Motor Neurons/physiology , Neural Stem Cells/physiology , Oligodendrocyte Transcription Factor 2 , Spinal Cord/cytology , Spinal Cord/growth & development
11.
J Neurosci ; 33(38): 15132-44, 2013 Sep 18.
Article in English | MEDLINE | ID: mdl-24048844

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.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Dendrites/metabolism , Ganglia, Sympathetic/cytology , Sensory Receptor Cells/cytology , Signal Transduction/physiology , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Archaeal Proteins/metabolism , Bone Morphogenetic Protein Receptors, Type I/deficiency , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Proteins/genetics , Cells, Cultured , DNA-Directed DNA Polymerase/metabolism , Embryo, Mammalian , Fluorescent Dyes/metabolism , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/metabolism , Imaging, Three-Dimensional , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Models, Neurological , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Smad4 Protein/deficiency , Smad4 Protein/genetics , Statistics, Nonparametric , Transcription Factors/metabolism
12.
Development ; 138(21): 4699-708, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21989914

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.


Subject(s)
Cell Proliferation , Cytokines/metabolism , Ganglia, Sympathetic/cytology , Neuroblastoma/physiopathology , Neurons/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Anaplastic Lymphoma Kinase , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chick Embryo , Cytokines/genetics , Enzyme Activation , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Humans , Midkine , Mutation , Neuroblastoma/pathology , Neurogenesis/physiology , Neurons/cytology , Neurons/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor, trkB/genetics , Receptor, trkB/metabolism
13.
Elife ; 122024 Mar 15.
Article in English | MEDLINE | ID: mdl-38488657

ABSTRACT

The pelvic organs (bladder, rectum, and sex organs) have been represented for a century as receiving autonomic innervation from two pathways - lumbar sympathetic and sacral parasympathetic - by way of a shared relay, the pelvic ganglion, conceived as an assemblage of sympathetic and parasympathetic neurons. Using single-cell RNA sequencing, we find that the mouse pelvic ganglion is made of four classes of neurons, distinct from both sympathetic and parasympathetic ones, albeit with a kinship to the former, but not the latter, through a complex genetic signature. We also show that spinal lumbar preganglionic neurons synapse in the pelvic ganglion onto equal numbers of noradrenergic and cholinergic cells, both of which therefore serve as sympathetic relays. Thus, the pelvic viscera receive no innervation from parasympathetic or typical sympathetic neurons, but instead from a divergent tail end of the sympathetic chains, in charge of its idiosyncratic functions.


Subject(s)
Neurons , Viscera , Mice , Animals , Neurons/physiology , Autonomic Nervous System , Sympathetic Nervous System/metabolism , Pelvis
14.
Dev Biol ; 363(1): 219-33, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22236961

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.


Subject(s)
Adrenergic Neurons/metabolism , Autonomic Nervous System/metabolism , Avian Proteins/genetics , Cell Differentiation/genetics , Homeodomain Proteins/genetics , Adrenergic Neurons/cytology , Animals , Autonomic Nervous System/cytology , Autonomic Nervous System/embryology , Avian Proteins/metabolism , Chick Embryo , Chickens , Dopamine beta-Hydroxylase/genetics , Dopamine beta-Hydroxylase/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Ganglia, Spinal/metabolism , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/embryology , Ganglia, Sympathetic/metabolism , Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Immunohistochemistry , In Situ Hybridization , Neural Crest/cytology , Neural Crest/embryology , Neural Crest/metabolism , Neural Tube/cytology , Neural Tube/embryology , Neural Tube/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
15.
Development ; 137(5): 775-84, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20147379

ABSTRACT

The highly related transcription factors Sox4 and Sox11 are expressed in the developing sympathetic nervous system. In the mouse, Sox11 appears first, whereas Sox4 is prevalent later. Using mouse mutagenesis and overexpression strategies in chicken, we studied the role of both SoxC proteins in this tissue. Neither Sox4 nor Sox11 predominantly functioned by promoting pan-neuronal or noradrenergic differentiation of sympathetic neurons as might have been expected from studies in neuronal precursors of the central nervous system. The transcriptional network that regulates the differentiation of sympathetic neurons remained intact and expression of noradrenergic markers showed only minor alterations. Instead, Sox11 was required in early sympathetic ganglia for proliferation of tyrosine hydroxylase-expressing cells, whereas Sox4 ensured the survival of these cells at later stages. In the absence of both Sox4 and Sox11, sympathetic ganglia remained hypoplastic throughout embryogenesis because of consecutive proliferation and survival defects. As a consequence, sympathetic ganglia were rudimentary in the adult and sympathetic innervation of target tissues was impaired leading to severe dysautonomia.


Subject(s)
SOXC Transcription Factors/physiology , Sympathetic Nervous System/embryology , Animals , Cell Lineage/genetics , Chick Embryo , Embryo, Mammalian , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/genetics , Norepinephrine/metabolism , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Sympathetic Nervous System/metabolism , Time Factors
17.
PLoS Biol ; 8(12): e1000565, 2010 Dec 21.
Article in English | MEDLINE | ID: mdl-21203589

ABSTRACT

Activation of Notch1 signaling in neural progenitor cells (NPCs) induces self-renewal and inhibits neurogenesis. Upon neuronal differentiation, NPCs overcome this inhibition, express proneural genes to induce Notch ligands, and activate Notch1 in neighboring NPCs. The molecular mechanism that coordinates Notch1 inactivation with initiation of neurogenesis remains elusive. Here, we provide evidence that Prox1, a transcription repressor and downstream target of proneural genes, counteracts Notch1 signaling via direct suppression of Notch1 gene expression. By expression studies in the developing spinal cord of chick and mouse embryo, we showed that Prox1 is limited to neuronal precursors residing between the Notch1+ NPCs and post-mitotic neurons. Physiological levels of Prox1 in this tissue are sufficient to allow binding at Notch1 promoter and they are critical for proper Notch1 transcriptional regulation in vivo. Gain-of-function studies in the chick neural tube and mouse NPCs suggest that Prox1-mediated suppression of Notch1 relieves its inhibition on neurogenesis and allows NPCs to exit the cell cycle and differentiate. Moreover, loss-of-function in the chick neural tube shows that Prox1 is necessary for suppression of Notch1 outside the ventricular zone, inhibition of active Notch signaling, down-regulation of NPC markers, and completion of neuronal differentiation program. Together these data suggest that Prox1 inhibits Notch1 gene expression to control the balance between NPC self-renewal and neuronal differentiation.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/metabolism , Neural Stem Cells/metabolism , Neural Tube/metabolism , Receptor, Notch1/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Cell Differentiation , Chick Embryo , Homeodomain Proteins/genetics , Mice , Neural Stem Cells/cytology , Neural Tube/cytology , Neural Tube/embryology , Neurogenesis , Receptor, Notch1/genetics , Signal Transduction , Tumor Suppressor Proteins/genetics
18.
Nat Commun ; 14(1): 2575, 2023 05 04.
Article in English | MEDLINE | ID: mdl-37142597

ABSTRACT

Noradrenergic and mesenchymal identities have been characterized in neuroblastoma cell lines according to their epigenetic landscapes and core regulatory circuitries. However, their relationship and relative contribution in patient tumors remain poorly defined. We now document spontaneous and reversible plasticity between the two identities, associated with epigenetic reprogramming, in several neuroblastoma models. Interestingly, xenografts with cells from each identity eventually harbor a noradrenergic phenotype suggesting that the microenvironment provides a powerful pressure towards this phenotype. Accordingly, such a noradrenergic cell identity is systematically observed in single-cell RNA-seq of 18 tumor biopsies and 15 PDX models. Yet, a subpopulation of these noradrenergic tumor cells presents with mesenchymal features that are shared with plasticity models, indicating that the plasticity described in these models has relevance in neuroblastoma patients. This work therefore emphasizes that intrinsic plasticity properties of neuroblastoma cells are dependent upon external cues of the environment to drive cell identity.


Subject(s)
Cell Plasticity , Neuroblastoma , Humans , Neuroblastoma/metabolism , Cell Line, Tumor , Tumor Microenvironment/genetics
19.
J Neurosci ; 31(17): 6379-91, 2011 Apr 27.
Article in English | MEDLINE | ID: mdl-21525278

ABSTRACT

Neural crest stem cells (NCSCs) give rise to the neurons and glia of the peripheral nervous system (PNS). NCSC-like cells can be isolated from multiple peripheral organs and maintained in neurosphere culture. Combining in vitro culture and transplantation, we show that expanded embryonic NCSC-like cells lose PNS traits and are reprogrammed to generate CNS cell types. When transplanted into the embryonic or adult mouse CNS, they differentiate predominantly into cells of the oligodendrocyte lineage without any signs of tumor formation. NCSC-derived oligodendrocytes generate CNS myelin and contribute to the repair of the myelin deficiency in shiverer mice. These results demonstrate a reprogramming of PNS progenitors to CNS fates without genetic modification and imply that PNS cells could be a potential source for cell-based CNS therapy.


Subject(s)
Brain Injuries/surgery , Gene Expression Regulation, Developmental/physiology , Myelin Sheath/metabolism , Neural Stem Cells/physiology , Oligodendroglia/physiology , Stem Cell Transplantation/methods , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Injuries/metabolism , Brain Injuries/physiopathology , Cell Differentiation/physiology , Cell Movement/drug effects , Cell Movement/genetics , Cells, Cultured , Disease Models, Animal , Embryo, Mammalian , Female , Ganglia, Spinal/cytology , Glial Fibrillary Acidic Protein/metabolism , Green Fluorescent Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubule-Associated Proteins/metabolism , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Myelin Sheath/ultrastructure , Nerve Tissue Proteins/metabolism , Neurofilament Proteins/metabolism , O Antigens/metabolism , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/ultrastructure , Transfection/methods , Tubulin/metabolism
20.
Dev Biol ; 355(1): 89-100, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21539825

ABSTRACT

Differentiation of sympathetic neurons is controlled by a group of transcription factors, including Phox2b, Ascl1, Hand2 and Gata3, induced by bone morphogenetic proteins (BMPs) in progenitors located in ganglion primordia at the dorsal aorta. Here, we address the function of the transcription factors AP-2ß and AP-2α, expressed in migrating neural crest cells (NCC) and maintained in sympathetic progenitors and differentiated neurons. The elimination of both AP-2α and AP-2ß results in the virtually complete absence of sympathetic and sensory ganglia due to apoptotic cell death of migrating NCC. In the AP-2ß knockout only sympathetic ganglia (SG) are targeted, leading to a reduction in ganglion size by about 40%, which is also caused by apoptotic death of neural crest progenitors. The conditional double knockout of AP-2α and AP-2ß in sympathetic progenitors and differentiated noradrenergic neurons results in a further decrease in neuron number, leading eventually to small sympathetic ganglion rudiments postnatally. The elimination of AP-2ß also leads to the complete absence of noradrenergic neurons of the Locus coeruleus (LC). Whereas AP-2α/ß transcription factors are in vivo not required for the onset or maintenance of noradrenergic differentiation, their essential survival functions are demonstrated for sympathetic progenitors and noradrenergic neurons.


Subject(s)
Ganglia, Sympathetic/metabolism , Neural Stem Cells/metabolism , Transcription Factor AP-2/metabolism , Animals , Cell Differentiation , Cell Survival , Cells, Cultured , Ganglia, Sensory/metabolism , Gene Expression Regulation, Developmental , Locus Coeruleus/metabolism , Mice , Mice, Knockout , Neural Crest/metabolism , Transcription Factor AP-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL