Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Exp Neurol ; 368: 114520, 2023 10.
Article in English | MEDLINE | ID: mdl-37634698

ABSTRACT

Wobbly hedgehog syndrome (WHS) has been long considered to be a myelin disease primarily affecting the four-toed hedgehog. In this study, we have shown for the first time that demyelination is accompanied by extensive remyelination in WHS. However, remyelination is not enough to compensate for the axonal degeneration and neuronal loss, resulting in a progressive neurodegenerative disease reminiscent of progressive forms of multiple sclerosis (MS) in humans. Thus, understanding the pathological features of WHS may shed light on the disease progression in progressive MS and ultimately help to develop therapeutic strategies for both diseases.


Subject(s)
Multiple Sclerosis , Neurodegenerative Diseases , Humans , Animals , Hedgehogs , Neurodegenerative Diseases/genetics , Disease Progression , Memory
2.
bioRxiv ; 2023 Jul 15.
Article in English | MEDLINE | ID: mdl-37503221

ABSTRACT

Wobbly hedgehog syndrome (WHS) has been long considered to be a myelin disease primarily affecting the four-toed hedgehog. In this study, we have shown for the first time that demyelination is accompanied by extensive remyelination in WHS. However, remyelination is not enough to compensate for the axonal degeneration and neuronal loss, resulting in a progressive neurodegenerative disease reminiscent of progressive forms of multiple sclerosis (MS) in humans. Thus, understanding the pathological features of WHS may shed light on the disease progression in progressive MS and ultimately help to develop therapeutic strategies for both diseases. Highlights: Wobbly hedgehog syndrome (WHS) is a progressive neurodegenerative disease.Spongy degeneration of the brain and spinal cord is the diagnostic feature of WHS.WHS affected brain and spinal cord show extensive demyelination and remyelination.Axonal degeneration is accompanied by loss of neurons in WHS.

3.
STAR Protoc ; 4(1): 102072, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36853716

ABSTRACT

Here, we present a protocol to assess demyelination in the corpus callosum of an acute cuprizone mouse model, which is routinely used to induce demyelination for studying myelin regeneration in the rodent brain. We describe the tracing of neural stem cells via intraperitoneal injection of tamoxifen into adult Gli1CreERT2;Ai9 mice and the induction of demyelination with cuprizone diet. We also detail EdU administration, cryosectioning of the mouse brain, EdU labeling, and immunofluorescence staining to examine proliferation and myelination. For complete details on the use and execution of this protocol, please refer to Radecki et al. (2020).1.


Subject(s)
Demyelinating Diseases , Remyelination , Mice , Animals , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Fluorescent Antibody Technique , Cell Proliferation
4.
Cells ; 11(13)2022 07 02.
Article in English | MEDLINE | ID: mdl-35805185

ABSTRACT

Oligodendrogenesis is essential for replacing worn-out oligodendrocytes, promoting myelin plasticity, and for myelin repair following a demyelinating injury in the adult mammalian brain. Neural stem cells are an important source of oligodendrocytes in the adult brain; however, there are considerable differences in oligodendrogenesis from neural stem cells residing in different areas of the adult brain. Amongst the distinct niches containing neural stem cells, the subventricular zone lining the lateral ventricles and the subgranular zone in the dentate gyrus of the hippocampus are considered the principle areas of adult neurogenesis. In addition to these areas, radial glia-like cells, which are the precursors of neural stem cells, are found in the lining of the third ventricle, where they are called tanycytes, and in the cerebellum, where they are called Bergmann glia. In this review, we will describe the contribution and regulation of each of these niches in adult oligodendrogenesis.


Subject(s)
Neural Stem Cells , Animals , Brain , Cell Differentiation/physiology , Lateral Ventricles/physiology , Mammals , Neural Stem Cells/physiology , Neurogenesis/physiology
5.
STAR Protoc ; 3(1): 101153, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35146452

ABSTRACT

Neural stem cells (NSCs) from the subventricular zone (SVZ) of the mouse brain can be expanded in vitro and grown as neurospheres, which can be stored long-term in liquid nitrogen. Here, we present a protocol for isolation and culture of NSCs from the adult mouse SVZ. We describe how to grow and expand primary NSCs to neurospheres, followed by differentiation and nucleofection/pharmacological treatments. Finally, we describe RNA extraction, EdU labeling of the cells, and immunofluorescent analysis to examine their proliferation. For complete details on the use and execution of this protocol, please refer to Radecki et al. (2020).


Subject(s)
Lateral Ventricles , Neural Stem Cells , Animals , Cell Differentiation/genetics , Cell Proliferation , Mice
6.
J Neurosci ; 42(2): 183-201, 2022 01 12.
Article in English | MEDLINE | ID: mdl-34772739

ABSTRACT

Peripheral nerves are organized into discrete compartments. Axons, Schwann cells (SCs), and endoneurial fibroblasts (EFs) reside within the endoneurium and are surrounded by the perineurium, a cellular sheath comprised of layers of perineurial glia (PNG). SC secretion of Desert Hedgehog (Dhh) regulates this organization. In Dhh nulls, the perineurium is deficient and the endoneurium is subdivided into small compartments termed minifascicles. Human Dhh mutations cause a neuropathy with similar defects. Here we examine the role of Gli1, a canonical transcriptional effector of hedgehog signaling, in regulating peripheral nerve organization in mice of both genders. We identify PNG, EFs, and pericytes as Gli1-expressing cells by genetic fate mapping. Although expression of Dhh by SCs and Gli1 in target cells is coordinately regulated with myelination, Gli1 expression unexpectedly persists in Dhh null EFs. Thus, Gli1 is expressed in EFs noncanonically (i.e., independent of hedgehog signaling). Gli1 and Dhh also have nonredundant activities. Unlike Dhh nulls, Gli1 nulls have a normal perineurium. Like Dhh nulls, Gli1 nulls form minifascicles, which we show likely arise from EFs. Thus, Dhh and Gli1 are independent signals: Gli1 is dispensable for perineurial development but functions cooperatively with Dhh to drive normal endoneurial development. During development, Gli1 also regulates endoneurial extracellular matrix production, nerve vascular organization, and has modest, nonautonomous effects on SC sorting and myelination of axons. Finally, in adult nerves, induced deletion of Gli1 is sufficient to drive minifascicle formation. Thus, Gli1 regulates the development and is required to maintain the endoneurial architecture of peripheral nerves.SIGNIFICANCE STATEMENT Peripheral nerves are organized into distinct cellular/ECM compartments: the epineurium, perineurium, and endoneurium. This organization, with its associated cellular constituents, is critical for the structural and metabolic support of nerves and their response to injury. Here, we show that Gli1, a transcription factor normally expressed downstream of hedgehog signaling, is required for the proper organization of the endoneurium but not the perineurium. Unexpectedly, Gli1 expression by endoneurial cells is independent of, and functions nonredundantly with, Schwann Cell-derived Desert Hedgehog in regulating peripheral nerve architecture. These results further delineate how peripheral nerves acquire their distinctive organization during normal development, and highlight mechanisms that may regulate their reorganization in pathologic settings, including peripheral neuropathies and nerve injury.


Subject(s)
Peripheral Nerves/metabolism , Zinc Finger Protein GLI1/metabolism , Animals , Axons/metabolism , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/metabolism , Female , Gene Expression Regulation , Male , Mice , Mice, Knockout , Schwann Cells/metabolism , Zinc Finger Protein GLI1/genetics
7.
Sci Data ; 8(1): 278, 2021 10 28.
Article in English | MEDLINE | ID: mdl-34711861

ABSTRACT

In the adult mammalian brain, Gli1 expressing neural stem cells reside in the subventricular zone and their progeny are recruited to sites of demyelination in the white matter where they generate new oligodendrocytes, the myelin forming cells. Remarkably, genetic loss or pharmacologic inhibition of Gli1 enhances the efficacy of remyelination by these neural stem cells. To understand the molecular mechanisms involved, we performed a transcriptomic analysis of this Gli1-pool of neural stem cells. We compared murine NSCs with either intact or deficient Gli1 expression from adult mice on a control diet or on a cuprizone diet which induces widespread demyelination. These data will be a valuable resource for identifying therapeutic targets for enhancing remyelination in demyelinating diseases like multiple sclerosis.


Subject(s)
Demyelinating Diseases/genetics , Neural Stem Cells/cytology , Transcriptome , Zinc Finger Protein GLI1/genetics , Animals , Cuprizone , Mice , Oligodendroglia/cytology
8.
Glia ; 69(6): 1583-1604, 2021 06.
Article in English | MEDLINE | ID: mdl-33620118

ABSTRACT

Microgliosis is a prominent pathological feature in many neurological diseases including multiple sclerosis (MS), a progressive auto-immune demyelinating disorder. The precise role of microglia, parenchymal central nervous system (CNS) macrophages, during demyelination, and the relative contributions of peripheral macrophages are incompletely understood. Classical markers used to identify microglia do not reliably discriminate between microglia and peripheral macrophages, confounding analyses. Here, we use a genetic fate mapping strategy to identify microglia as predominant responders and key effectors of demyelination in the cuprizone (CUP) model. Colony-stimulating factor 1 (CSF1), also known as macrophage colony-stimulating factor (M-CSF) - a secreted cytokine that regulates microglia development and survival-is upregulated in demyelinated white matter lesions. Depletion of microglia with the CSF1R inhibitor PLX3397 greatly abrogates the demyelination, loss of oligodendrocytes, and reactive astrocytosis that results from CUP treatment. Electron microscopy (EM) and serial block face imaging show myelin sheaths remain intact in CUP treated mice depleted of microglia. However, these CUP-damaged myelin sheaths are lost and robustly phagocytosed upon-repopulation of microglia. Direct injection of CSF1 into CNS white matter induces focal microgliosis and demyelination indicating active CSF1 signaling can promote demyelination. Finally, mice defective in adopting a toxic astrocyte phenotype that is driven by microglia nevertheless demyelinate normally upon CUP treatment implicating microglia rather than astrocytes as the primary drivers of CUP-mediated demyelination. Together, these studies indicate activated microglia are required for and can drive demyelination directly and implicate CSF1 signaling in these events.


Subject(s)
Demyelinating Diseases , Microglia , Animals , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Disease Models, Animal , Macrophages , Mice , Receptors, Colony-Stimulating Factor , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Signal Transduction
9.
Stem Cell Reports ; 15(5): 1047-1055, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33125874

ABSTRACT

Enhancing repair of myelin is an important therapeutic goal in many neurological disorders characterized by demyelination. In the healthy adult brain, ventral neural stem cells (vNSCs) in the subventricular zone, marked by GLI1 expression, do not generate oligodendrocytes. However, in response to demyelination, their progeny are recruited to lesions where they differentiate into oligodendrocytes and ablation of GLI1 further enhances remyelination. GLI1 and GLI2 are closely related transcriptional activators but the role of GLI2 in remyelination by vNSCs is not clear. Here, we show that genetic ablation of Gli1 in vNSCs increases GLI2 expression and combined loss of both transcription factors decreases the recruitment and differentiation of their progeny in demyelinated lesions. These results indicate that GLI1 and GLI2 have distinct, non-redundant functions in vNSCs and their relative levels play an essential role in the response to demyelination.


Subject(s)
Demyelinating Diseases/metabolism , Neural Stem Cells/metabolism , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein Gli2/metabolism , Animals , Cell Differentiation , Demyelinating Diseases/genetics , Lateral Ventricles/metabolism , Mice , Mice, Inbred C57BL , Oligodendrocyte Precursor Cells/metabolism , Oligodendroglia/metabolism , Remyelination , Sequence Deletion , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein Gli2/genetics
10.
Nature ; 526(7573): 448-52, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26416758

ABSTRACT

Enhancing repair of myelin is an important but still elusive therapeutic goal in many neurological disorders. In multiple sclerosis, an inflammatory demyelinating disease, endogenous remyelination does occur but is frequently insufficient to restore function. Both parenchymal oligodendrocyte progenitor cells and endogenous adult neural stem cells resident within the subventricular zone are known sources of remyelinating cells. Here we characterize the contribution to remyelination of a subset of adult neural stem cells, identified by their expression of Gli1, a transcriptional effector of the sonic hedgehog pathway. We show that these cells are recruited from the subventricular zone to populate demyelinated lesions in the forebrain but never enter healthy, white matter tracts. Unexpectedly, recruitment of this pool of neural stem cells, and their differentiation into oligodendrocytes, is significantly enhanced by genetic or pharmacological inhibition of Gli1. Importantly, complete inhibition of canonical hedgehog signalling was ineffective, indicating that the role of Gli1 both in augmenting hedgehog signalling and in retarding myelination is specialized. Indeed, inhibition of Gli1 improves the functional outcome in a relapsing/remitting model of experimental autoimmune encephalomyelitis and is neuroprotective. Thus, endogenous neural stem cells can be mobilized for the repair of demyelinated lesions by inhibiting Gli1, identifying a new therapeutic avenue for the treatment of demyelinating disorders.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Kruppel-Like Transcription Factors/antagonists & inhibitors , Myelin Sheath/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/physiology , White Matter/metabolism , White Matter/pathology , Adult Stem Cells/cytology , Adult Stem Cells/metabolism , Animals , Cell Differentiation , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Hedgehog Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Lateral Ventricles , Mice , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Neuroprotective Agents/antagonists & inhibitors , Neuroprotective Agents/metabolism , Oligodendroglia/cytology , Prosencephalon/metabolism , Prosencephalon/pathology , Signal Transduction , White Matter/cytology , Zinc Finger Protein GLI1
11.
Dev Cell ; 34(2): 129-30, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-26218317

ABSTRACT

The mechanisms that drive the spiral wrapping of the myelin sheath around axons are poorly understood. Two papers in this issue of Developmental Cell demonstrate that actin disassembly, rather than actin assembly, predominates during oligodendrocyte maturation and is critical for the genesis of the central myelin sheath.


Subject(s)
Actin Cytoskeleton/metabolism , Actin-Related Protein 2-3 Complex/metabolism , Central Nervous System/growth & development , Myelin Sheath/physiology , Oligodendroglia/physiology , Animals
12.
Stroke ; 41(2): 357-62, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20019326

ABSTRACT

BACKGROUND AND PURPOSE: Bone morphogenetic proteins and their receptors are expressed in adult brains, and their expression levels increase after cerebral ischemia. The brain also expresses an inhibitor of bone morphogenetic protein signaling, noggin, but the role of noggin in ischemic disease outcome has not been studied. METHODS: We used transgenic mice overexpressing noggin to assess whether inhibition of bone morphogenetic protein signaling affects ischemic injury responses after permanent middle cerebral artery occlusion. RESULTS: Transgenic mice overexpressing noggin mice had significantly smaller infarct volumes and lower motor deficits compared to wild-type mice. CD11b(+) and IBA1(+) microglia along with oligodendroglial progenitors were significantly increased in transgenic mice overexpressing noggin mice at 14 days after permanent middle cerebral artery occlusion. CONCLUSIONS: These results provide genetic evidence that overexpression of noggin reduces ischemic brain injury after permanent middle cerebral artery occlusion via enhanced activation of microglia and oligodendrogenesis.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Brain Ischemia/genetics , Brain Ischemia/therapy , Carrier Proteins/genetics , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/therapy , Animals , Bone Morphogenetic Proteins/antagonists & inhibitors , Brain Infarction/pathology , Brain Infarction/physiopathology , Brain Infarction/therapy , Brain Ischemia/physiopathology , Cell Proliferation , Cytoprotection/physiology , Disease Models, Animal , Genetic Therapy/methods , Gliosis/pathology , Gliosis/physiopathology , Gliosis/therapy , Infarction, Middle Cerebral Artery/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Nerve Regeneration/physiology , Oligodendroglia/metabolism , Paresis/pathology , Paresis/physiopathology , Paresis/therapy , Recovery of Function/physiology , Stem Cells/metabolism
13.
J Neurosci ; 27(28): 7397-407, 2007 Jul 11.
Article in English | MEDLINE | ID: mdl-17626200

ABSTRACT

Progenitor cells that express the transcription factor olig1 generate several neural cell types including oligodendrocytes and GABAergic interneurons in the dorsal cortex. The fate of these progenitor cells is regulated by a number of signals including bone morphogenetic proteins (BMPs) secreted in the dorsal forebrain. BMPs signal by binding to heteromeric serine-threonine kinase receptors formed by type I (BMPR1a, BMPR1b, Alk2) and type II (BMPRII) subunits. To determine the specific role of the BMPR1a subunit in lineage commitment by olig1-expressing cells, we used a cre/loxP genetic approach to ablate BMPR1a in these cells while leaving signaling from other subunits intact. There was a reduction in numbers of immature oligodendrocytes in the BMPR1a-null mutant brains at birth. However, by postnatal day 20, the BMPR1a-null mice had a significant increase in the number of mature and immature oligodendrocytes compared with wild-type littermates. There was also an increase in the proportion of calbindin-positive interneurons in the dorsomedial cortex of BMPR1a-null mice at birth without any change in the number of parvalbumin- or calretinin-positive cells. These effects were attributable, at least in part, to a decrease in the length of the cell cycle in subventricular zone progenitor cells. Thus, our findings indicate that BMPR1a mediates the suppressive effects of BMP signaling on oligodendrocyte lineage commitment and on the specification of calbindin-positive interneurons in the dorsomedial cortex.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I/metabolism , Cerebral Cortex/cytology , Interneurons/cytology , Interneurons/metabolism , Oligodendroglia/cytology , S100 Calcium Binding Protein G/metabolism , Signal Transduction/physiology , Aging , Animals , Animals, Newborn , Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Morphogenetic Protein Receptors, Type I/deficiency , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Proteins/metabolism , Calbindins , Cell Count , Cell Cycle , Cell Lineage , Cellular Senescence , Cerebral Cortex/metabolism , Death , Interneurons/classification , Mice , Mice, Inbred C57BL , Mutation , Neurons/metabolism , Oligodendroglia/physiology , Smad Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , gamma-Aminobutyric Acid/metabolism
14.
Development ; 132(24): 5503-14, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16314487

ABSTRACT

Bone morphogenetic protein (BMP) and leukemia inhibitory factor (LIF) signaling both promote the differentiation of neural stem/progenitor cells into glial fibrillary acidic protein (GFAP) immunoreactive cells. This study compares the cellular and molecular characteristics, and the potentiality, of GFAP(+) cells generated by these different signaling pathways. Treatment of cultured embryonic subventricular zone (SVZ) progenitor cells with LIF generates GFAP(+) cells that have a bipolar/tripolar morphology, remain in cell cycle, contain progenitor cell markers and demonstrate self-renewal with enhanced neurogenesis - characteristics that are typical of adult SVZ and subgranular zone (SGZ) stem cells/astrocytes. By contrast, BMP-induced GFAP(+) cells are stellate, exit the cell cycle, and lack progenitor traits and self-renewal--characteristics that are typical of astrocytes in the non-neurogenic adult cortex. In vivo, transgenic overexpression of BMP4 increases the number of GFAP(+) astrocytes but depletes the GFAP(+) progenitor cell pool, whereas transgenic inhibition of BMP signaling increases the size of the GFAP(+) progenitor cell pool but reduces the overall numbers of astrocytes. We conclude that LIF and BMP signaling generate different astrocytic cell types, and propose that these cells are, respectively, adult progenitor cells and mature astrocytes.


Subject(s)
Astrocytes/cytology , Bone Morphogenetic Proteins/physiology , Embryonic Stem Cells/cytology , Glial Fibrillary Acidic Protein/biosynthesis , Leukemia Inhibitory Factor/physiology , Animals , Astrocytes/metabolism , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Carrier Proteins/genetics , Cell Cycle , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells/metabolism , Leukemia Inhibitory Factor/genetics , Mice , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , Signal Transduction
15.
Development ; 131(17): 4131-42, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15280210

ABSTRACT

Bone morphogenetic protein (BMP) signaling inhibits the generation of oligodendroglia and enhances generation of astrocytes by neural progenitor cells both in vitro and in vivo. This study examined the mechanisms underlying the effects of BMP signaling on glial lineage commitment. Treatment of cultured neural progenitor cells with BMP4 induced expression of all four members of the inhibitor of differentiation (ID) family of helix-loop-helix transcriptional inhibitors and blocked oligodendrocyte (OL) lineage commitment. Overexpression of Id4 or Id2 but not Id1 or Id3 in cultured progenitor cells reproduced both the inhibitory effects of BMP4 treatment on OL lineage commitment and the stimulatory effects on astrogliogenesis. Conversely, decreasing the levels of Id4 mRNA by RNA interference enhanced OL differentiation and inhibited the effects of BMP4 on glial lineage commitment. This suggests that induction of Id4 expression mediates effects of BMP signaling. Bacterial two-hybrid and co-immunoprecipitation studies demonstrated that ID4, and to a lesser extent ID2, complexed with the basic-helix-loop-helix transcription (bHLH) factors OLIG1 and OLIG2, which are required for the generation of OLs. By contrast, ID1 and ID3 did not complex with the OLIG proteins. In addition, the OLIG and ID proteins both interacted with the E2A proteins E12 and E47. Further, exposure of cultured progenitor cells to BMP4 changed the intracellular localization of OLIG1 and OLIG2 from a predominantly nuclear to a predominantly cytoplasmic localization. These observations suggest that the induction of ID4 and ID2, and their sequestration of both OLIG proteins and E2A proteins mediate the inhibitory effects of BMP signaling on OL lineage commitment and contribute to the generation of astrocytes.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Oligodendroglia/cytology , Animals , Astrocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors , Blotting, Western , Bone Morphogenetic Protein 4 , Cell Differentiation , Cell Line , Cell Lineage , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Green Fluorescent Proteins , Humans , Immunohistochemistry , Luminescent Proteins/metabolism , Mice , Microscopy, Fluorescence , Models, Biological , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Oligodendrocyte Transcription Factor 2 , Phenotype , Precipitin Tests , Protein Binding , RNA Interference , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Time Factors , Transcription Factors/metabolism , Transfection , Two-Hybrid System Techniques
16.
Development ; 131(5): 1175-86, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14973275

ABSTRACT

The role of target-derived BMP signaling in development of sensory ganglia and the sensory innervation of the skin was examined in transgenic animals that overexpress either the BMP inhibitor noggin or BMP4 under the control of a keratin 14 (K14) promoter. Overexpression of noggin resulted in a significant increase in the number of neurons in the trigeminal and dorsal root ganglia. Conversely, overexpression of BMP4 resulted in a significant decrease in the number of dorsal root ganglion neurons. There was no significant change in proliferation of trigeminal ganglion neurons in the noggin transgenic animals, and neuron numbers did not undergo the normal developmental decrease between E12.5 and the adult, suggesting that programmed cell death was decreased in these animals. The increase in neuron numbers in the K14-noggin animals was followed by an extraordinary increase in the density of innervation in the skin and a marked change in the pattern of innervation by different types of fibers. Conversely, the density of innervation of the skin was decreased in the BMP4 overexpressing animals. Further Merkel cells and their innervation were increased in the K14-noggin mice and decreased in the K14-BMP4 mice. The changes in neuron numbers and the density of innervation were not accompanied by a change in the levels of neurotrophins in the skin. These findings indicate that the normal developmental decrease in neuron numbers in sensory ganglia depends upon BMP signaling, and that BMPs may limit both the final neuron number in sensory ganglia as well as the extent of innervation of targets. Coupled with prior observations, this suggests that BMP signaling may regulate the acquisition of dependence of neurons on neurotrophins for survival, as well as their dependence on target-derived neurotrophins for determining the density of innervation of the target.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Neurons, Afferent/cytology , Neurons, Afferent/metabolism , Peripheral Nerves/embryology , Animals , Base Sequence , Bone Morphogenetic Protein 4 , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/pharmacology , Carrier Proteins , Cell Count , DNA, Complementary/genetics , Ganglia, Spinal/embryology , Ganglia, Spinal/growth & development , Ganglia, Spinal/metabolism , Gene Expression Regulation, Developmental , In Situ Hybridization , Keratin-14 , Keratins/genetics , Mice , Mice, Transgenic , Nerve Growth Factor/pharmacology , Neurons, Afferent/drug effects , Peripheral Nerves/growth & development , Peripheral Nerves/metabolism , Promoter Regions, Genetic , Proteins/genetics , Proteins/metabolism , Signal Transduction , Skin/innervation , Trigeminal Ganglion/embryology , Trigeminal Ganglion/growth & development , Trigeminal Ganglion/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...