Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 40
1.
Cells ; 13(8)2024 Apr 13.
Article En | MEDLINE | ID: mdl-38667291

Both Hedgehog and androgen signaling pathways are known to promote myelin regeneration in the central nervous system. Remarkably, the combined administration of agonists of each pathway revealed their functional cooperation towards higher regeneration in demyelination models in males. Since multiple sclerosis, the most common demyelinating disease, predominates in women, and androgen effects were reported to diverge according to sex, it seemed essential to assess the existence of such cooperation in females. Here, we developed an intranasal formulation containing the Hedgehog signaling agonist SAG, either alone or in combination with testosterone. We show that SAG promotes myelin regeneration and presumably a pro-regenerative phenotype of microglia, thus mimicking the effects previously observed in males. However, unlike in males, the combined molecules failed to cooperate in the demyelinated females, as shown by the level of functional improvement observed. Consistent with this observation, SAG administered in the absence of testosterone amplified peripheral inflammation by presumably activating NK cells and thus counteracting a testosterone-induced reduction in Th17 cells when the molecules were combined. Altogether, the data uncover a sex-dependent effect of the Hedgehog signaling agonist SAG on the peripheral innate immune system that conditions its ability to cooperate or not with androgens in the context of demyelination.


Demyelinating Diseases , Testosterone , Animals , Female , Male , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Demyelinating Diseases/drug therapy , Mice , Testosterone/pharmacology , Hedgehog Proteins/metabolism , Hedgehog Proteins/agonists , Mice, Inbred C57BL , Central Nervous System/drug effects , Central Nervous System/immunology , Central Nervous System/pathology , Central Nervous System/metabolism , Smoothened Receptor/metabolism , Smoothened Receptor/agonists , Myelin Sheath/metabolism , Disease Models, Animal , Signal Transduction/drug effects , Immune System/drug effects , Microglia/drug effects , Microglia/metabolism , Microglia/immunology , Sex Characteristics
2.
Nat Commun ; 14(1): 1592, 2023 03 22.
Article En | MEDLINE | ID: mdl-36949062

Neuroprotective, anti-inflammatory, and remyelinating properties of androgens are well-characterized in demyelinated male mice and men suffering from multiple sclerosis. However, androgen effects mediated by the androgen receptor (AR), have been only poorly studied in females who make low androgen levels. Here, we show a predominant microglial AR expression in demyelinated lesions from female mice and women with multiple sclerosis, but virtually undetectable AR expression in lesions from male animals and men with multiple sclerosis. In female mice, androgens and estrogens act in a synergistic way while androgens drive microglia response towards regeneration. Transcriptomic comparisons of demyelinated mouse spinal cords indicate that, regardless of the sex, androgens up-regulate genes related to neuronal function integrity and myelin production. Depending on the sex, androgens down-regulate genes related to the immune system in females and lipid catabolism in males. Thus, androgens are required for proper myelin regeneration in females and therapeutic approaches of demyelinating diseases need to consider male-female differences.


Androgens , Multiple Sclerosis , Animals , Mice , Female , Male , Disease Models, Animal , Myelin Sheath/physiology , Multiple Sclerosis/genetics , Multiple Sclerosis/pathology , Neurons/pathology
3.
Int J Mol Sci ; 22(7)2021 Mar 26.
Article En | MEDLINE | ID: mdl-33810425

Motor neurons and their axons reaching the skeletal muscle have long been considered as the best characterized targets of the degenerative process observed in amyotrophic lateral sclerosis (ALS). However, the involvement of glial cells was also more recently reported. Although oligodendrocytes have been underestimated for a longer time than other cells, they are presently considered as critically involved in axonal injury and also conversely constitute a target for the toxic effects of the degenerative neurons. In the present review, we highlight the recent advances regarding oligodendroglial cell involvement in the pathogenesis of ALS. First, we present the oligodendroglial cells, the process of myelination, and the tight relationship between axons and myelin. The histological abnormalities observed in ALS and animal models of the disease are described, including myelin defects and oligodendroglial accumulation of pathological protein aggregates. Then, we present data that establish the existence of dysfunctional and degenerating oligodendroglial cells, the chain of events resulting in oligodendrocyte degeneration, and the most recent molecular mechanisms supporting oligodendrocyte death and dysfunction. Finally, we review the arguments in support of the primary versus secondary involvement of oligodendrocytes in the disease and discuss the therapeutic perspectives related to oligodendrocyte implication in ALS pathogenesis.


Amyotrophic Lateral Sclerosis/metabolism , Demyelinating Diseases/physiopathology , Oligodendroglia/cytology , Animals , Astrocytes/cytology , Axons/metabolism , Cell Death , Cell Lineage , Cell Proliferation , DNA-Binding Proteins/chemistry , Disease Models, Animal , Disease Progression , Humans , Mice , Motor Neurons/metabolism , Myelin Sheath/metabolism , Neuroglia/metabolism , Oxygen/chemistry , Prosencephalon/metabolism , Rats , Spinal Cord/metabolism , Superoxide Dismutase-1/metabolism
4.
Glia ; 69(6): 1369-1392, 2021 06.
Article En | MEDLINE | ID: mdl-33484204

Hedgehog morphogens control fundamental cellular processes during tissue development and regeneration. In the central nervous system (CNS), Hedgehog signaling has been implicated in oligodendrocyte and myelin production, where it functions in a concerted manner with other pathways. Since androgen receptor (AR) plays a key role in establishing the sexual phenotype of myelin during development and is required for spontaneous myelin regeneration in the adult CNS, we hypothesized the existence of a possible coordination between Hedgehog and androgen signals in oligodendrocyte and myelin production. Here, we report complementary activities of both pathways during early postnatal oligodendrogenesis further revealing that persistent Hedgehog signaling activation impedes myelin production. The data also uncover prominent pro-myelinating activity of testosterone and involvement of AR in the control of neural stem cell commitment toward the oligodendroglial lineage. In the context of CNS demyelination, we provide evidence for the functional cooperation of the pathways leading to acceleration of myelin regeneration that might be related to their respective role on microglial and astroglial responses, higher preservation of axonal integrity, lower neuroinflammation, and functional improvement of animals in an immune model of CNS demyelination. Strong decreases of deleterious cytokines in the CNS (GM-CSF, TNF-α, IL-17A) and spleen (IL-2, IFN-γ) stand as unique features of the combined drugs while the potent therapeutic activity of testosterone on peripheral immune cells contributes to increase tolerogenic CD11c+ dendritic cells, reduce the clonal expansion of conventional CD4+ T cells and increase CD4+ Foxp3+ regulatory T cells. Altogether, these data might open promising perspectives for demyelinating diseases.


Signal Transduction , Androgens , Animals , Demyelinating Diseases , Hedgehog Proteins , Myelin Sheath , Neuroinflammatory Diseases , Oligodendroglia , Testosterone
5.
Front Cell Neurosci ; 15: 801704, 2021.
Article En | MEDLINE | ID: mdl-35082605

The regeneration of myelin is known to restore axonal conduction velocity after a demyelinating event. Remyelination failure in the central nervous system contributes to the severity and progression of demyelinating diseases such as multiple sclerosis. Remyelination is controlled by many signaling pathways, such as the Sonic hedgehog (Shh) pathway, as shown by the canonical activation of its key effector Smoothened (Smo), which increases the proliferation of oligodendrocyte precursor cells via the upregulation of the transcription factor Gli1. On the other hand, the inhibition of Gli1 was also found to promote the recruitment of a subset of adult neural stem cells and their subsequent differentiation into oligodendrocytes. Since Smo is also able to transduce Shh signals via various non-canonical pathways such as the blockade of Gli1, we addressed the potential of non-canonical Smo signaling to contribute to oligodendroglial cell maturation in myelinating cells using the non-canonical Smo agonist GSA-10, which downregulates Gli1. Using the Oli-neuM cell line, we show that GSA-10 promotes Gli2 upregulation, MBP and MAL/OPALIN expression via Smo/AMP-activated Protein Kinase (AMPK) signaling, and efficiently increases the number of axonal contact/ensheathment for each oligodendroglial cell. Moreover, GSA-10 promotes the recruitment and differentiation of oligodendroglial progenitors into the demyelinated corpus callosum in vivo. Altogether, our data indicate that non-canonical signaling involving Smo/AMPK modulation and Gli1 downregulation promotes oligodendroglia maturation until axon engagement. Thus, GSA-10, by activation of this signaling pathway, represents a novel potential remyelinating agent.

6.
FASEB J ; 34(10): 13641-13653, 2020 10.
Article En | MEDLINE | ID: mdl-32862444

Leucine-rich repeat and immunoglobin-domain containing (LRRIG) proteins that are commonly involved in protein-protein interactions play important roles in nervous system development and maintenance. LINGO-1, one of this family members, is characterized as a negative regulator of neuronal survival, axonal regeneration, and oligodendrocyte precursor cell (OPC) differentiation into mature myelinating oligodendrocytes. Three LINGO-1 homologs named LINGO-2, LINGO-3, and LINGO-4 have been described. However, their relative expression and functions remain unexplored. Here, we show by in situ hybridization and quantitative polymerase chain reaction that the transcripts of LINGO homologs are differentially expressed in the central nervous system. The immunostaining of brain slices confirmed this observation and showed the co-expression of LINGO-1 with its homologs. Using BRET (bioluminescence resonance energy transfer) analysis, we demonstrate that LINGO proteins can physically interact with each of the other ones with comparable affinities and thus form the oligomeric states. Furthermore, co-immunoprecipitation experiments indicate that LINGO proteins form heterocomplexes in both heterologous systems and cortical neurons. Since LINGO-1 is a promising target for the treatment of demyelinating diseases, its ability to form heteromeric complexes reveals a new level of complexity in its functioning and opens the way for new strategies to achieve diverse and nuanced LINGO-1 regulation.


Brain/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Multimerization , Animals , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Protein Binding
7.
Front Cell Neurosci ; 14: 79, 2020.
Article En | MEDLINE | ID: mdl-32317939

Myelination is an essential process that consists of the ensheathment of axons by myelin. In the central nervous system (CNS), myelin is synthesized by oligodendrocytes. The proliferation, migration, and differentiation of oligodendrocyte precursor cells constitute a prerequisite before mature oligodendrocytes extend their processes around the axons and progressively generate a multilamellar lipidic sheath. Although myelination is predominately driven by oligodendrocytes, the other glial cells including astrocytes and microglia, also contribute to this process. The present review is an update of the most recent emerging mechanisms involving astrocyte and microglia in myelin production. The contribution of these cells will be first described during developmental myelination that occurs in the early postnatal period and is critical for the proper development of cognition and behavior. Then, we will report the novel findings regarding the beneficial or deleterious effects of astroglia and microglia, which respectively promote or impair the endogenous capacity of oligodendrocyte progenitor cells (OPCs) to induce spontaneous remyelination after myelin loss. Acute delineation of astrocyte and microglia activities and cross-talk should uncover the way towards novel therapeutic perspectives aimed at recovering proper myelination during development or at breaking down the barriers impeding the regeneration of the damaged myelin that occurs in CNS demyelinating diseases.

8.
Development ; 146(9)2019 05 02.
Article En | MEDLINE | ID: mdl-31048318

Myelination leads to the formation of myelin sheaths surrounding neuronal axons and is crucial for function, plasticity and repair of the central nervous system (CNS). It relies on the interaction of the axons and the oligodendrocytes: the glial cells producing CNS myelin. Here, we have investigated the role of a crucial component of the Sonic hedgehog (Shh) signalling pathway, the co-receptor Boc, in developmental and repairing myelination. During development, Boc mutant mice display a transient decrease in oligodendroglial cell density together with delayed myelination. Despite recovery of oligodendroglial cells at later stages, adult mutants still exhibit a lower production of myelin basic protein correlated with a significant decrease in the calibre of callosal axons and a reduced amount of the neurofilament NF-M. During myelin repair, the altered OPC differentiation observed in the mutant is reminiscent of the phenotype observed after blockade of Shh signalling. In addition, Boc mutant microglia/macrophages unexpectedly exhibit the apparent inability to transition from a highly to a faintly ramified morphology in vivo Altogether, these results identify Boc as an important component of myelin formation and repair.


Immunoglobulin G/metabolism , Myelin Sheath/metabolism , Receptors, Cell Surface/metabolism , Animals , Blotting, Western , Cell Differentiation/drug effects , Cells, Cultured , Cuprizone/pharmacology , Demyelinating Diseases/chemically induced , Demyelinating Diseases/metabolism , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Immunoglobulin G/genetics , Intermediate Filaments/drug effects , Intermediate Filaments/metabolism , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microglia/metabolism , Myelin Sheath/drug effects , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Receptors, Cell Surface/genetics
9.
Front Cell Neurosci ; 12: 305, 2018.
Article En | MEDLINE | ID: mdl-30237763

Since the discovery of its role as a morphogen directing ventral patterning of the spinal cord, the secreted protein Sonic Hedgehog (Shh) has been implicated in a wide array of events contributing to the development, maintenance and repair of the central nervous system (CNS). One of these events is the generation of oligodendrocytes, the glial cells of the CNS responsible for axon myelination. In embryo, the first oligodendroglial cells arise from the ventral ventricular zone in the developing brain and spinal cord where Shh induces the basic helix-loop-helix transcription factors Olig1 and Olig2 both necessary and sufficient for oligodendrocyte production. Later on, Shh signaling participates in the production of oligodendroglial cells in the dorsal ventricular-subventricular zone in the postnatal forebrain. Finally, the modulation of Hedgehog signaling activity promotes the repair of demyelinated lesions. This mini-review article focuses on the Shh-dependent molecular mechanisms involved in the spatial and temporal control of oligodendrocyte lineage appearance. The apparent intricacy of the roles of two essential components of Shh signaling, Smoothened and Gli1, in the postnatal production of myelin and its regeneration following a demyelinating event is also highlighted. A deeper understanding of the implication of each of the components that regulate oligodendrogenesis and myelination should beneficially influence the therapeutic strategies in the field of myelin diseases.

10.
Proc Natl Acad Sci U S A ; 113(51): 14829-14834, 2016 12 20.
Article En | MEDLINE | ID: mdl-27930320

Lost myelin can be replaced after injury or during demyelinating diseases in a regenerative process called remyelination. In the central nervous system (CNS), the myelin sheaths, which protect axons and allow the fast propagation of electrical impulses, are produced by oligodendrocytes. The abundance and widespread distribution of oligodendrocyte progenitors (OPs) within the adult CNS account for this remarkable regenerative potential. Here, we report a key role for the male gonad, testosterone, and androgen receptor (AR) in CNS remyelination. After lysolecithin-induced demyelination of the male mouse ventral spinal cord white matter, the recruitment of glial fibrillary acidic protein-expressing astrocytes was compromised in the absence of testes and testosterone signaling via AR. Concomitantly, the differentiation of OPs into oligodendrocytes forming myelin basic protein (MBP)+ and proteolipid protein-positive myelin was impaired. Instead, in the absence of astrocytes, axons were remyelinated by protein zero (P0)+ and peripheral myelin protein 22-kDa (PMP22)+ myelin, normally only produced by Schwann cells in the peripheral nervous system. Thus, testosterone favors astrocyte recruitment and spontaneous oligodendrocyte-mediated remyelination. This finding may have important implications for demyelinating diseases, psychiatric disorders, and cognitive aging. The testosterone dependency of CNS oligodendrocyte remyelination may have roots in the evolutionary history of the AR, because the receptor has evolved from an ancestral 3-ketosteroid receptor through gene duplication at the time when myelin appeared in jawed vertebrates.


Myelin Sheath/metabolism , Receptors, Androgen/metabolism , Remyelination , Androgens/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , Central Nervous System/metabolism , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Neuroglia/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Receptors, Steroid/metabolism , Schwann Cells/metabolism , Signal Transduction , Testis/metabolism , Testosterone/metabolism
11.
J Dev Biol ; 4(3)2016 Sep 08.
Article En | MEDLINE | ID: mdl-29615592

The Hedgehog morphogen aroused an enormous interest since it was characterized as an essential signal for ventral patterning of the spinal cord two decades ago. The pathway is notably implicated in the initial appearance of the progenitors of oligodendrocytes (OPCs), the glial cells of the central nervous system which after maturation are responsible for axon myelination. In accordance with the requirement for Hedgehog signaling in ventral patterning, the earliest identifiable cells in the oligodendrocyte lineage are derived from the ventral ventricular zone of the developing spinal cord and brain. Here, we present the current knowledge about the involvement of Hedgehog signaling in the strict spatial and temporal regulation which characterizes the initiation and progression of the oligodendrocyte lineage. We notably describe the ability of the Hedgehog signaling to tightly orchestrate the appearance of specific combinations of genes in concert with other pathways. We document the molecular mechanisms controlling Hedgehog temporal activity during OPC specification. The contribution of the pathway to aspects of OPC development different from their specification is also highlighted especially in the optic nerve. Finally, we report the data demonstrating that Hedgehog signaling-dependency is not a universal situation for oligodendrocyte generation as evidenced in the dorsal spinal cord in contrast to the dorsal forebrain.

12.
Med Sci (Paris) ; 31(12): 1115-25, 2015 Dec.
Article Fr | MEDLINE | ID: mdl-26672665

Development and repair of the nervous system are based on the existence of neural stem cells (NSCs) able to generate neurons and glial cells. Among the mechanisms that are involved in the control of embryo or adult NSCs, the Notch signalling plays a major role. In embryo, the pathway participates in the maintenance of NSCs during all steps of development of the central nervous system which starts with the production of neurons also called neurogenesis and continues with gliogenesis giving rise to astrocytes and oligodendrocytes. During the postnatal and adult period, Notch signalling is still present in the major neurogenic areas, the subventricular zone of the lateral ventricles and the subgranular zone of the hippocampus. In these regions, Notch maintains NSC quiescence, contributes to the heterogeneity of these cells and displays pleiotropic effects during the regeneration process occurring after a lesion.


Neural Stem Cells , Receptors, Notch/physiology , Signal Transduction/physiology , Age Factors , Aging , Animals , Humans , Nervous System/growth & development
13.
Neuroscientist ; 21(4): 356-71, 2015 Aug.
Article En | MEDLINE | ID: mdl-24743306

Since the discovery of the segment polarity gene Hedgehog in Drosophila three decades ago, our knowledge of Hedgehog signaling pathway has considerably improved and paved the way to a wide field of investigations in the developing and adult central nervous system. Its peculiar transduction mechanism together with its implication in tissue patterning, neural stem cell biology, and neural tissue homeostasis make Hedgehog pathway of interest in a high number of normal or pathological contexts. Consistent with its role during brain development, misregulation of Hedgehog signaling is associated with congenital diseases and tumorigenic processes while its recruitment in damaged neural tissue may be part of the repairing process. This review focuses on the most recent data regarding the Hedgehog pathway in the developing and adult central nervous system and also its relevance as a therapeutic target in brain and spinal cord diseases.


Brain/growth & development , Hedgehog Proteins/metabolism , Spinal Cord/growth & development , Animals , Brain/metabolism , Humans , Nervous System Malformations/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , Signal Transduction , Spinal Cord/metabolism
14.
Stem Cell Reports ; 3(2): 312-23, 2014 Aug 12.
Article En | MEDLINE | ID: mdl-25254344

In the adult brain, self-renewal is essential for the persistence of neural stem cells (NSCs) throughout life, but its regulation is still poorly understood. One NSC can give birth to two NSCs or one NSC and one transient progenitor. A correct balance is necessary for the maintenance of germinal areas, and understanding the molecular mechanisms underlying NSC division mode is clearly important. Here, we report a function of the Sonic Hedgehog (SHH) receptor Patched in the direct control of long-term NSC self-renewal in the subependymal zone. We show that genetic conditional activation of SHH signaling in adult NSCs leads to their expansion and the depletion of their direct progeny. These phenotypes are associated in vitro with an increase in NSC symmetric division in a process involving NOTCH signaling. Together, our results demonstrate a tight control of adult neurogenesis and NSC renewal driven by Patched.


Hedgehog Proteins/metabolism , Neural Stem Cells/cytology , Amino Acid Transport System X-AG/genetics , Animals , Brain/metabolism , Cell Proliferation/drug effects , Hedgehog Proteins/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neurogenesis , Patched Receptors , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Notch/metabolism , Selective Estrogen Receptor Modulators/pharmacology , Signal Transduction , Tamoxifen/pharmacology , Up-Regulation , Zinc Finger Protein GLI1
16.
Arterioscler Thromb Vasc Biol ; 33(12): 2858-66, 2013 Dec.
Article En | MEDLINE | ID: mdl-24135022

OBJECTIVE: The purpose of this study is to further document alteration of signal transduction pathways, more particularly of hedgehog (Hh) signaling, causing impaired ischemic muscle repair in old mice. APPROACH AND RESULTS: We used 12-week-old (young mice) and 20- to 24-month-old C57BL/6 mice (old mice) to investigate the activity of Hh signaling in the setting of hindlimb ischemia-induced angiogenesis and skeletal muscle repair. In this model, delayed ischemic muscle repair observed in old mice was associated with an impaired upregulation of Gli1. Sonic Hh expression was not different in old mice compared with young mice, whereas desert Hh (Dhh) expression was downregulated in the skeletal muscle of old mice both in healthy and ischemic conditions. The rescue of Dhh expression by gene therapy in old mice promoted ischemia-induced angiogenesis and increased nerve density; nevertheless, it failed to promote myogenesis or to increase Gli1 mRNA expression. After further investigation, we found that, in addition to Dhh, smoothened expression was significantly downregulated in old mice. We used smoothened haploinsufficient mice to demonstrate that smoothened knockdown by 50% is sufficient to impair activation of Hh signaling and ischemia-induced muscle repair. CONCLUSIONS: The present study demonstrates that Hh signaling is impaired in aged mice because of Dhh and smoothened downregulation. Moreover, it shows that hegdehog-dependent regulation of angiogenesis and myogenesis involves distinct mechanisms.


Aging/metabolism , Hedgehog Proteins/metabolism , Ischemia/metabolism , Muscle Development , Muscle, Skeletal/blood supply , Muscle, Skeletal/metabolism , Neovascularization, Physiologic , Age Factors , Aging/genetics , Animals , COS Cells , Chlorocebus aethiops , Disease Models, Animal , Gene Expression Regulation , Genetic Therapy , Hedgehog Proteins/genetics , Hindlimb , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Ischemia/genetics , Ischemia/pathology , Ischemia/physiopathology , Ischemia/therapy , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Regeneration , Signal Transduction , Smoothened Receptor , Transfection , Zinc Finger Protein GLI1
17.
Differentiation ; 85(4-5): 182-9, 2013.
Article En | MEDLINE | ID: mdl-23827460

The transgenic plp-GFP mouse line expressing the green fluorescent protein (GFP) driven by the mouse myelin proteolipid protein (plp) gene promoter has been previously used to study the contribution of the plp lineage to oligodendrocyte development in the embryonic brain. Here, we show that the GFP fluorescence reflects the developmental expression of proteolipid protein during the postnatal development until adulthood in brain slices and in primary cultures of plp-GFP(+) cells derived from postnatal animals. In the adult brain, plp-GFP-expressing cells are mature oligodendrocytes but not oligodendroglial progenitors. In the model of focal demyelination induced by lysolecithin (LPC) in the corpus callosum of adult plp-GFP animals, we observed an up-regulation of the morphogen Sonic Hedgehog (Shh) in the LPC-induced lesion but not in the control animals. Moreover, we show that the adenovirus-mediated transfer of Shh in the lesion results in the attenuation of the demyelination extent as evidenced by GFP fluorescence analysis in Shh-treated and control animals. Altogether these data show how plp-GFP fluorescence can be monitored to follow the oligodendrocyte lineage during demyelination and identify Shh morphogen as an important factor during repair.


Demyelinating Diseases/genetics , Myelin Proteolipid Protein/metabolism , Myelin Sheath/pathology , Oligodendroglia/metabolism , Promoter Regions, Genetic/physiology , Animals , Cell Differentiation/physiology , Cell Lineage , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Oligodendroglia/cytology , Oligodendroglia/pathology , Up-Regulation
18.
Best Pract Res Clin Endocrinol Metab ; 27(3): 429-42, 2013 Jun.
Article En | MEDLINE | ID: mdl-23856270

The calcium sensing receptor (CaSR) is expressed by subpopulations of neuronal and glial cells throughout the brain and is activated by extracellular calcium [Formula: see text] . During development, the CaSR regulates neuronal cell growth and migration as well as oligodendroglial maturation and function. Emerging evidence suggests that in nerve terminals, CaSR is implicated in synaptic plasticity and neurotransmission. In this review, we analyze the roles attributed to CaSR in regulating diverse brain functions, including central regulation of body fluid composition and blood pressure. We also discuss the potential relevance of Ca(2+)-sensing in brain by other family C G protein-coupled receptors. Finally, evidence that the CaSR contributes to the pathogenesis of various brain disorders raises the possibility that pharmacological modulators of the CaSR may have therapeutic benefit.


Calcium Signaling/physiology , Calcium/metabolism , Central Nervous System/metabolism , Receptors, Calcium-Sensing/physiology , Synaptic Transmission/physiology , Animals , Brain/metabolism , Cell Movement/physiology , Humans , Myelin Sheath/metabolism , Neuroglia/metabolism , Neuronal Plasticity/physiology , Neurons/metabolism
19.
Mol Aspects Med ; 34(2-3): 646-54, 2013.
Article En | MEDLINE | ID: mdl-23506897

The Na(+)-independent, high affinity choline carrier system proposed to supply choline for the synthesis of cell membrane phospholipids was recently associated with SLC44 family members (SLC44A1-5) also called choline-like transporter family. SLC44A1 is widely expressed throughout the nervous system in both neurons and oligodendrocytes, while SLC44A2-4 are mainly detected in peripheral tissues. The subcellular localization of the proteins was mainly addressed for SLC44A1 through the development of specific antibodies. SLC44A1 is detected in both the plasma and mitochondrial membranes where the protein is able to transport choline at high affinity and in a Na(+)-independent manner. The physiological relevance of SLC44A1 as a choline carrier is indicated by its likely involvement in membrane synthesis for cell growth or repair, and also by its role in phospholipid production for the generation of lung surfactant. Moreover, an autoimmune disease has been related to the blockade of SLC44A2 function, which results in the alteration of hair cells in the inner ear and leads to autoimmune hearing loss. In the alloimmune syndrome called transfusion-related acute lung injury, antibodies to SLC44A2 cause a deleterious aggregation of granulocytes. Therefore transporters of the SLC44 family represent attractive and promising targets for therapeutic and diagnostic applications regarding both immune and degenerative diseases.


Antigens, CD/genetics , Antigens, CD/physiology , Choline/metabolism , Gene Expression Regulation/physiology , Models, Molecular , Multigene Family/genetics , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/physiology , Antigens, CD/metabolism , Blotting, Northern , Cell Membrane/physiology , Humans , Models, Biological , Organic Cation Transport Proteins/metabolism , Phylogeny , Species Specificity
20.
Mol Pharmacol ; 83(5): 1020-9, 2013 May.
Article En | MEDLINE | ID: mdl-23448715

Activation of the Smoothened (Smo) receptor mediates Hedgehog (Hh) signaling. Hh inhibitors are in clinical trials for cancer, and small-molecule Smo agonists may have therapeutic interests in regenerative medicine. Here, we have generated and validated a pharmacophoric model for Smo agonists and used this model for the virtual screening of a library of commercially available compounds. Among the 20 top-scoring ligands, we have identified and characterized a novel quinolinecarboxamide derivative, propyl 4-(1-hexyl-4-hydroxy-2-oxo-1,2-dihydroquinoline-3-carboxamido) benzoate, (GSA-10), as a Smo agonist. GSA-10 fits to the agonist pharmacophoric model with two hydrogen bond acceptor groups and four hydrophobic regions. Using pharmacological, biochemical, and molecular approaches, we provide compelling evidence that GSA-10 acts at Smo to promote the differentiation of multipotent mesenchymal progenitor cells into osteoblasts. However, this molecule does not display the hallmarks of reference Smo agonists. Remarkably, GSA-10 does not recognize the classic bodipy-cyclopamine binding site. Its effect on cell differentiation is inhibited by Smo antagonists, such as MRT-83, SANT-1, LDE225, and M25 in the nanomolar range, by GDC-0449 in the micromolar range, but not by cyclopamine and CUR61414. Thus, GSA-10 allows the pharmacological characterization of a novel Smo active site, which is notably not targeted to the primary cilium and strongly potentiated by forskolin and cholera toxin. GSA-10 belongs to a new class of Smo agonists and will be helpful for dissecting Hh mechanism of action, with important implications in physiology and in therapy.


Quinolines/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Benzoates/pharmacology , Binding Sites/drug effects , Bone Morphogenetic Protein Receptors/metabolism , Cell Differentiation/drug effects , Cell Line , Cyclic AMP/metabolism , Cyclohexylamines/pharmacology , HEK293 Cells , Hedgehog Proteins/metabolism , Humans , Ligands , Receptors, G-Protein-Coupled/antagonists & inhibitors , Small Molecule Libraries , Smoothened Receptor , Thiophenes/pharmacology , Transcription Factors/metabolism , Wnt Proteins/metabolism , Zinc Finger Protein GLI1
...