Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Glia ; 72(8): 1518-1540, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38794866

ABSTRACT

In the central nervous system, the formation of myelin by oligodendrocytes (OLs) relies on the switch from the polymerization of the actin cytoskeleton to its depolymerization. The molecular mechanisms that trigger this switch have yet to be elucidated. Here, we identified P21-activated kinase 1 (PAK1) as a major regulator of actin depolymerization in OLs. Our results demonstrate that PAK1 accumulates in OLs in a kinase-inhibited form, triggering actin disassembly and, consequently, myelin membrane expansion. Remarkably, proteomic analysis of PAK1 binding partners enabled the identification of NF2/Merlin as its endogenous inhibitor. Our findings indicate that Nf2 knockdown in OLs results in PAK1 activation, actin polymerization, and a reduction in OL myelin membrane expansion. This effect is rescued by treatment with a PAK1 inhibitor. We also provide evidence that the specific Pak1 loss-of-function in oligodendroglia stimulates the thickening of myelin sheaths in vivo. Overall, our data indicate that the antagonistic actions of PAK1 and NF2/Merlin on the actin cytoskeleton of the OLs are critical for proper myelin formation. These findings have broad mechanistic and therapeutic implications in demyelinating diseases and neurodevelopmental disorders.


Subject(s)
Myelin Sheath , Oligodendroglia , p21-Activated Kinases , p21-Activated Kinases/metabolism , Oligodendroglia/metabolism , Animals , Myelin Sheath/metabolism , Neurofibromin 2/metabolism , Neurofibromin 2/genetics , Rats , Actins/metabolism , Cells, Cultured , Mice , Mice, Inbred C57BL , Actin Cytoskeleton/metabolism
2.
Stem Cell Reports ; 17(11): 2467-2483, 2022 11 08.
Article in English | MEDLINE | ID: mdl-36351367

ABSTRACT

The presence of putative stem/progenitor cells has been suggested in adult peripheral nervous system (PNS) tissue, including the dorsal root ganglion (DRG). To date, their identification and fate in pathophysiological conditions have not been addressed. Combining multiple in vitro and in vivo approaches, we identified the presence of stem cells in the adult DRG satellite glial population, and progenitors were present in the DRGs and sciatic nerve. Cell-specific transgenic mouse lines highlighted the proliferative potential of DRG stem cells and progenitors in vitro. DRG stem cells had gliogenic and neurogenic potentials, whereas progenitors were essentially gliogenic. Lineage tracing showed that, under physiological conditions, adult DRG stem cells maintained DRG homeostasis by supplying satellite glia. Under pathological conditions, adult DRG stem cells replaced DRG neurons lost to injury in addition of renewing the satellite glial pool. These novel findings open new avenues for development of therapeutic strategies targeting DRG stem cells for PNS disorders.


Subject(s)
Adult Stem Cells , Ganglia, Spinal , Mice , Animals , Neuroglia , Neurons , Stem Cells
3.
Brain Commun ; 4(1): fcac025, 2022.
Article in English | MEDLINE | ID: mdl-35224490

ABSTRACT

The p70 ribosomal S6 kinases (p70 ribosomal S6 kinase 1 and p70 ribosomal S6 kinase 2) are downstream targets of the mechanistic target of rapamycin signalling pathway. p70 ribosomal S6 kinase 1 specifically has demonstrated functions in regulating cell size in Drosophila and in insulin-sensitive cell populations in mammals. Prior studies demonstrated that the mechanistic target of the rapamycin pathway promotes oligodendrocyte differentiation and developmental myelination; however, how the immediate downstream targets of mechanistic target of rapamycin regulate these processes has not been elucidated. Here, we tested the hypothesis that p70 ribosomal S6 kinase 1 regulates oligodendrocyte differentiation during developmental myelination and remyelination processes in the CNS. We demonstrate that p70 ribosomal S6 kinase activity peaks in oligodendrocyte lineage cells at the time when they transition to myelinating oligodendrocytes during developmental myelination in the mouse spinal cord. We further show p70 ribosomal S6 kinase activity in differentiating oligodendrocytes in acute demyelinating lesions induced by lysophosphatidylcholine injection or by experimental autoimmune encephalomyelitis in mice. In demyelinated lesions, the expression of the p70 ribosomal S6 kinase target, phosphorylated S6 ribosomal protein, was transient and highest in maturing oligodendrocytes. Interestingly, we also identified p70 ribosomal S6 kinase activity in oligodendrocyte lineage cells in active multiple sclerosis lesions. Consistent with its predicted function in promoting oligodendrocyte differentiation, we demonstrate that specifically inhibiting p70 ribosomal S6 kinase 1 in cultured oligodendrocyte precursor cells significantly impairs cell lineage progression and expression of myelin basic protein. Finally, we used zebrafish to show in vivo that inhibiting p70 ribosomal S6 kinase 1 function in oligodendroglial cells reduces their differentiation and the number of myelin internodes produced. These data reveal an essential function of p70 ribosomal S6 kinase 1 in promoting oligodendrocyte differentiation during development and remyelination across multiple species.

4.
J Med Chem ; 64(9): 5667-5688, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33949859

ABSTRACT

Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system (CNS) that causes severe motor, sensory, and cognitive impairments. Kallikrein-related peptidase (KLK)6 is the most abundant serine protease secreted in the CNS, mainly by oligodendrocytes, the myelin-producing cells of the CNS, and KLK6 is assumed to be a robust biomarker of MS, since it is highly increased in the cerebrospinal fluid (CSF) of MS patients. Here, we report the design and biological evaluation of KLK6's low-molecular-weight inhibitors, para-aminobenzyl derivatives. Interestingly, selected hit compounds were selective of the KLK6 proteolytic network encompassing KLK1 and plasmin that also participate in the development of MS physiopathology. Moreover, hits were found noncytotoxic on primary cultures of murine neurons and oligodendrocyte precursor cells (OPCs). Among them, two compounds (32 and 42) were shown to promote the differentiation of OPCs into mature oligodendrocytes in vitro constituting thus emerging leads for the development of regenerative therapies.


Subject(s)
Cell Differentiation/drug effects , Kallikreins/antagonists & inhibitors , Serine Proteinase Inhibitors/pharmacology , Animals , Benzene Derivatives/chemistry , Benzene Derivatives/metabolism , Benzene Derivatives/pharmacology , Binding Sites , Catalytic Domain , Cells, Cultured , Drug Design , Fibrinolysin/antagonists & inhibitors , Fibrinolysin/metabolism , Humans , Kallikreins/metabolism , Kinetics , Mice , Molecular Docking Simulation , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Neurons/cytology , Neurons/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Structure-Activity Relationship
5.
Glia ; 69(5): 1094-1109, 2021 05.
Article in English | MEDLINE | ID: mdl-33301181

ABSTRACT

Oligodendrocytes are extensively coupled to astrocytes, a phenomenon ensuring glial homeostasis and maintenance of central nervous system myelin. Molecular disruption of this communication occurs in demyelinating diseases such as multiple sclerosis. Less is known about the vulnerability and reconstruction of the panglial network during adult demyelination-remyelination. Here, we took advantage of lysolcithin-induced demyelination to investigate the expression dynamics of the oligodendrocyte specific connexin 47 (Cx47) and to some extent that of astrocyte Cx43, and whether this dynamic could be modulated by grafted induced pluripotent stem cell (iPSC)-neural progeny. Our data show that disruption of Cx43-Cx47 mediated hetero-cellular gap-junction intercellular communication following demyelination is larger in size than demyelination. Loss of Cx47 expression is timely rescued during remyelination and accelerated by the grafted neural precursors. Moreover, mouse and human iPSC-derived oligodendrocytes express Cx47, which co-labels with astrocyte Cx43, indicating their integration into the panglial network. These data suggest that in rodents, full lesion repair following transplantation occurs by panglial reconstruction in addition to remyelination. Targeting panglial elements by cell therapy or pharmacological compounds may help accelerating or stabilizing re/myelination in myelin disorders.


Subject(s)
Connexins , Induced Pluripotent Stem Cells , Multiple Sclerosis , Remyelination , Animals , Astrocytes , Connexin 43/genetics , Mice , Oligodendroglia
6.
Acta Neuropathol ; 138(3): 457-476, 2019 09.
Article in English | MEDLINE | ID: mdl-31011859

ABSTRACT

Schwann cells (SC) enter the central nervous system (CNS) in pathophysiological conditions. However, how SC invade the CNS to remyelinate central axons remains undetermined. We studied SC migratory behavior ex vivo and in vivo after exogenous transplantation in the demyelinated spinal cord. The data highlight for the first time that SC migrate preferentially along blood vessels in perivascular extracellular matrix (ECM), avoiding CNS myelin. We demonstrate in vitro and in vivo that this migration route occurs by virtue of a dual mode of action of Eph/ephrin signaling. Indeed, EphrinB3, enriched in myelin, interacts with SC Eph receptors, to drive SC away from CNS myelin, and triggers their preferential adhesion to ECM components, such as fibronectin via integrinß1 interactions. This complex interplay enhances SC migration along the blood vessel network and together with lesion-induced vascular remodeling facilitates their timely invasion of the lesion site. These novel findings elucidate the mechanism by which SC invade and contribute to spinal cord repair.


Subject(s)
Blood Vessels , Cell Movement/physiology , Ephrin-B3/metabolism , Remyelination/physiology , Schwann Cells/physiology , Spinal Cord/metabolism , Animals , Demyelinating Diseases/pathology , Female , Fibronectins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/physiology , Spinal Cord/pathology
7.
Glia ; 66(10): 2221-2232, 2018 10.
Article in English | MEDLINE | ID: mdl-30152028

ABSTRACT

Oligodendrocyte development is a critical process timely and spatially regulated to ensure proper myelination of the central nervous system. HMG-box transcription factors are key regulators of oligodendrocyte lineage progression. Among these factors, Sox17 was previously identified as a positive regulator of oligodendrocyte development. However, the role of Sox17 in oligodendroglial cell lineage progression and differentiation is still poorly understood. To define the functional role of Sox17, we generated new transgenic mouse models with inducible overexpression of Sox17, specifically in oligodendroglial cells. Here, we report that gain of Sox17 function has no effect on oligodendrocyte progenitor cells (OPCs) specification. During early postnatal development, Sox17 overexpression increases the pool of OPCs at the expense of differentiated oligodendrocytes. However, the oligodendroglial cell population, OPC proliferation and apoptosis remained unchanged in Sox17 transgenic mice. RNA sequencing, quantitative RT-PCR and immunohistochemical analysis showed that Sox17 represses the expression of the major myelin genes, resulting in a severe CNS hypomyelination. Overall, our data highlight an unexpected role for Sox17 as a negative regulator of OPC differentiation and myelination, suggesting stage specific functions for this factor during oligodendroglial cell lineage progression.


Subject(s)
Cell Differentiation/physiology , HMGB Proteins/metabolism , Oligodendrocyte Precursor Cells/metabolism , SOXF Transcription Factors/metabolism , Animals , Apoptosis/physiology , Gene Expression Regulation, Developmental , HMGB Proteins/genetics , Mice, Transgenic , Oligodendrocyte Precursor Cells/pathology , Oligodendroglia/metabolism , Oligodendroglia/pathology , SOXF Transcription Factors/genetics , Spinal Cord/growth & development , Spinal Cord/metabolism , Spinal Cord/pathology , Transcriptome
8.
Stem Cells ; 34(4): 984-96, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26676415

ABSTRACT

Pelizaeus-Merzbacher disease (PMD) results from an X-linked misexpression of proteolipid protein 1 (PLP1). This leukodystrophy causes severe hypomyelination with progressive inflammation, leading to neurological dysfunctions and shortened life expectancy. While no cure exists for PMD, experimental cell-based therapy in the dysmyelinated shiverer model suggested that human oligodendrocyte progenitor cells (hOPCs) or human neural precursor cells (hNPCs) are promising candidates to treat myelinopathies. However, the fate and restorative advantages of human NPCs/OPCs in a relevant model of PMD has not yet been addressed. Using a model of Plp1 overexpression, resulting in demyelination with progressive inflammation, we compared side-by-side the therapeutic benefits of intracerebrally grafted hNPCs and hOPCs. Our findings reveal equal integration of the donor cells within presumptive white matter tracks. While the onset of exogenous remyelination was earlier in hOPCs-grafted mice than in hNPC-grafted mice, extended lifespan occurred only in hNPCs-grafted animals. This improved survival was correlated with reduced neuroinflammation (microglial and astrocytosis loads) and microglia polarization toward M2-like phenotype followed by remyelination. Thus modulation of neuroinflammation combined with myelin restoration is crucial to prevent PMD pathology progression and ensure successful rescue of PMD mice. These findings should help to design novel therapeutic strategies combining immunomodulation and stem/progenitor cell-based therapy for disorders associating hypomyelination with inflammation as observed in PMD.


Subject(s)
Immunity, Innate , Inflammation/therapy , Neural Stem Cells/transplantation , Oligodendroglia/transplantation , Pelizaeus-Merzbacher Disease/therapy , Animals , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Gene Expression Regulation, Developmental , Humans , Immunomodulation , Inflammation/immunology , Inflammation/pathology , Mice , Microglia/immunology , Microglia/pathology , Myelin Proteolipid Protein/biosynthesis , Myelin Sheath/metabolism , Neural Stem Cells/immunology , Oligodendroglia/immunology , Pelizaeus-Merzbacher Disease/immunology , Pelizaeus-Merzbacher Disease/pathology , Regeneration
9.
J Clin Invest ; 125(9): 3642-56, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26301815

ABSTRACT

Induced pluripotent stem cell-derived (iPS-derived) neural precursor cells may represent the ideal autologous cell source for cell-based therapy to promote remyelination and neuroprotection in myelin diseases. So far, the therapeutic potential of reprogrammed cells has been evaluated in neonatal demyelinating models. However, the repair efficacy and safety of these cells has not been well addressed in the demyelinated adult CNS, which has decreased cell plasticity and scarring. Moreover, it is not clear if these induced pluripotent-derived cells have the same reparative capacity as physiologically committed CNS-derived precursors. Here, we performed a side-by-side comparison of CNS-derived and skin-derived neural precursors in culture and following engraftment in murine models of adult spinal cord demyelination. Grafted induced neural precursors exhibited a high capacity for survival, safe integration, migration, and timely differentiation into mature bona fide oligodendrocytes. Moreover, grafted skin-derived neural precursors generated compact myelin around host axons and restored nodes of Ranvier and conduction velocity as efficiently as CNS-derived precursors while outcompeting endogenous cells. Together, these results provide important insights into the biology of reprogrammed cells in adult demyelinating conditions and support use of these cells for regenerative biomedicine of myelin diseases that affect the adult CNS.


Subject(s)
Cell Differentiation , Demyelinating Diseases/therapy , Myelin Sheath/metabolism , Neural Stem Cells/metabolism , Skin/metabolism , Stem Cell Transplantation , Animals , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mice , Mice, Knockout , Neural Stem Cells/pathology , Neural Stem Cells/transplantation , Regenerative Medicine/methods , Skin/pathology
10.
Proc Natl Acad Sci U S A ; 112(24): 7587-92, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26023184

ABSTRACT

The identification of new pathways governing myelination provides innovative avenues for remyelination. Liver X receptors (LXRs) α and ß are nuclear receptors activated by oxysterols that originated from the oxidation of cholesterol. They are crucial for cholesterol homeostasis, a major lipid constituent of myelin sheaths that are formed by oligodendrocytes. However, the role of LXRs in myelin generation and maintenance is poorly understood. Here, we show that LXRs are involved in myelination and remyelination processes. LXRs and their ligands are present in oligodendrocytes. We found that mice invalidated for LXRs exhibit altered motor coordination and spatial learning, thinner myelin sheaths, and reduced myelin gene expression. Conversely, activation of LXRs by either 25-hydroxycholesterol or synthetic TO901317 stimulates myelin gene expression at the promoter, mRNA, and protein levels, directly implicating LXRα/ß in the transcriptional control of myelin gene expression. Interestingly, activation of LXRs also promotes oligodendroglial cell maturation and remyelination after lysolecithin-induced demyelination of organotypic cerebellar slice cultures. Together, our findings represent a conceptual advance in the transcriptional control of myelin gene expression and strongly support a new role of LXRs as positive modulators in central (re)myelination processes.


Subject(s)
Cerebellum/physiology , Myelin Sheath/physiology , Orphan Nuclear Receptors/physiology , Animals , Cell Differentiation/drug effects , Cerebellum/cytology , Cerebellum/drug effects , Cholesterol/metabolism , Gene Expression Regulation/drug effects , Homeostasis , Hydrocarbons, Fluorinated/pharmacology , Hydroxycholesterols/pharmacology , Liver X Receptors , Male , Mice , Mice, Knockout , Myelin Sheath/drug effects , Myelin Sheath/genetics , Oligodendroglia/cytology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Organ Culture Techniques , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/deficiency , Promoter Regions, Genetic , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spatial Learning/drug effects , Spatial Learning/physiology , Sulfonamides/pharmacology
11.
Stem Cells ; 33(6): 2011-24, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25786382

ABSTRACT

It has been proposed that the adult dorsal root ganglia (DRG) harbor neural stem/progenitor cells (NPCs) derived from the neural crest. However, the thorough characterization of their stemness and differentiation plasticity was not addressed. In this study, we investigated adult DRG-NPC stem cell properties overtime, and their fate when ectopically grafted in the central nervous system. We compared them in vitro and in vivo to the well-characterized adult spinal cord-NPCs derived from the same donors. Using micro-dissection and neurosphere cultures, we demonstrate that adult DRG-NPCs have quasi unlimited self-expansion capacities without compromising their tissue specific molecular signature. Moreover, they differentiate into multiple peripheral lineages in vitro. After transplantation, adult DRG-NPCs generate pericytes in the developing forebrain but remyelinating Schwann cells in response to spinal cord demyelination. In addition, we show that axonal and endothelial/astrocytic factors as well astrocytes regulate the fate of adult DRG-NPCs in culture. Although the adult DRG-NPC multipotency is restricted to the neural crest lineage, their dual responsiveness to developmental and lesion cues highlights their impressive adaptive and repair potentials making them valuable targets for regenerative medicine.


Subject(s)
Cell Differentiation/physiology , Demyelinating Diseases/pathology , Ganglia, Spinal/cytology , Myelin Sheath/metabolism , Pericytes/cytology , Schwann Cells/cytology , Adult Stem Cells/cytology , Animals , Cells, Cultured , Demyelinating Diseases/therapy , Ganglia, Spinal/metabolism , Mice, Inbred C57BL , Mice, Nude , Nerve Regeneration/physiology , Neural Crest/cytology , Neurons/cytology
12.
Brain ; 138(Pt 1): 120-35, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25564492

ABSTRACT

The basic helix-loop-helix transcription factor Olig2 is a key determinant for the specification of neural precursor cells into oligodendrocyte progenitor cells. However, the functional role of Olig2 in oligodendrocyte migration and differentiation remains elusive both during developmental myelination and under demyelinating conditions of the adult central nervous system. To decipher Olig2 functions, we generated transgenic mice (TetOlig2:Sox10(rtTA/+)) overexpressing Olig2 in Sox10(+) oligodendroglial cells in a doxycycline inducible manner. We show that Olig2 overexpression increases the generation of differentiated oligodendrocytes, leading to precocious myelination of the central nervous system. Unexpectedly, we found that gain of Olig2 function in oligodendrocyte progenitor cells enhances their migration rate. To determine whether Olig2 overexpression in adult oligodendrocyte progenitor cells promotes oligodendrocyte regeneration for myelin repair, we induced lysophosphatidylcholine demyelination in the corpus callosum of TetOlig2:Sox10(rtTA/+) and control mice. We found that Olig2 overexpression enhanced oligodendrocyte progenitor cell differentiation and remyelination. To assess the relevance of these findings in demyelinating diseases, we also examined OLIG2 expression in multiple sclerosis lesions. We demonstrate that OLIG2 displays a differential expression pattern in multiple sclerosis lesions that correlates with lesion activity. Strikingly, OLIG2 was predominantly detected in NOGO-A(+) (now known as RTN4-A) maturing oligodendrocytes, which prevailed in active lesion borders, rather than chronic silent and shadow plaques. Taken together, our data provide proof of principle indicating that OLIG2 overexpression in oligodendrocyte progenitor cells might be a possible therapeutic mechanism for enhancing myelin repair.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Demyelinating Diseases/metabolism , Myelin Sheath/physiology , Nerve Tissue Proteins/metabolism , Oligodendroglia/physiology , Regeneration/genetics , Spinal Cord/cytology , Stem Cells/physiology , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/genetics , Cells, Cultured , Demyelinating Diseases/chemically induced , Demyelinating Diseases/pathology , Disease Models, Animal , Doxycycline/pharmacology , Embryo, Mammalian , Gene Expression Regulation/genetics , Lysophosphatidylcholines/toxicity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/pathology , Nerve Tissue Proteins/genetics , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/pathology , Oligodendroglia/ultrastructure , Regeneration/drug effects , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Spinal Cord/pathology
13.
PLoS One ; 8(2): e57430, 2013.
Article in English | MEDLINE | ID: mdl-23468987

ABSTRACT

Neural precursor (NPC) based therapies are used to restore neurons or oligodendrocytes and/or provide neuroprotection in a large variety of neurological diseases. In multiple sclerosis models, intravenously (i.v) -delivered NPCs reduced clinical signs via immunomodulation. We demonstrated recently that NPCs were able to cross cerebral endothelial cells in vitro and that the multifunctional signalling molecule, CD44 involved in trans-endothelial migration of lymphocytes to sites of inflammation, plays a crucial role in extravasation of syngeneic NPCs. In view of the role of CD44 in NPCs trans-endothelial migration in vitro, we questioned presently the benefit of CD44 overexpression by NPCs in vitro and in vivo, in EAE mice. We show that overexpression of CD44 by NPCs enhanced over 2 folds their trans-endothelial migration in vitro, without impinging on the proliferation or differentiation potential of the transduced cells. Moreover, CD44 overexpression by NPCs improved significantly their elongation, spreading and number of filopodia over the extracellular matrix protein laminin in vitro. We then tested the effect of CD44 overexpression after i.v. delivery in the tail vein of EAE mice. CD44 overexpression was functional invivo as it accelerated trans-endothelial migration and facilitated invasion of HA expressing perivascular sites. These in vitro and in vivo data suggest that CD44 may be crucial not only for NPC crossing the endothelial layer but also for facilitating invasion of extravascular tissues.


Subject(s)
Cell Movement , Endothelium, Vascular/metabolism , Hyaluronan Receptors/metabolism , Neural Stem Cells/metabolism , Animals , Base Sequence , Cell Line , DNA Primers , Endothelium, Vascular/cytology , Flow Cytometry , Immunohistochemistry , Mice , Neural Stem Cells/cytology , Polymerase Chain Reaction
14.
Proc Natl Acad Sci U S A ; 108(26): 10714-9, 2011 Jun 28.
Article in English | MEDLINE | ID: mdl-21670295

ABSTRACT

Boundary cap cells (BC), which express the transcription factor Krox20, participate in the formation of the boundary between the central nervous system and the peripheral nervous system. To study BC stemness, we developed a method to purify and amplify BC in vitro from Krox20(Cre/+), R26R(YFP/+) mouse embryos. We show that BC progeny are EGF/FGF2-responsive, form spheres, and express neural crest markers. Upon growth factor withdrawal, BC progeny gave rise to multiple neural crest and CNS lineages. Transplanted into the developing murine forebrain, they successfully survived, migrated, and integrated within the host environment. Surprisingly, BC progeny generated exclusively CNS cells, including neurons, astrocytes, and myelin-forming oligodendrocytes. In vitro experiments indicated that a sequential combination of ventralizing morphogens and glial growth factors was necessary to reprogram BC into oligodendrocytes. Thus, BC progeny are endowed with differentiation plasticity beyond the peripheral nervous system. The demonstration that CNS developmental cues can reprogram neural crest-derived stem cells into CNS derivatives suggests that BC could serve as a source of cell type-specific lineages, including oligodendrocytes, for cell-based therapies to treat CNS disorders.


Subject(s)
Cell Differentiation , Peripheral Nervous System/cytology , Stem Cells/cytology , Animals , Cell Lineage , Cell Movement , Cells, Cultured , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Oligodendroglia/metabolism
15.
J Cell Biol ; 193(2): 397-407, 2011 Apr 18.
Article in English | MEDLINE | ID: mdl-21502361

ABSTRACT

The specific functions of intrinsic regulators of oligodendrocyte progenitor cell (OPC) division are poorly understood. Type 2 cyclin-dependent kinase (Cdk2) controls cell cycle progression of OPCs, but whether it acts during myelination and repair of demyelinating lesions remains unexplored. Here, we took advantage of a viable Cdk2(-/-) mutant mouse to investigate the function of this cell cycle regulator in OPC proliferation and differentiation in normal and pathological conditions. During central nervous system (CNS) development, Cdk2 loss does not affect OPC cell cycle, oligodendrocyte cell numbers, or myelination. However, in response to CNS demyelination, it clearly alters adult OPC renewal, cell cycle exit, and differentiation. Importantly, Cdk2 loss accelerates CNS remyelination of demyelinated axons. Thus, Cdk2 is dispensable for myelination but is important for adult OPC renewal, and could be one of the underlying mechanisms that drive adult progenitors to differentiate and thus regenerate myelin.


Subject(s)
Central Nervous System/metabolism , Cyclin-Dependent Kinase 2/genetics , Neurogenesis/genetics , Animals , Axons/metabolism , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nerve Regeneration/physiology , Neural Stem Cells/metabolism , Oligodendroglia/metabolism
16.
Brain ; 134(Pt 4): 1168-83, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21459827

ABSTRACT

Improving oligodendroglial differentiation from human foetal neural progenitor cells remains a primordial issue to accomplish successful cell-based therapies in myelin diseases. Here, we combined in situ, in vitro and in vivo approaches to assess the oligodendrogenic potential of different human foetal forebrain regions during the first trimester of gestation. We show for the first time that the initial wave of oligodendrocyte progenitor emergence in the ventral telencephalon onsets as early as 7.5 weeks into gestation. Interestingly, in vitro, isolation of ganglionic eminences yielded oligodendrocyte progenitor-enriched cultures, as compared with cortex and thalamus. Most importantly, single injection of human neural progenitors into rodent models of focal gliotoxic demyelination revealed the great capacity of these cells to survive, extensively migrate and successfully remyelinate the spinal cord, irrespective of their origin. Thus, our study brings novel insights into the biology of early human foetal neural progenitor cells and offers new support for the development of cellular therapeutics for myelin disorders.


Subject(s)
Cell Differentiation/physiology , Myelin Sheath/metabolism , Nerve Regeneration/physiology , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Spinal Cord/metabolism , Analysis of Variance , Animals , Cell Movement/physiology , Humans , Immunohistochemistry , Mice , Mice, Nude , Neural Stem Cells/cytology , Oligodendroglia/cytology , Spinal Cord/cytology
17.
J Neuroimmunol ; 223(1-2): 131-4, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20400187

ABSTRACT

It has been recently shown that systemically injected neural precursor cells (NPCs) could cross brain endothelium and favor functional recovery in animal models of multiple sclerosis (MS). Here we show that human NPCs express receptors of the chemokines IL8 and CXCL13 (CXCR1 and CXCR5, respectively) and migrate across brain endothelial cells in vitro, in response to these chemokines. Considering that these chemokines have been found overexpressed in MS in active, but not inactive areas of demyelination, our data suggest that systemically injected human NPCs may be considered for targeting active areas of demyelination in therapeutic approaches of MS.


Subject(s)
Brain/immunology , Chemokine CXCL13/physiology , Chemotaxis, Leukocyte/immunology , Embryonic Stem Cells/immunology , Endothelial Cells/immunology , Interleukin-8/physiology , Neurons/immunology , Brain/metabolism , Cells, Cultured , Chemokine CXCL13/biosynthesis , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Endothelial Cells/metabolism , Endothelial Cells/transplantation , Humans , Interleukin-8/biosynthesis , Neurons/metabolism , Neurons/transplantation
18.
J Neurosci Res ; 87(15): 3438-46, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19739249

ABSTRACT

Multipotential neural stem/precursor cells of the central nervous system were extensively studied for their properties of generating myelinating oligodendrocytes both in vitro and in vivo upon engraftment in animal models of myelin disorders, such as leucodystrophy and multiple sclerosis. These studies provided proof-of-principle that efficient myelination can be achieved by cell transplantation. However, one major drawback of cell-based therapy of myelin diseases is the difficulty in generating oligodendrocytes efficiently from human fetal neural stem/precursor cells (hNPC). Here we explored whether overexpression of the basic helix-loop-helix (bHLH) transcription factor Olig2 in fetal hNPC could enhance the generation of oligodendrocytes both in vitro and in vivo. We report that transduction of hNPC with Olig2-encoding lentiviral vectors enhances their commitment toward an oligodendroglial fate. Moreover, Olig2-transduced hNPC, grafted into the dysmyelinated shiverer mouse brain, survived up to 9 weeks, migrated extensively, and differentiated into MBP(+) myelinating oligodendrocytes. In contrast, control hNPC remained at a less mature stage and generated very few myelinating oligodendrocytes. Our study indicates that bHLH transcription factors, such as Olig2, are interesting targets for directing hNPC into myelinating oligodendrocytes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain/embryology , Brain/metabolism , Cell Differentiation/genetics , Nerve Tissue Proteins/metabolism , Oligodendroglia/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain/cytology , Brain Tissue Transplantation/methods , Cell Lineage/genetics , Cells, Cultured , Fetus , Gene Expression Regulation, Developmental/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Genetic Vectors/pharmacology , Humans , Lentivirus/genetics , Mice , Mice, Neurologic Mutants , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Nerve Regeneration/genetics , Nerve Tissue Proteins/genetics , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/cytology , Spheroids, Cellular , Stem Cell Transplantation/methods , Stem Cells/cytology , Transduction, Genetic/methods
19.
Stem Cells ; 26(7): 1673-82, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18450824

ABSTRACT

Systemically injected neural precursor cells (NPCs) were unexpectedly shown to reach the cerebral parenchyma and induce recovery in various diffuse brain pathologies, including animal models of multiple sclerosis. However, the molecular mechanisms supporting NPC migration across brain endothelium remain elusive. Brain endothelium constitutes the blood-brain barrier, which uniquely controls the access of drugs and trafficking of cells, including leukocytes, from the blood to the brain. Taking advantage of the availability of in vitro models of human and rat blood-brain barrier developed in our laboratory and validated by us and others, we show here that soluble hyaluronic acid, the major ligand of the adhesion molecule CD44, as well as anti-CD44 blocking antibodies, largely prevents NPC adhesion to and migration across brain endothelium in inflammatory conditions. We present further evidence that NPCs, surprisingly, induce the formation of apical cups at the surface of brain endothelial cells, enriched in CD44 and other adhesion molecules, thus hijacking the endothelial signaling recently shown to be involved in leukocyte extravasation. These results demonstrate the pivotal role of CD44 in the trans-endothelial migration of NPCs across brain endothelial cells: we propose that they may help design new strategies for the delivery of therapeutic NPCs to the brain by systemic administration.


Subject(s)
Brain/metabolism , Endothelial Cells/metabolism , Hyaluronan Receptors/biosynthesis , Hyaluronan Receptors/physiology , Neurons/cytology , Stem Cells/cytology , Animals , Brain/embryology , Cell Adhesion , Cell Movement , Endothelial Cells/cytology , Humans , Hyaluronic Acid/chemistry , Hyaluronic Acid/metabolism , Leukocytes/cytology , Mice , Rats , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...