Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
Add more filters










Publication year range
1.
ASN Neuro ; 16(1): 2371163, 2024.
Article in English | MEDLINE | ID: mdl-39024549

ABSTRACT

The fifteen canonical paracrine fibroblast growth factors (FGFs) are organized in five subfamilies that interact with four FGF-receptors (FGFRs) and heparan sulfate proteoglycan (HSPG) co-receptors. Many of these FGFs are expressed in CNS regions where oligodendrocyte (OL) progenitors originate, migrate or differentiate. FGF2 (basic FGF) is considered a prototype FGF and the information about the effects of FGF signaling on OL-lineage cells has evolved largely from the study of FGF2. However, other FGFs from four subfamilies ((FGF1 (FGF1,-2), FGF4 (FGF4,-5,-6), FGF8 (FGF8,-17,-18) and FGF9 (FGF9,-16,-20)) that can interact with the isoforms of FGFRs expressed in OL-lineage cells may also play important roles. We previously reported OL-responses to FGF8 family members. Here, we investigate the effects of members of the FGF1,-4, and -9 subfamilies on proliferation and differentiation of OL progenitors (OPCs), and on cell cycle re-entry and down-regulation of myelin proteins by mature OLs. We found that while FGF2 induced all these responses strongly, FGF4,-6,-9 could do so only transiently and in the presence of exogenous HSPGs, and that FGF5,-16,-20 could not do so even in the presence of heparin or at higher concentrations. Furthermore, we noted that structurally similar FGFs within subfamilies did not always show similarities in their biological effects on OL-lineage cells. Taken together, these studies reveal that FGFs differ in the way they regulate the OL-lineage cells, emphasizes the selectivity and importance of HSPGs as FGF co-receptors in OL-lineage cells and suggests that structural similarity among FGF-subfamily members may not always predict their overlapping biological functions.


Structurally similar members of the FGF1, -4, and -9 subfamilies trigger diverse biological responses in oligodendrocyte-lineage cells and exhibit selective requirement for heparan sulfate proteoglycans as FGF co-receptors.


Subject(s)
Cell Differentiation , Fibroblast Growth Factors , Oligodendroglia , Animals , Oligodendroglia/metabolism , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Cell Differentiation/physiology , Cell Differentiation/drug effects , Cell Proliferation/physiology , Cell Proliferation/drug effects , Cells, Cultured , Structure-Activity Relationship , Rats
2.
Biomolecules ; 14(4)2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38672460

ABSTRACT

A considerable effort has been spent in the past decades to develop targeted therapies for the treatment of demyelinating diseases, such as multiple sclerosis (MS). Among drugs with free radical scavenging activity and oligodendrocyte protecting effects, Edaravone (Radicava) has recently received increasing attention because of being able to enhance remyelination in experimental in vitro and in vivo disease models. While its beneficial effects are greatly supported by experimental evidence, there is a current paucity of information regarding its mechanism of action and main molecular targets. By using high-throughput RNA-seq and biochemical experiments in murine oligodendrocyte progenitors and SH-SY5Y neuroblastoma cells combined with molecular docking and molecular dynamics simulation, we here provide evidence that Edaravone triggers the activation of aryl hydrocarbon receptor (AHR) signaling by eliciting AHR nuclear translocation and the transcriptional-mediated induction of key cytoprotective gene expression. We also show that an Edaravone-dependent AHR signaling transduction occurs in the zebrafish experimental model, associated with a downstream upregulation of the NRF2 signaling pathway. We finally demonstrate that its rapid cytoprotective and antioxidant actions boost increased expression of the promyelinating Olig2 protein as well as of an Olig2:GFP transgene in vivo. We therefore shed light on a still undescribed potential mechanism of action for this drug, providing further support to its therapeutic potential in the context of debilitating demyelinating conditions.


Subject(s)
Antioxidants , Edaravone , Receptors, Aryl Hydrocarbon , Signal Transduction , Animals , Humans , Mice , Antioxidants/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Edaravone/pharmacology , Molecular Docking Simulation , Molecular Dynamics Simulation , NF-E2-Related Factor 2/metabolism , Receptors, Aryl Hydrocarbon/drug effects , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction/drug effects , Zebrafish/metabolism
3.
Cells ; 12(18)2023 09 11.
Article in English | MEDLINE | ID: mdl-37759473

ABSTRACT

Intracranial hypertension (ICP) and visual impairment intracranial pressure (VIIP) are some of the sequels of long-term space missions. Here we sought to determine how space microgravity (µG) impacts the metabolomics profile of oligodendrocyte progenitors (OLPs), the myelin-forming cells in the central nervous system. We report increased glutamate and energy metabolism while the OLPs were in space for 26 days. We also show that after space flight, OLPs (SPC OLPs) display significantly increased mitochondrial respiration and glycolysis. These data are in agreement with our previous work using simulated microgravity. In addition, our global metabolomics approach allowed for the discovery of endogenous metabolites secreted by OLPs while in space that are significantly modulated by microgravity. Our results provide, for the first time, relevant information about the energetic state of OLPs while in space and after space flight. The functional and molecular relevance of these specific pathways are promising targets for therapeutic intervention for humans in long-term space missions to the moon, Mars and beyond.


Subject(s)
Metabolomics , Secretome , Humans , Oligodendroglia , Myelin Sheath , Glutamic Acid
4.
Biomolecules ; 13(2)2023 01 19.
Article in English | MEDLINE | ID: mdl-36830573

ABSTRACT

Intracranial hypertension (ICP) and visual impairment intracranial pressure (VIIP) are some of the consequences of long-term space missions. Here we examined the behavior of oligodendrocyte progenitors (OLPs) after space flight using time-lapse microscopy. We show that most OLPs divided more than ground control (GC) counterparts did. Nonetheless, a subpopulation of OLPs flown to space presented a significant increase in autophagic cell death. Examination of the proteomic profile of the secretome of space flown OLPs (SPC-OLPs) revealed that the stress protein heat shock protein-90 beta "HSP-90ß" was the 5th most enriched (6.8 times) and the secreted protein acidic and rich in cysteine "SPARC" was the 7th most enriched (5.2 times), with respect to ground control cells. SPARC induces endoplasmic reticulum stress, which leads to autophagy. Given the roles and importance of these two proteins in mammalian cells' metabolism, their upregulation may hold the key to modulating cell proliferation and autophagy, in order to mitigate ICP and VIIP during and after space missions.


Subject(s)
Oligodendrocyte Precursor Cells , Space Flight , Animals , Osteonectin , Proteomics , Autophagy , Cell Proliferation , Mammals
5.
Brain Pathol ; 33(4): e13148, 2023 07.
Article in English | MEDLINE | ID: mdl-36623505

ABSTRACT

The developmental functions of primary cilia and the downstream signaling pathways have been widely studied; however, the roles of primary cilia in the developing neurovascular system are not clearly understood. In this study, we found that ablation of genes encoding ciliary transport proteins such as intraflagellar transport homolog 88 (Ift88) and kinesin family member 3a (Kif3a) in cortical radial progenitors led to periventricular heterotopia during late mouse embryogenesis. Conditional mutation of primary cilia unexpectedly caused breakdown of both the neuroepithelial lining and the blood-choroid plexus barrier. Choroidal leakage was partially caused by enlargement of the choroid plexus in the cilia mutants. We found that the choroid plexus expressed platelet-derived growth factor A (Pdgf-A) and that Pdgf-A expression was ectopically increased in cilia-mutant embryos. Cortices obtained from embryos in utero electroporated with Pdgfa mimicked periventricular heterotopic nodules of the cilia mutant. These results suggest that defective ciliogenesis in both cortical progenitors and the choroid plexus leads to breakdown of cortical and choroidal barriers causing forebrain neuronal dysplasia, which may be related to developmental cortical malformation.


Subject(s)
Cilia , Neurons , Mice , Animals , Cilia/genetics , Cilia/metabolism , Neurons/metabolism , Prosencephalon , Signal Transduction , Carrier Proteins/metabolism
6.
Life (Basel) ; 12(6)2022 May 27.
Article in English | MEDLINE | ID: mdl-35743828

ABSTRACT

In previous studies, we examined the effects of space microgravity on human neural stem cells. To date, there are no studies on a different type of cell that is critical for myelination and electrical signals transmission, oligodendrocyte progenitors (OLPs). The purpose of the present study was to examine the behavior of space-flown OLPs (SPC-OLPs) as they were adapting to Earth's gravity. We found that SPC-OLPs survived, and most of them proliferated normally. Nonetheless, some of them displayed incomplete cytokinesis. Both morphological and ontogenetic analyses showed that they remained healthy and expressed the immature OLP markers Sox2, PDGFR-α, and transferrin (Tf) after space flight, which confirmed that SPC-OLPs displayed a more immature phenotype than their ground control (GC) counterparts. In contrast, GC OLPs expressed markers that usually appear later (GPDH, O4, and ferritin), indicating a delay in SPC-OLPs' development. These cells remained immature even after treatment with culture media designed to support oligodendrocyte (OL) maturation. The most remarkable and surprising finding was that the iron carrier glycoprotein Tf, previously described as an early marker for OLPs, was expressed ectopically in the nucleus of all SPC-OLPs. In contrast, their GC counterparts expressed it exclusively in the cytoplasm, as previously described. In addition, analysis of the secretome demonstrated that SPC-OLPs contained 3.5 times more Tf than that of GC cells, indicating that Tf is gravitationally regulated, opening two main fields of study to understand the upregulation of the Tf gene and secretion of the protein that keep OLPs at a progenitor stage rather than moving forward to more mature phenotypes. Alternatively, because Tf is an autocrine and paracrine factor in the central nervous system (CNS), in the absence of neurons, it accumulated in the secretome collected after space flight. We conclude that microgravity is becoming a novel platform to study why in some myelin disorders OLPs are present but do not mature.

7.
Cells ; 11(3)2022 02 02.
Article in English | MEDLINE | ID: mdl-35159329

ABSTRACT

The oligodendrocyte progenitors (OPCs) are at the front of the glial reaction to the traumatic brain injury. However, regulatory pathways steering the OPC reaction as well as the role of reactive OPCs remain largely unknown. Here, we compared a long-lasting, exacerbated reaction of OPCs to the adult zebrafish brain injury with a timely restricted OPC activation to identify the specific molecular mechanisms regulating OPC reactivity and their contribution to regeneration. We demonstrated that the influx of the cerebrospinal fluid into the brain parenchyma after injury simultaneously activates the toll-like receptor 2 (Tlr2) and the chemokine receptor 3 (Cxcr3) innate immunity pathways, leading to increased OPC proliferation and thereby exacerbated glial reactivity. These pathways were critical for long-lasting OPC accumulation even after the ablation of microglia and infiltrating monocytes. Importantly, interference with the Tlr1/2 and Cxcr3 pathways after injury alleviated reactive gliosis, increased new neuron recruitment, and improved tissue restoration.


Subject(s)
Oligodendrocyte Precursor Cells , Animals , Brain , Gliosis/metabolism , Immunity, Innate , Oligodendrocyte Precursor Cells/metabolism , Zebrafish
8.
Int J Mol Sci ; 22(17)2021 Aug 30.
Article in English | MEDLINE | ID: mdl-34502342

ABSTRACT

Our previous studies have demonstrated that specific peroxisome proliferator-activated receptor-γ (PPAR-γ) agonists play a fundamental role in oligodendrocyte progenitor (OP) differentiation, protecting them against oxidative and inflammatory damage. The antihypertensive drug Telmisartan (TLM) was shown to act as a PPAR-γ modulator. This study investigates the TLM effect on OP differentiation and validates its capability to restore damage in a pharmacological model of Niemann-Pick type C (NPC) disease through a PPAR-γ-mediated mechanism. For the first time in purified OPs, we demonstrate that TLM-induced PPAR-γ activation downregulates the type 1 angiotensin II receptor (AT1), the level of which naturally decreases during differentiation. Like other PPAR-γ agonists, we show that TLM promotes peroxisomal proliferation and promotes OP differentiation. Furthermore, TLM can offset the OP maturation arrest induced by a lysosomal cholesterol transport inhibitor (U18666A), which reproduces an NPC1-like phenotype. In the NPC1 model, TLM also reduces cholesterol accumulation within peroxisomal and lysosomal compartments and the contacts between lysosomes and peroxisomes, revealing that TLM can regulate intracellular cholesterol transport, crucial for myelin formation. Altogether, these data indicate a new potential use of TLM in hypomyelination pathologies such as NPC1, underlining the possible repositioning of the drug already used in other pathologies.


Subject(s)
Antihypertensive Agents/pharmacology , Cell Differentiation , Cholesterol/metabolism , Oligodendroglia/drug effects , PPAR gamma/metabolism , Protective Agents/pharmacology , Telmisartan/pharmacology , Animals , Oligodendroglia/metabolism , PPAR gamma/genetics , Rats , Rats, Wistar
9.
Semin Cell Dev Biol ; 116: 16-24, 2021 08.
Article in English | MEDLINE | ID: mdl-34110985

ABSTRACT

Oligodendrocytes are highly specialized glial cells, responsible for producing myelin in the central nervous system (CNS). The multi-stage process of oligodendrocyte development is tightly regulated to ensure proper lineage progression of oligodendrocyte progenitor cells (OPCs) to mature myelin producing oligodendrocytes. This developmental process involves complex interactions between several intrinsic signaling pathways that are modulated by an array of extrinsic factors. Understanding these regulatory processes is of crucial importance, as it may help to identify specific molecular targets both to enhance plasticity in the normal CNS and to promote endogenous recovery following injury or disease. This review describes two major regulators that play important functional roles in distinct phases of oligodendrocyte development: OPC proliferation and differentiation. Specifically, we highlight the roles of the extracellular astrocyte/radial glia-derived protein Endothelin-1 in OPC proliferation and the intracellular Akt/mTOR pathway in OPC differentiation. Lastly, we reflect on how recent advances in neuroscience and scientific technology will enable greater understanding into how intrinsic and extrinsic regulators interact to generate oligodendrocyte diversity.


Subject(s)
Oligodendroglia/metabolism , Stem Cells/metabolism , Cell Differentiation , Cell Proliferation , Humans
10.
Stem Cell Reports ; 14(6): 1076-1092, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32413277

ABSTRACT

The subventricular zone (SVZ) of the lateral ventricles and the subgranular zone (SGZ) of the dentate gyrus in the hippocampus are known as neurogenic niches. We show that the median eminence (ME) of the hypothalamus comprises BrdU+ newly proliferating cells co-expressing NG2 (oligodendrocyte progenitors) and RIP (pre-myelinating oligodendrocytes), suggesting their differentiation toward mature oligodendrocytes (OLs). ME cells can generate neurospheres (NS) in vitro, which differentiate mostly to OLs compared with SVZ-NS that typically generate neurons. Interestingly, this population of oligodendrocyte progenitors is increased in the ME from experimental autoimmune encephalomyelitis (EAE)-affected mice. Notably, the thrombospondin 1 (TSP1) expressed by astrocytes, acts as negative regulator of oligodendrogenesis in vitro and is downregulated in the ME of EAE mice. Importantly, transplanted ME-NS preferentially differentiate to MBP+ OLs compared with SVZ-NS in Shiverer mice. Hence, discovering the ME as a new site for myelin-producing cells has a great importance for advising future therapy for demyelinating diseases and spinal cord injury.


Subject(s)
Median Eminence/cytology , Neural Stem Cells/cytology , Oligodendroglia/cytology , Stem Cell Niche , Animals , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neurogenesis , Oligodendroglia/metabolism , Thrombospondin 1/genetics , Thrombospondin 1/metabolism
11.
Glia ; 68(10): 1957-1967, 2020 10.
Article in English | MEDLINE | ID: mdl-32086854

ABSTRACT

Remyelination, namely, the formation of new myelin sheaths around denuded axons, counteracts axonal degeneration and restores neuronal function. Considerable advances have been made in understanding this regenerative process that often fails in diseases like multiple sclerosis, leaving axons demyelinated and vulnerable to damage, thus contributing to disease progression. The identification of the membrane receptor GPR17 on a subset of oligodendrocyte precursor cells (OPCs), which mediate remyelination in the adult central nervous system (CNS), has led to a huge amount of evidence that validated this receptor as a new attractive target for remyelinating therapies. Here, we summarize the role of GPR17 in OPC function, myelination and remyelination, describing its atypical pharmacology, its downstream signaling, and the genetic and epigenetic factors modulating its activity. We also highlight crucial insights into GPR17 pathophysiology coming from the demonstration that oligodendrocyte injury, associated with inflammation in chronic neurodegenerative conditions, is invariably characterized by abnormal and persistent GPR17 upregulation, which, in turn, is accompanied by a block of OPCs at immature premyelinating stages. Finally, we discuss the current literature in light of the potential exploitment of GPR17 as a therapeutic target to promote remyelination.


Subject(s)
Myelin Sheath/metabolism , Oligodendroglia/metabolism , Receptors, G-Protein-Coupled/metabolism , Remyelination/physiology , Signal Transduction/physiology , Animals , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Epigenesis, Genetic/physiology , Humans , Myelin Sheath/genetics , Receptors, G-Protein-Coupled/genetics
13.
Neurochem Res ; 45(3): 591-605, 2020 Mar.
Article in English | MEDLINE | ID: mdl-30906970

ABSTRACT

Oligodendrocytes (OL) are the only myelinating cells of the central nervous system thus interferences, either environmental or genetic, with their maturation or function have devastating consequences. Albeit so far neglected, one of the less appreciated, nevertheless possible, regulators of OL maturation and function is the circadian cycle. Yet, disruptions in these rhythms are unfortunately becoming a common "disorder" in the today's world. The temporal patterning of behaviour and physiology is controlled by a circadian timing system based in the anterior hypothalamus. At the molecular level, circadian rhythms are generated by a transcriptional/translational feedback system that regulates transcription and has a major impact on cellular function(s). Fundamental cellular properties/functions in most cell types vary with the daily circadian cycle: OL are unlikely an exception! To be clear, the presence of circadian oscillators or the cell-specific function(s) of the circadian clock in OL has yet to be defined. Furthermore, we wish to entertain the idea of links between the "thin" evidence on OL intrinsic circadian rhythms and their interjection(s) at different stages of lineage progression as well as in supporting/regulating OL crucial function: myelination. Individuals with intellectual and developmental syndromes as well as neurodegenerative diseases present with a disrupted sleep/wake cycle; hence, we raise the possibility that these disturbances in timing can contribute to the loss of white matter observed in these disorders. Preclinical and clinical work in this area is needed for a better understanding of how circadian rhythms influence OL maturation and function(s), to aid the development of new therapeutic strategies and standards of care for these patients.


Subject(s)
Circadian Rhythm , Oligodendroglia/metabolism , Sleep/physiology , Animals , Humans
14.
Front Cell Dev Biol ; 7: 303, 2019.
Article in English | MEDLINE | ID: mdl-31850342

ABSTRACT

The limited access to functional human brain tissue has led to the development of stem cell-based alternative models. The differentiation of human pluripotent stem cells into cerebral organoids with self-organized architecture has created novel opportunities to study the early stages of the human cerebral formation. Here we applied state-of-the-art label-free shotgun proteomics to compare the proteome of stem cell-derived cerebral organoids to the human fetal brain. We identified 3,073 proteins associated with different developmental stages, from neural progenitors to neurons, astrocytes, or oligodendrocytes. The major protein groups are associated with neurogenesis, axon guidance, synaptogenesis, and cortical brain development. Glial cell proteins related to cell growth and maintenance, energy metabolism, cell communication, and signaling were also described. Our data support the variety of cells and neural network functional pathways observed within cell-derived cerebral organoids, confirming their usefulness as an alternative model. The characterization of brain organoid proteome is key to explore, in a dish, atypical and disrupted processes during brain development or neurodevelopmental, neurodegenerative, and neuropsychiatric diseases.

15.
Iran J Pharm Res ; 18(1): 286-295, 2019.
Article in English | MEDLINE | ID: mdl-31089363

ABSTRACT

The generation of oligodendrocyte progenitor cells (OPCs) offers tremendous opportunities for cell replacement therapy in demyelinating diseases such as multiple sclerosis (MS) and spinal cord injury. Recently, the prospect of reprogramming terminally differentiated adult cells towards another mature somatic cell or progenitor cells without an intermediate pluripotent state has been of interest. Trichostatin A is a histone deacetylase inhibitor which opens the chromatin and facilitates the transcription of silence genes. In this study, we have treated human astrocytes line U87 and primary culture of mouse astrocytes with TSA for 12 h, prior their transfer to OPC induction medium. Then we evaluated the morphology and the fate of the treated astrocytes at post-treatment days. Both cell lines acquired OPC morphology and expressed OPC specific markers. Following transfer to differentiation medium, U87-derived iOPCs differentiated to oligodendrocyte like cells and expressed PLP as a mature oligodendrocyte marker. Our results introduced TSA as an inducer for production of OPCs from astrocytes and could be considered a potential way for the treatment of demyelinating diseases.

16.
Wiley Interdiscip Rev Dev Biol ; 8(4): e342, 2019 07.
Article in English | MEDLINE | ID: mdl-30945456

ABSTRACT

Brain tumors such as adult glioblastomas and pediatric high-grade gliomas or medulloblastomas are among the leading causes of cancer-related deaths, exhibiting poor prognoses with little improvement in outcomes in the past several decades. These tumors are heterogeneous and can be initiated from various neural cell types, contributing to therapy resistance. How such heterogeneity arises is linked to the tumor cell of origin and their genetic alterations. Brain tumorigenesis and progression recapitulate key features associated with normal neurogenesis; however, the underlying mechanisms are quite dysregulated as tumor cells grow and divide in an uncontrolled manner. Recent comprehensive genomic, transcriptomic, and epigenomic studies at single-cell resolution have shed new light onto diverse tumor-driving events, cellular heterogeneity, and cells of origin in different brain tumors. Primary and secondary glioblastomas develop through different genetic alterations and pathways, such as EGFR amplification and IDH1/2 or TP53 mutation, respectively. Mutations such as histone H3K27M impacting epigenetic modifications define a distinct group of pediatric high-grade gliomas such as diffuse intrinsic pontine glioma. The identification of distinct genetic, epigenomic profiles and cellular heterogeneity has led to new classifications of adult and pediatric brain tumor subtypes, affording insights into molecular and lineage-specific vulnerabilities for treatment stratification. This review discusses our current understanding of tumor cells of origin, heterogeneity, recurring genetic and epigenetic alterations, oncogenic drivers and signaling pathways for adult glioblastomas, pediatric high-grade gliomas, and medulloblastomas, the genetically heterogeneous groups of malignant brain tumors. This article is categorized under: Gene Expression and Transcriptional Hierarchies > Gene Networks and Genomics Adult Stem Cells, Tissue Renewal, and Regeneration > Stem Cell Differentiation and Reversion Signaling Pathways > Cell Fate Signaling.


Subject(s)
Brain Neoplasms/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Epigenesis, Genetic/genetics , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Humans , Medulloblastoma/genetics , Medulloblastoma/metabolism , Medulloblastoma/pathology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Oligodendrocyte Precursor Cells/cytology , Oligodendrocyte Precursor Cells/metabolism
17.
Neural Regen Res ; 13(4): 628-632, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29722306

ABSTRACT

Chondroitin sulfate proteoglycan-4 (CSPG4) is a surface component of two key cell types (oligodendrocyte progenitor cells (OPCs) and myeloid cells) present in lysolecithin-induced lesions in mouse spinal cord. Two types of CSPG4 manipulations have been used to study the roles of these cells in myelin damage and repair: (1) OPC and myeloid-specific ablation of CSPG4, and (2) transplantation of enhanced green fluorescent protein (EGFP)-labeled progenitors to distinguish between bone marrow-derived macrophages and resident microglia. Ablation of CSPG4 in OPCs does not affect myelin damage, but decreases myelin repair, due to reduced proliferation of CSPG4-null OPCs that diminishes generation of mature oligodendrocytes for remyelination. Ablation of CSPG4 in myeloid cells greatly decreases recruitment of macrophages to spinal cord lesions, resulting in smaller initial lesions, but also in significantly diminished myelin repair. In the absence of macrophage recruitment, OPC proliferation is greatly impaired, again leading to decreased generation of myelinating oligodendrocytes. Macrophages may promote OPC proliferation via phagocytosis of myelin debris and/or secretion of factors that stimulate OPC mitosis. Microglia are not able to substitute for macrophages in promoting OPC proliferation. An additional feature of lesions in myeloid-specific CSPG4 null mice is the persistence of poorly-differentiated platelet-derived growth factor receptor α (PDGFRα) + macrophages that may prolong damage.

18.
Mol Neurobiol ; 55(5): 4388-4402, 2018 May.
Article in English | MEDLINE | ID: mdl-28660484

ABSTRACT

Perinatal asphyxia results from the action of different risk factors like complications during pregnancy, preterm delivery, or long and difficult labor. Nowadays, it is still the leading cause of neonatal brain injury known as hypoxic-ischemic encephalopathy (HIE) and resulting neurological disorders. A temporal limitation of oxygen, glucose, and trophic factors supply results in alteration of neural cell differentiation and functioning and/or leads to their death. Among the affected cells are oligodendrocytes, responsible for myelinating the central nervous system (CNS) and formation of white matter. Therefore, one of the major consequences of the experienced HIE is leukodystrophic diseases resulting from oligodendrocyte deficiency or malfunctioning. The therapeutic strategies applied after perinatal asphyxia are aimed at reducing brain damage and promoting the endogenous neuroreparative mechanisms. In this review, we focus on the biology of oligodendrocytes and discuss present clinical treatments in the context of their efficiency in preserving white matter structure and preventing cognitive and behavioral deficits after perinatal asphyxia.


Subject(s)
Asphyxia/complications , Leukoencephalopathies/etiology , Leukoencephalopathies/therapy , Myelin Sheath/pathology , Oligodendroglia/pathology , Animals , Cell Transplantation , Humans , Nerve Regeneration
19.
Acta Neuropathol ; 134(3): 441-458, 2017 09.
Article in English | MEDLINE | ID: mdl-28685323

ABSTRACT

Whereas microglia involvement in virtually all brain diseases is well accepted their role in the control of homeostasis in the central nervous system (CNS) is mainly thought to be the maintenance of neuronal function through the formation, refinement, and monitoring of synapses in both the developing and adult brain. Although the prenatal origin as well as the neuron-centered function of cortical microglia has recently been elucidated, much less is known about a distinct amoeboid microglia population formerly described as the "fountain of microglia" that appears only postnatally in myelinated regions such as corpus callosum and cerebellum. Using large-scale transcriptional profiling, fate mapping, and genetic targeting approaches, we identified a unique molecular signature of this microglia subset that arose from a CNS endogenous microglia pool independent from circulating myeloid cells. Microglia depletion experiments revealed an essential role of postnatal microglia for the proper development and homeostasis of oligodendrocytes and their progenitors. Our data provide new cellular and molecular insights into the myelin-supporting function of microglia in the normal CNS.


Subject(s)
Microglia/physiology , Myelin Sheath/physiology , Oligodendrocyte Precursor Cells/physiology , Oligodendroglia/physiology , Animals , Cell Proliferation/physiology , Mice
20.
J Tissue Eng Regen Med ; 11(5): 1442-1455, 2017 05.
Article in English | MEDLINE | ID: mdl-26118416

ABSTRACT

Oligodendrocyte progenitors (OPCs) are ranked among the most likely candidates for cell-based strategies aimed at treating neurodegenerative diseases accompanied by dys/demyelination of the central nervous system (CNS). In this regard, different sources of stem cells are being tested to elaborate xeno-free protocols for efficient generation of OPCs for clinical applications. In the present study, neural stem cells of human umbilical cord blood (HUCB-NSCs) have been used to derive OPCs and subsequently to differentiate them into mature, GalC-expressing oligodendrocytes. Applied components of the extracellular matrix (ECM) and the analogues of physiological substances known to increase glial commitment of neural stem cells have been shown to significantly increase the yield of the resulting OPC fraction. The efficiency of ECM components in promoting oligodendrocyte commitment and differentiation prompted us to investigate the potential role of gelatinases in those processes. Subsequently, endogenous and ECM metalloproteinases (MMPs) activity has been compared with that detected in primary cultures of rat oligodendrocytes in vitro, as well as in rat brains in vivo. The data indicate that gelatinases are engaged in gliogenesis both in vitro and in vivo, although differently, which presumably results from distinct extracellular conditions. In conclusion, the study presents an efficient xeno-free method of deriving oligodendrocyte from HUCB-NSCs and analyses the engagement of MMP-2/MMP-9 in the processes of cell commitment and maturation. Copyright © 2015 John Wiley & Sons, Ltd.


Subject(s)
Cell Differentiation , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Umbilical Cord/metabolism , Animals , Cell Line , Cell Separation , Humans , Neural Stem Cells/cytology , Oligodendroglia/cytology , Rats , Rats, Wistar , Umbilical Cord/cytology
SELECTION OF CITATIONS
SEARCH DETAIL