Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cells ; 13(6)2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38534318

RESUMEN

Parkinson's disease (PD) is a progressive neurodegenerative disease characterized by resting tremor, bradykinesia, rigidity, and postural instability that also includes non-motor symptoms such as mood dysregulation. Dopamine (DA) is the primary neurotransmitter involved in this disease, but cholinergic imbalance has also been implicated. Current intervention in PD is focused on replenishing central DA, which provides remarkable temporary symptomatic relief but does not address neuronal loss and the progression of the disease. It has been well established that neuronal nicotinic cholinergic receptors (nAChRs) can regulate DA release and that nicotine itself may have neuroprotective effects. Recent studies identified nAChRs in nonneuronal cell types, including glial cells, where they may regulate inflammatory responses. Given the crucial role of neuroinflammation in dopaminergic degeneration and the involvement of microglia and astrocytes in this response, glial nAChRs may provide a novel therapeutic target in the prevention and/or treatment of PD. In this review, following a brief discussion of PD, we focus on the role of glial cells and, specifically, their nAChRs in PD pathology and/or treatment.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedad de Parkinson , Receptores Nicotínicos , Humanos , Enfermedad de Parkinson/metabolismo , Receptores Nicotínicos/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Nicotina/metabolismo , Dopamina/metabolismo , Astrocitos/metabolismo
2.
Cell Mol Life Sci ; 80(9): 260, 2023 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-37594553

RESUMEN

Oligodendrocytes are generated via a two-step mechanism from pluripotent neural stem cells (NSCs): after differentiation of NSCs to oligodendrocyte precursor/NG2 cells (OPCs), they further develop into mature oligodendrocytes. The first step of this differentiation process is only incompletely understood. In this study, we utilized the neurosphere assay to investigate NSC to OPC differentiation in a time course-dependent manner by mass spectrometry-based (phospho-) proteomics. We identify doublecortin-like kinase 1 (Dclk1) as one of the most prominently regulated proteins in both datasets, and show that it undergoes a gradual transition between its short/long isoform during NSC to OPC differentiation. This is regulated by phosphorylation of its SP-rich region, resulting in inhibition of proteolytic Dclk1 long cleavage, and therefore Dclk1 short generation. Through interactome analyses of different Dclk1 isoforms by proximity biotinylation, we characterize their individual putative interaction partners and substrates. All data are available via ProteomeXchange with identifier PXD040652.


Asunto(s)
Células-Madre Neurales , Células Precursoras de Oligodendrocitos , Diferenciación Celular , Quinasas Similares a Doblecortina , Oligodendroglía , Fosforilación , Proteínas Serina-Treonina Quinasas , Proteómica
3.
Glia ; 71(9): 2210-2233, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37226895

RESUMEN

Oligodendrocyte precursor cells (OPCs) generate oligodendrocytes, a process that may be tuned by neuronal activity, possibly via synaptic connections to OPCs. However, a developmental role of synaptic signaling to OPCs has so far not been shown unequivocally. To address this question, we comparatively analyzed functional and molecular characteristics of highly proliferative and migratory OPCs in the embryonic brain. Embryonic OPCs in mice (E18.5) shared the expression of voltage-gated ion channels and their dendritic morphology with postnatal OPCs, but almost completely lacked functional synaptic currents. Transcriptomic profiling of PDGFRα+ OPCs revealed a limited abundance of genes coding for postsynaptic signaling and synaptogenic cell adhesion molecules in the embryonic versus the postnatal period. RNA sequencing of single OPCs showed that embryonic synapse-lacking OPCs are found in clusters distinct from postnatal OPCs and with similarities to early progenitors. Furthermore, single-cell transcriptomics demonstrated that synaptic genes are transiently expressed only by postnatal OPCs until they start to differentiate. Taken together, our results indicate that embryonic OPCs represent a unique developmental stage biologically resembling postnatal OPCs but without synaptic input and a transcriptional signature in the continuum between OPCs and neural precursors.


Asunto(s)
Células Precursoras de Oligodendrocitos , Ratones , Animales , Células Precursoras de Oligodendrocitos/metabolismo , Ratones Transgénicos , Oligodendroglía/metabolismo , Neuronas/fisiología , Neurogénesis/fisiología , Diferenciación Celular/fisiología
4.
J Chem Neuroanat ; 127: 102192, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36403746

RESUMEN

Brain injury triggers a complex response involving morphological changes, cellular proliferation, and differentiation of newly formed neuroglial subpopulations. These processes have been extensively studied in animal stroke models with permanent large vessel occlusion. However, less is known about neuroglial response after transient cerebral ischemia. Herein, we aimed to determine an astrocytic and NG2 glial proliferative response, potential changes in expression of developmental neuroglial markers: vimentin, nestin, oligodendrocyte transcription marker (Olig2), and a role of neuroglial subpopulations as a source of cells replenishing structural deficiencies in the ischemic brain. Results showed an induction of a proliferative neuroglial response in the peri-infarct area reflected in an increased percentage of GFAP/Ki67 + and NG2/Ki67 + cells within 4 weeks after transient MCAO. The peak of GFAP+ astrocytes proliferation of 30.3 ± 10.3% was observed in the first week, and a peak of NG2 + cells proliferation of 23.1 ± 11.8% in the second week after stroke. The presence of GFAP/Vimentin+ and GFAP/Nestin+ cells, as well as GFAP/Olig2 + and NG2/Olig2 + cells indicated an induction of developmental phenotypes with a differentiation potential. Finally, observed between day 1 and week 3 transient GFAP/NG2 + colocalization suggests the heterogeneous source of the reactive neuroglia after transient MCAO. Altogether, one-hour MCAO is a sufficient pathological stimulus to trigger a strong proliferative response of GFAP+ and NG2 + neuroglial cells and induce their early developmental phenotype. Our results suggest that transient ischemia may initiate a change in the direction of differentiation within the neuroglia cell population.


Asunto(s)
Ataque Isquémico Transitorio , Accidente Cerebrovascular , Animales , Ataque Isquémico Transitorio/patología , Nestina/metabolismo , Vimentina/metabolismo , Antígeno Ki-67/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Neuroglía/metabolismo , Astrocitos/metabolismo , Diferenciación Celular/fisiología , Accidente Cerebrovascular/metabolismo , Proliferación Celular
5.
Neurosci Res ; 188: 28-38, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36375656

RESUMEN

The aqueduct of Sylvius connects the third with the fourth ventricle and is surrounded by the Periaqueductal Grey. Here, we report a novel niche of cells in the dorsal section of the aqueduct, hereby named dorsal aqueduct niche or DAN, by applying a battery of selective markers and transgenic mouse lines. The somata of DAN cells are located toward the lumen of the ventricle forming multiple layers in close association with the cerebrospinal fluid (CSF). A single process emerges from the soma and run with the blood vessels. Cells of the DAN express radial glia/stem cell markers such as GFAP, vimentin and nestin, and the glutamate transporter GLAST or the oligodendrocyte precursor/pericyte marker NG2, thereby suggesting their potential for the generation of new cells. Morphologically, DAN cells resemble tanycytes of the third ventricle, which transfer biochemical signals from the CSF to the central nervous system and display proliferative capacity. The aqueduct ependymal lining can proliferate as observed by the integration of BrdU and expression of Ki67. Thus, the dorsal section of the aqueduct of Sylvius possesses cells that may act a niche of new glial cells in the adult mouse brain.


Asunto(s)
Acueducto del Mesencéfalo , Tercer Ventrículo , Animales , Ratones , Acueducto del Mesencéfalo/metabolismo , Proteína Ácida Fibrilar de la Glía/metabolismo , Tercer Ventrículo/metabolismo , Neuroglía/metabolismo , Epéndimo/metabolismo , Ratones Transgénicos
6.
Front Cell Neurosci ; 16: 1005399, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36467604

RESUMEN

Spinal cord injury (SCI) is a serious neurological trauma that is challenging to treat. After SCI, many neurons in the injured area die due to necrosis or apoptosis, and astrocytes, oligodendrocytes, microglia and other non-neuronal cells become dysfunctional, hindering the repair of the injured spinal cord. Corrective surgery and biological, physical and pharmacological therapies are commonly used treatment modalities for SCI; however, no current therapeutic strategies can achieve complete recovery. Somatic cell reprogramming is a promising technology that has gradually become a feasible therapeutic approach for repairing the injured spinal cord. This revolutionary technology can reprogram fibroblasts, astrocytes, NG2 cells and neural progenitor cells into neurons or oligodendrocytes for spinal cord repair. In this review, we provide an overview of the transcription factors, genes, microRNAs (miRNAs), small molecules and combinations of these factors that can mediate somatic cell reprogramming to repair the injured spinal cord. Although many challenges and questions related to this technique remain, we believe that the beneficial effect of somatic cell reprogramming provides new ideas for achieving functional recovery after SCI and a direction for the development of treatments for SCI.

7.
Exp Neurol ; 355: 114147, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35738417

RESUMEN

Following injury in the central nervous system, a population of astrocytes occupy the lesion site, form glial bridges and facilitate axon regeneration. These astrocytes originate primarily from resident astrocytes or NG2+ oligodendrocyte progenitor cells. However, the extent to which these cell types give rise to the lesion-filling astrocytes, and whether the astrocytes derived from different cell types contribute similarly to optic nerve regeneration remain unclear. Here we examine the distribution of astrocytes and NG2+ cells in an optic nerve crush model. We show that optic nerve astrocytes partially fill the injury site over time after a crush injury. Viral mediated expression of a growth-promoting factor, ciliary neurotrophic factor (CNTF), in retinal ganglion cells (RGCs) promotes axon regeneration without altering the lesion size or the degree of lesion-filling GFAP+ cells. Strikingly, using inducible NG2CreER driver mice, we found that CNTF overexpression in RGCs increases the occupancy of NG2+ cell-derived astrocytes in the optic nerve lesion. An EdU pulse-chase experiment shows that the increase in NG2 cell-derived astrocytes is not due to an increase in cell proliferation. Lastly, we performed RNA-sequencing on the injured optic nerve and reveal that CNTF overexpression in RGCs results in significant changes in the expression of distinct genes, including those that encode chemokines, growth factor receptors, and immune cell modulators. Even though CNTF-induced axon regeneration has long been recognized, this is the first evidence of this procedure affecting glial cell fate at the optic nerve crush site. We discuss possible implication of these results for axon regeneration.


Asunto(s)
Traumatismos del Nervio Óptico , Traumatismos del Sistema Nervioso , Animales , Astrocitos/metabolismo , Axones/patología , Factor Neurotrófico Ciliar , Citocinas/metabolismo , Ratones , Regeneración Nerviosa/fisiología , Traumatismos del Nervio Óptico/patología , Células Ganglionares de la Retina/metabolismo , Traumatismos del Sistema Nervioso/metabolismo
8.
Folia Morphol (Warsz) ; 80(4): 766-775, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34699052

RESUMEN

The dynamic development of studies on neuroglia in recent years indicates its previously underestimated role in maintaining proper brain function, both in physiological and pathological conditions. The use of modern research methods such as single-cell techniques as well as in vivo and in vitro models enriched the state of our knowledge. The most important issues regarding the maturation and development of neuroglia include cooperation between glial cell groups and with neurons in neurogenesis, neuroregeneration, (re)myelination and how the early developmental roles of glia contribute to nervous system dysfunction in neurodevelopmental and neurodegenerative disorders. There is still growing evidence emphasizing the importance of astroglia in maintaining the brain physiological homeostasis, regulation of immune response, cerebral blood flow, and involvement in the reactive neurogliosis, precisely adapted to the nature of pathological stimulus and the depth of tissue damage. The important issues related to the function of oligodendrocytes include explanation of the mechanisms of interaction between the glial cells and myelinated axons, important not only in myelination, but also in development of cognitive processes and memory. Further studies are required for understanding the mechanisms of demyelination occurring in several central nervous system (CNS) diseases. An interesting area of research is related with explanation of the NG2 glia function, characterised by significant proliferative potential and ability to differentiate in both in physiological conditions and in pathology, as well as the presence of synaptic neural-glial connections, which are especially numerous during development. The increasing knowledge of microglia comprises the presence of specialised subsets of microglia, their role the myelination process and neurovascular unit functioning. We are only beginning to understand how microglia enter the brain and develop distinct functional states during ontogeny. This review summarises the current state of knowledge on the development and role in the CNS of different, heterogeneous cell populations defined by a common term neuroglia.


Asunto(s)
Neurogénesis , Neuroglía , Astrocitos , Neuronas , Oligodendroglía
9.
Cells ; 10(8)2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34440814

RESUMEN

Adult neural stem and progenitor cells (NSPCs) contribute to learning, memory, maintenance of homeostasis, energy metabolism and many other essential processes. They are highly heterogeneous populations that require input from a regionally distinct microenvironment including a mix of neurons, oligodendrocytes, astrocytes, ependymal cells, NG2+ glia, vasculature, cerebrospinal fluid (CSF), and others. The diversity of NSPCs is present in all three major parts of the CNS, i.e., the brain, spinal cord, and retina. Intrinsic and extrinsic signals, e.g., neurotrophic and growth factors, master transcription factors, and mechanical properties of the extracellular matrix (ECM), collectively regulate activities and characteristics of NSPCs: quiescence/survival, proliferation, migration, differentiation, and integration. This review discusses the heterogeneous NSPC populations in the normal physiology and highlights their potentials and roles in injured/diseased states for regenerative medicine.


Asunto(s)
Células Madre Adultas/fisiología , Células-Madre Neurales/fisiología , Enfermedades Neurodegenerativas/patología , Traumatismos de la Médula Espinal/patología , Células Madre Adultas/citología , Células Madre Adultas/trasplante , Animales , Antígenos/metabolismo , Diferenciación Celular , Epéndimo/citología , Epéndimo/fisiología , Humanos , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante , Enfermedades Neurodegenerativas/terapia , Proteoglicanos/metabolismo , Medicina Regenerativa , Traumatismos de la Médula Espinal/terapia
10.
Cell Mol Neurobiol ; 41(2): 309-326, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32335774

RESUMEN

Following the transection of peripheral sympathetic preganglionic axons comprising the cervical sympathetic trunk (CST), we observe robust glial and neuronal plasticity at 1 week post-injury in the rat spinal cord intermediolateral cell column (IML), which houses the injured parent neuronal cell bodies. This plasticity contributes to neuroprotection, as no neuronal loss in the IML is present at 16 weeks post-injury. Here, we administered the antibiotic minocycline or vehicle (VEH) daily for 1 week after CST transection to investigate the role of activated microglia in IML glial and neuronal plasticity and subsequent neuronal survival. At 1 week post-injury, minocycline treatment did not alter microglia number in the IML, but led to a dampened microglia activation state. In addition, the increases in oligodendrocyte (OL) lineage cells and activated astrocytes following injury in VEH rats were attenuated in the minocycline-treated rats. Further, the normal downregulation of choline acetyltransferase (ChAT) in the injured neurons was blunted. At 16 weeks post-injury, fewer ChAT+ neurons were present in the minocycline-treated rats, suggesting that activated microglia together with the glial and neuronal plasticity at 1 week post-injury contribute to the long-term survival of the injured neurons. These results provide evidence for beneficial crosstalk between activated microglia and neurons as well as other glial cells in the cord following peripheral axon injury, which ultimately leads to neuroprotection. The influences of microglia activation in promoting neuronal survival should be considered when developing therapies to administer minocycline for the treatment of neurological pathologies.


Asunto(s)
Axones/patología , Microglía/patología , Plasticidad Neuronal , Médula Espinal/patología , Factor de Transcripción Activador 3/metabolismo , Animales , Astrocitos/efectos de los fármacos , Axones/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Colina O-Acetiltransferasa/metabolismo , Femenino , Microglía/efectos de los fármacos , Microglía/metabolismo , Minociclina/farmacología , Plasticidad Neuronal/efectos de los fármacos , Oligodendroglía/efectos de los fármacos , Oligodendroglía/patología , Ratas Sprague-Dawley , Factores de Tiempo
11.
Prog Retin Eye Res ; 81: 100886, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32771538

RESUMEN

Glial cells are critically important for maintenance of neuronal activity in the central nervous system (CNS), including the optic nerve (ON). However, the ON has several unique characteristics, such as an extremely high myelination level of retinal ganglion cell (RGC) axons throughout the length of the nerve (with virtually all fibers myelinated by 7 months of age in humans), lack of synapses and very narrow geometry. Moreover, the optic nerve head (ONH) - a region where the RGC axons exit the eye - represents an interesting area that is morphologically distinct in different species. In many cases of multiple sclerosis (demyelinating disease of the CNS) vision problems are the first manifestation of the disease, suggesting that RGCs and/or glia in the ON are more sensitive to pathological conditions than cells in other parts of the CNS. Here, we summarize current knowledge on glial organization and function in the ON, focusing on glial support of RGCs. We cover both well-established concepts on the important role of glial cells in ON health and new findings, including novel insights into mechanisms of remyelination, microglia/NG2 cell-cell interaction, astrocyte reactivity and the regulation of reactive astrogliosis by mitochondrial fragmentation in microglia.


Asunto(s)
Neuroglía/fisiología , Nervio Óptico/citología , Animales , Axones/fisiología , Humanos , Células Ganglionares de la Retina/fisiología
12.
Adv Exp Med Biol ; 1175: 181-197, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31583589

RESUMEN

Ageing reduces the functional capacity of all organs, so does that of the nervous system; the latter is evident in the reduction of cognitive abilities, learning and memory. While the exact mechanisms of ageing of the nervous system remain elusive, it is without doubt that morpho-functional changes in a variety of neuroglial cells contribute to this process. The age-dependent changes in neuroglia are characterised by a progressive loss of function. This reduces glial ability to homeostatically nurture, protect and regenerate the nervous tissue. Such neuroglial paralysis also facilitates neurodegenerative processes. Ageing of neuroglia is variable and can be affected by environmental factors and comorbidities.


Asunto(s)
Envejecimiento , Neuroglía/fisiología , Humanos
13.
Glia ; 67(11): 2178-2202, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31444938

RESUMEN

Spinal cord injury (SCI) affects over 17,000 individuals in the United States per year, resulting in sudden motor, sensory and autonomic impairments below the level of injury. These deficits may be due at least in part to the loss of oligodendrocytes and demyelination of spared axons as it leads to slowed or blocked conduction through the lesion site. It has long been accepted that progenitor cells form new oligodendrocytes after SCI, resulting in the acute formation of new myelin on demyelinated axons. However, the chronicity of demyelination and the functional significance of remyelination remain contentious. Here we review work examining demyelination and remyelination after SCI as well as the current understanding of oligodendrocyte lineage cell responses to spinal trauma, including the surprisingly long-lasting response of NG2+ oligodendrocyte progenitor cells (OPCs) to proliferate and differentiate into new myelinating oligodendrocytes for months after SCI. OPCs are highly sensitive to microenvironmental changes, and therefore respond to the ever-changing post-SCI milieu, including influx of blood, monocytes and neutrophils; activation of microglia and macrophages; changes in cytokines, chemokines and growth factors such as ciliary neurotrophic factor and fibroblast growth factor-2; glutamate excitotoxicity; and axon degeneration and sprouting. We discuss how these changes relate to spontaneous oligodendrogenesis and remyelination, the evidence for and against demyelination being an important clinical problem and if remyelination contributes to motor recovery.


Asunto(s)
Enfermedades Desmielinizantes/patología , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Traumatismos de la Médula Espinal/patología , Animales , Humanos , Remielinización/fisiología , Traumatismos de la Médula Espinal/fisiopatología , Células Madre/fisiología
14.
J Neurosci ; 39(16): 3013-3027, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30760627

RESUMEN

In the CNS, oligodendrocytes are responsible for myelin formation and maintenance. Following spinal cord injury, oligodendrocyte loss and an inhibitory milieu compromise remyelination and recovery. Here, we explored the role of retinoic acid receptor-beta (RARß) signaling in remyelination. Using a male Sprague Dawley rat model of PNS-CNS injury, we show that oral treatment with a novel drug like RARß agonist, C286, induces neuronal expression of the proteoglycan decorin and promotes myelination and differentiation of oligodendrocyte precursor cells (NG2+ cells) in a decorin-mediated neuron-glia cross talk. Decorin promoted the activation of RARα in NG2+ cells by increasing the availability of the endogenous ligand RA. NG2+ cells synthesize RA, which is released in association with exosomes. We found that decorin prevents this secretion through regulation of the EGFR-calcium pathway. Using functional and pharmacological studies, we further show that RARα signaling is both required and sufficient for oligodendrocyte differentiation. These findings illustrate that RARß and RARα are important regulators of oligodendrocyte differentiation, providing new targets for myelination.SIGNIFICANCE STATEMENT This study identifies novel therapeutic targets for remyelination after PNS-CNS injury. Pharmacological and knock-down experiments show that the retinoic acid (RA) signaling promotes differentiation of oligodendrocyte precursor cells (OPCs) and remyelination in a cross talk between neuronal RA receptor-beta (RARß) and RARα in NG2+ cells. We show that stimulation of RARα is required for the differentiation of OPCs and we describe for the first time how oral treatment with a RARß agonist (C286, currently being tested in a Phase 1 trial, ISRCTN12424734) leads to the endogenous synthesis of RA through retinaldehyde dehydrogenase 2 (Raldh2) in NG2 cells and controls exosome-associated-RA intracellular levels through a decorin-Ca2+ pathway. Although RARß has been implicated in distinct aspects of CNS regeneration, this study identifies a novel function for both RARß and RARα in remyelination.


Asunto(s)
Exosomas/metabolismo , Vaina de Mielina/metabolismo , Regeneración Nerviosa/efectos de los fármacos , Receptores de Ácido Retinoico/agonistas , Traumatismos de la Médula Espinal/tratamiento farmacológico , Tretinoina/metabolismo , Animales , Decorina/metabolismo , Receptores ErbB/metabolismo , Vaina de Mielina/efectos de los fármacos , Regeneración Nerviosa/fisiología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Traumatismos de la Médula Espinal/metabolismo
15.
Eur Psychiatry ; 50: 60-69, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29503098

RESUMEN

Growing evidence points to synaptic pathology as a core component of the pathophysiology of schizophrenia (SZ). Significant reductions of dendritic spine density and altered expression of their structural and molecular components have been reported in several brain regions, suggesting a deficit of synaptic plasticity. Regulation of synaptic plasticity is a complex process, one that requires not only interactions between pre- and post-synaptic terminals, but also glial cells and the extracellular matrix (ECM). Together, these elements are referred to as the 'tetrapartite synapse', an emerging concept supported by accumulating evidence for a role of glial cells and the extracellular matrix in regulating structural and functional aspects of synaptic plasticity. In particular, chondroitin sulfate proteoglycans (CSPGs), one of the main components of the ECM, have been shown to be synthesized predominantly by glial cells, to form organized perisynaptic aggregates known as perineuronal nets (PNNs), and to modulate synaptic signaling and plasticity during postnatal development and adulthood. Notably, recent findings from our group and others have shown marked CSPG abnormalities in several brain regions of people with SZ. These abnormalities were found to affect specialized ECM structures, including PNNs, as well as glial cells expressing the corresponding CSPGs. The purpose of this review is to bring forth the hypothesis that synaptic pathology in SZ arises from a disruption of the interactions between elements of the tetrapartite synapse.


Asunto(s)
Encéfalo/patología , Plasticidad Neuronal/fisiología , Esquizofrenia/patología , Sinapsis/patología , Animales , Encéfalo/metabolismo , Encéfalo/fisiopatología , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Matriz Extracelular/metabolismo , Humanos , Esquizofrenia/metabolismo , Esquizofrenia/fisiopatología , Sinapsis/metabolismo
16.
Glia ; 66(5): 1068-1081, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29393544

RESUMEN

NG2 cells represent precursors of oligodendrocytes under physiological conditions; however, following cerebral ischemia they play an important role in glial scar formation. Here, we compared the expression profiles of oligodendroglial lineage cells, after focal cerebral ischemia (FCI) and in Alzheimer's-like pathology using transgenic mice, which enables genetic fate-mapping of Cspg4-positive NG2 cells and their progeny, based on the expression of red fluorescent protein tdTomato. tdTomato-positive cells possessed the expression profile of NG2 cells and oligodendrocytes; however, based on the expression of cell type-specific genes, we were able to distinguish between them. To shed light on the changes in the expression patterns caused by FCI, we employed self-organizing Kohonen maps, enabling the division of NG2 cells and oligodendrocytes into subpopulations based on similarities in the expression profiles of individual cells. We identified three subpopulations of NG2 cells emerging after FCI: proliferative; astrocyte-like and oligodendrocyte-like NG2 cells; such phenotypes were further confirmed by immunohistochemistry. Oligodendrocytes themselves formed four subpopulations, which reflected the process of oligodendrocytes maturation. Finally, we used 5-ethynyl-2' deoxyuridine (EdU) labeling to reveal that NG2 cells can differentiate directly into reactive astrocytes without preceding proliferation. In contrast, in Alzheimer's-like pathology we failed to identify these subpopulations. Collectively, here we identified several yet unknown differences between the expression profiles of NG2 cells and oligodendrocytes, and characterized specific genes contributing to oligodendrocyte maturation and phenotypical changes of NG2 cells after FCI. Moreover, our results suggest that, unlike in Alzheimer's-like pathology, NG2 cells acquire a multipotent phenotype following FCI.


Asunto(s)
Isquemia Encefálica/fisiopatología , Regeneración Nerviosa/fisiología , Células Precursoras de Oligodendrocitos/fisiología , Animales , Astrocitos/patología , Astrocitos/fisiología , Encéfalo/patología , Encéfalo/fisiopatología , Isquemia Encefálica/patología , Proliferación Celular/fisiología , Modelos Animales de Enfermedad , Femenino , Ratones Transgénicos , Células Precursoras de Oligodendrocitos/patología , Análisis de la Célula Individual
17.
Neuron ; 96(5): 1003-1012.e7, 2017 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-29103804

RESUMEN

Blood-brain barrier (BBB) disruption alters the composition of the brain microenvironment by allowing blood proteins into the CNS. However, whether blood-derived molecules serve as extrinsic inhibitors of remyelination is unknown. Here we show that the coagulation factor fibrinogen activates the bone morphogenetic protein (BMP) signaling pathway in oligodendrocyte progenitor cells (OPCs) and suppresses remyelination. Fibrinogen induces phosphorylation of Smad 1/5/8 and inhibits OPC differentiation into myelinating oligodendrocytes (OLs) while promoting an astrocytic fate in vitro. Fibrinogen effects are rescued by BMP type I receptor inhibition using dorsomorphin homolog 1 (DMH1) or CRISPR/Cas9 activin A receptor type I (ACVR1) knockout in OPCs. Fibrinogen and the BMP target Id2 are increased in demyelinated multiple sclerosis (MS) lesions. Therapeutic depletion of fibrinogen decreases BMP signaling and enhances remyelination in vivo. Targeting fibrinogen may be an upstream therapeutic strategy to promote the regenerative potential of CNS progenitors in diseases with remyelination failure.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Fibrinógeno/farmacología , Células Precursoras de Oligodendrocitos/metabolismo , Remielinización/efectos de los fármacos , Receptores de Activinas Tipo I/efectos de los fármacos , Receptores de Activinas Tipo I/genética , Receptores de Activinas Tipo I/metabolismo , Animales , Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/patología , Fibrinógeno/antagonistas & inhibidores , Lisofosfatidilcolinas/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis por Micromatrices , Vaina de Mielina/metabolismo , Células Precursoras de Oligodendrocitos/efectos de los fármacos , Plásmidos/genética , Transducción de Señal/efectos de los fármacos
18.
J Neurosci ; 37(43): 10290-10309, 2017 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-28931573

RESUMEN

Oligodendrocyte precursor cells (OPCs) act as a reservoir of new oligodendrocytes (OLs) in homeostatic and pathological conditions. OPCs are activated in response to injury to generate myelinating OLs, but the underlying mechanisms remain poorly understood. Here, we show that chromodomain helicase DNA binding protein 7 (Chd7) regulates OPC activation after spinal cord injury (SCI). Chd7 is expressed in OPCs in the adult spinal cord and its expression is upregulated with a concomitant increase in Sox2 expression after SCI. OPC-specific ablation of Chd7 in injured mice leads to reduced OPC proliferation, the loss of OPC identity, and impaired OPC differentiation. Ablation of Chd7 or Sox2 in cultured OPCs shows similar phenotypes to those observed in Chd7 knock-out mice. Chd7 and Sox2 form a complex in OPCs and bind to the promoters or enhancers of the regulator of cell cycle (Rgcc) and protein kinase Cθ (PKCθ) genes, thereby inducing their expression. The expression of Rgcc and PKCθ is reduced in the OPCs of the injured Chd7 knock-out mice. In cultured OPCs, overexpression and knock-down of Rgcc or PKCθ promote and suppress OPC proliferation, respectively. Furthermore, overexpression of both Rgcc and PKCθ rescues the Chd7 deletion phenotypes. Chd7 is thus a key regulator of OPC activation, in which it cooperates with Sox2 and acts via direct induction of Rgcc and PKCθ expression.SIGNIFICANCE STATEMENT Spinal cord injury (SCI) leads to oligodendrocyte (OL) loss and demyelination, along with neuronal death, resulting in impairment of motor or sensory functions. Oligodendrocyte precursor cells (OPCs) activated in response to injury are potential sources of OL replacement and are thought to contribute to remyelination and functional recovery after SCI. However, the molecular mechanisms underlying OPC activation, especially its epigenetic regulation, remain largely unclear. We demonstrate here that the chromatin remodeler chromodomain helicase DNA binding protein 7 (Chd7) regulates the proliferation and identity of OPCs after SCI. We have further identified regulator of cell cycle (Rgcc) and protein kinase Cθ (PKCθ) as novel targets of Chd7 for OPC activation.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Oligodendroglía/metabolismo , Factores de Transcripción SOXB1/fisiología , Traumatismos de la Médula Espinal/metabolismo , Células Madre/metabolismo , Animales , Proliferación Celular/fisiología , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Noqueados , Ratones Transgénicos , Oligodendroglía/patología , Embarazo , Unión Proteica/fisiología , Traumatismos de la Médula Espinal/patología , Células Madre/patología
19.
Biochem Pharmacol ; 141: 42-55, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28522408

RESUMEN

NG2 cells represent one of the most proliferative glial cell populations in the intact mammalian central nervous system (CNS). They are well-known for their ability to renew themselves or to generate new oligodendrocytes during development as well as in adulthood, therefore also being termed oligodendrocyte progenitor cells. Following CNS injuries, such as demyelination, trauma or ischemia, the proliferative capacity of NG2 cells rapidly increases and moreover, their differentiation potential broadens, as documented by numerous reports also describing their differentiation into astrocytes or even neurons. Here, we summarize the current knowledge about NG2 cells proliferation, their fate plasticity during embryogenesis as well as in postnatal CNS under physiological and pathological conditions, with the main emphasis on the role of various signaling molecules, growth factors, hormones or even neurotransmitters on the fate potential of NG2 cells.


Asunto(s)
Células Madre Multipotentes/fisiología , Neurogénesis/fisiología , Neuroglía/fisiología , Células Madre/fisiología , Animales , Antígenos/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Fármacos del Sistema Nervioso Central/farmacología , Fármacos del Sistema Nervioso Central/uso terapéutico , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Madre Multipotentes/efectos de los fármacos , Células Madre Multipotentes/trasplante , Neurogénesis/efectos de los fármacos , Neuroglía/efectos de los fármacos , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/fisiología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/fisiología , Proteoglicanos/metabolismo , Células Madre/efectos de los fármacos
20.
BMC Neurosci ; 18(1): 31, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28279169

RESUMEN

BACKGROUND: Increased motor activity or social interactions through enriched environment are strong stimulators of grey and white matter plasticity in the adult rodent brain. In the present study we evaluated whether specific reaching training of the dominant forelimb (RT) and stimulation of unspecific motor activity through enriched environment (EE) influence the generation of distinct oligodendrocyte subpopulations in the sensorimotor cortex and corpus callosum of the adult rat brain. Animals were placed in three different housing conditions: one group was transferred to an EE, a second group received daily RT, whereas a third group remained in the standard cage. Bromodeoxyuridine (BrdU) was applied at days 2-6 after start of experiments and animals were allowed to survive for 10 and 42 days. RESULTS: Enriched environment and daily reaching training of the dominant forelimb significantly increased the number of newly differentiated GSTπ+ oligodendrocytes at day 10 and newly differentiated CNPase+ oligodendrocytes in the sensorimotor cortex at day 42. The myelin level as measured by CNPase expression was increased in the frontal cortex at day 42. Distribution of newly differentiated NG2+ subpopulations changed between 10 and 42 days with an increase of GSTπ+ subtypes and a decrease of NG2+ cells in the sensorimotor cortex and corpus callosum. Analysis of neuronal marker doublecortin (DCX) showed that more than half of NG2+ cells express DCX in the cortex. The number of new DCX+NG2+ cells was reduced by EE at day 10. CONCLUSIONS: Our results indicate for the first time that specific and unspecific motor training conditions differentially alter the process of differentiation from oligodendrocyte subpopulations, in particular NG2+DCX+ cells, in the sensorimotor cortex and corpus callosum.


Asunto(s)
Cuerpo Calloso/fisiología , Vivienda para Animales , Destreza Motora/fisiología , Oligodendroglía/fisiología , Práctica Psicológica , Corteza Sensoriomotora/fisiología , 2',3'-Nucleótido Cíclico Fosfodiesterasas/metabolismo , Animales , Antígenos/metabolismo , Bromodesoxiuridina , Cuerpo Calloso/citología , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Miembro Anterior/fisiología , Lóbulo Frontal/citología , Lóbulo Frontal/fisiología , Masculino , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Animales , Neurogénesis/fisiología , Neuropéptidos/metabolismo , Oligodendroglía/citología , Proteoglicanos/metabolismo , Distribución Aleatoria , Ratas Wistar , Tiempo de Reacción , Corteza Sensoriomotora/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA