Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Glia ; 71(8): 1791-1803, 2023 08.
Article in English | MEDLINE | ID: mdl-36866453

ABSTRACT

Zika virus (ZIKV) is a strongly neurotropic flavivirus whose infection has been associated with microcephaly in neonates. However, clinical and experimental evidence indicate that ZIKV also affects the adult nervous system. In this regard, in vitro and in vivo studies have shown the ability of ZIKV to infect glial cells. In the central nervous system (CNS), glial cells are represented by astrocytes, microglia, and oligodendrocytes. In contrast, the peripheral nervous system (PNS) constitutes a highly heterogeneous group of cells (Schwann cells, satellite glial cells, and enteric glial cells) spread through the body. These cells are critical in both physiological and pathological conditions; as such, ZIKV-induced glial dysfunctions can be associated with the development and progression of neurological complications, including those related to the adult and aging brain. This review will address the effects of ZIKV infection on CNS and PNS glial cells, focusing on cellular and molecular mechanisms, including changes in the inflammatory response, oxidative stress, mitochondrial dysfunction, Ca2+ and glutamate homeostasis, neural metabolism, and neuron-glia communication. Of note, preventive and therapeutic strategies that focus on glial cells may emerge to delay and/or prevent the development of ZIKV-induced neurodegeneration and its consequences.


Subject(s)
Zika Virus Infection , Zika Virus , Humans , Zika Virus/physiology , Zika Virus Infection/complications , Zika Virus Infection/drug therapy , Zika Virus Infection/pathology , Neuroglia/metabolism , Central Nervous System/metabolism , Brain/metabolism
2.
Front Immunol ; 13: 956340, 2022.
Article in English | MEDLINE | ID: mdl-36072579

ABSTRACT

Clostridioides difficile (C. difficile) produces toxins A (TcdA) and B (TcdB), both associated with intestinal damage and diarrhea. Pannexin-1 (Panx1) channels allows the passage of messenger molecules, such as adenosine triphosphate (ATP), which in turn activate the P2X7 receptors (P2X7R) that regulate inflammation and cell death in inflammatory bowel diseases. The aim of this study was to verify the effect of C. difficile infection (CDI) in the expression of Panx1 and P2X7R in intestinal tissues of mice, as well as their role in cell death and IL-6 expression induced by TcdA and TcdB in enteric glial cells (EGCs). Male C57BL/6 mice (8 weeks of age) were infected with C. difficile VPI10463, and the control group received only vehicle per gavage. After three days post-infection (p.i.), cecum and colon samples were collected to evaluate the expression of Panx1 by immunohistochemistry. In vitro, EGCs (PK060399egfr) were challenged with TcdA or TcdB, in the presence or absence of the Panx1 inhibitor (10Panx trifluoroacetate) or P2X7R antagonist (A438079), and Panx1 and P2X7R expression, caspase-3/7 activity and phosphatidylserine binding to annexin-V, as well as IL-6 expression were assessed. CDI increased the levels of Panx1 in cecum and colon of mice compared to the control group. Panx1 inhibitor decreased caspase-3/7 activity and phosphatidylserine-annexin-V binding, but not IL-6 gene expression in TcdA and TcdB-challenged EGCs. P2X7 receptor antagonist accentually reduced caspase-3/7 activity, phosphatidylserine-annexin-V binding, and IL-6 gene expression in TcdA and TcdB-challenged EGCs. In conclusion, Panx1 is increased during CDI and plays an important role in the effects of C. difficile toxins in EGCs, participating in cell death induced by both toxins by promoting caspase-3/7 activation via P2X7R, which is also involved in IL-6 expression induced by both toxins.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Connexins , Nerve Tissue Proteins , Receptors, Purinergic P2X7 , Animals , Annexins , Bacterial Toxins/metabolism , Caspase 3/metabolism , Cell Death , Connexins/genetics , Connexins/metabolism , Inflammation Mediators , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroglia/metabolism , Phosphatidylserines , Receptors, Purinergic P2X7/genetics
3.
Dev Biol ; 442(2): 249-261, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30071216

ABSTRACT

The development of the sensory nervous system is the result of fine-tuned waves of neurogenesis and apoptosis which control the appropriate number of precursors and newly generated neurons and orient them toward a specific lineage. Neurotrophins and their tyrosine-kinase receptors (RTK) orchestrate this process. They have long been in the scope of the neurotrophic theory which established that a neuron is committed to die unless a trophic factor generated by its target provides it with a survival signal. The neural death has thus always been described as a "default" program, survival being the major player to control the number of cells. New insights have been brought by the gain of function studies which recently demonstrated that TrkC (NTRK3) is a "dependence receptor" able to actively trigger apoptosis in absence of its ligand NT-3. In order to address the role of TrkC pro-apoptotic activity in the control of sensory neurons number, we generated a TrkC gene-trap mutant mice. We found out that this new murine model recapitulates the sensory phenotype of TrkC constitutive mutants, with reduced DRG size and reduced number of DRG neurons. We engineered these mice strain with a lacZ reporter in order to follow the fate of neurons committed to a TrkC lineage and observed that they are specifically protected from NT-3 mediated apoptosis in NT-3/TrkC double knock-out embryos. Finally, using a chicken model we demonstrated that silencing NT-3 emanating from the ventral neural tube induced apoptosis in the DRG anlage. This apoptosis was inhibited by silencing TrkC. This work thus demonstrates that, during in vivo DRG development, TrkC behaves as a two-sided receptor transducing positive signals of neuronal survival in response to NT-3, but actively inducing neuronal cell death when unbound. This functional duality sets adequate number of neurons committed to a TrkC identity in the forming DRG.


Subject(s)
Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Receptor, trkC/metabolism , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism , Animals , Apoptosis/physiology , Cell Line , Cell Survival/physiology , Chick Embryo , Female , Ganglia, Spinal/embryology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism
4.
Front Cell Neurosci ; 12: 235, 2018.
Article in English | MEDLINE | ID: mdl-30123112

ABSTRACT

In recent years, the functions of glial cells, namely, astrocytes and microglia, have gained prominence in several diseases of the central nervous system, especially in glioblastoma (GB), the most malignant primary brain tumor that leads to poor clinical outcomes. Studies showed that microglial cells or astrocytes play a critical role in promoting GB growth. Based on the recent findings, the complex network of the interaction between microglial/astrocytes cells and GB may constitute a potential therapeutic target to overcome tumor malignancy. In the present review, we summarize the most important mechanisms and functions of the molecular factors involved in the microglia or astrocytes-GB interactions, which is particularly the alterations that occur in the cell's extracellular matrix and the cytoskeleton. We overview the cytokines, chemokines, neurotrophic, morphogenic, metabolic factors, and non-coding RNAs actions crucial to these interactions. We have also discussed the most recent studies regarding the mechanisms of transportation and communication between microglial/astrocytes - GB cells, namely through the ABC transporters or by extracellular vesicles. Lastly, we highlight the therapeutic challenges and improvements regarding the crosstalk between these glial cells and GB.

5.
Dev Biol ; 444 Suppl 1: S47-S59, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29614271

ABSTRACT

In the neural primordium of vertebrate embryos, the neural crest (NC) displays a unique character: the capacity of its component cells to leave the neural primordium, migrate along definite (and, for long, not identified) routes in the developing embryo and invade virtually all tissues and organs, while producing a large array of differentiated cell types. The most striking diversity of the NC derivatives is found in its cephalic domain that produces, not only melanocytes and peripheral nerves and ganglia, but also various mesenchymal derivatives (connective tissues, bones, cartilages…) which, in other parts of the body, are mesoderm-derived. The aim of this article was to review the large amount of work that has been devoted to solving the problem of the differentiation capacities of individual NC cells (NCC) arising from both the cephalic and trunk levels of the neural axis. A variety of experimental designs applied to NCC either in vivo or in vitro are evaluated, including the possibility to culture them in crestospheres, a technique previously designed for cells of the CNS, and which reinforces the notion, previously put forward, of the existence of NC stem cells. At the trunk level, the developmental potentialities of the NCC are more restricted than in their cephalic counterparts, but, in addition to the neural-melanocytic fate that they exclusively express in vivo, it was clearly shown that they harbor mesenchymal capacities that can be revealed in vitro. Finally, a large amount of evidence has been obtained that, during the migration process, most of the NCC are multipotent with a variable array of potentialities among the cells considered. Investigations carried out in adults have shown that multipotent NC stem cells persist in the various sites of the body occupied by NCC. Enlightening new developments concerning the invasive capacity of NCC, the growing peripheral nerves were revealed as migration routes for NCC travelling to distant ventrolateral regions of the body. Designated "Schwann cell precursors" in the mouse embryo, these NCC can leave the nerves and are able to convert to a novel fate. The convertibility of the NC-derived cells, particularly evident in the Schwann cell-melanocyte lineage transition, has also been demonstrated for neuroendocrine cells of the adult carotid body and for the differentiation of parasympathetic neurons of ganglia distant from their origin, the NC. All these new developments attest the vitality of the research on the NC, a field that characterizes vertebrate development and for which the interest has constantly increased during the last decades.


Subject(s)
Multipotent Stem Cells/cytology , Neural Crest/cytology , Neural Crest/physiology , Animals , Biological Evolution , Body Patterning , Cell Differentiation/physiology , Cell Movement/physiology , Central Nervous System/physiology , Embryonic Development , Epithelial-Mesenchymal Transition/physiology , Humans , Melanocytes/cytology , Mesoderm , Neural Crest/metabolism , Neural Plate/physiology , Neural Stem Cells/cytology , Neurogenesis/physiology , Neuronal Plasticity/physiology , Neurons/cytology , Schwann Cells , Vertebrates
6.
J Neuroinflammation ; 14(1): 79, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28388962

ABSTRACT

BACKGROUND: The irinotecan (CPT-11) causes intestinal mucositis and diarrhea that may be related to changes in the enteric nervous system (ENS). In inflammatory condition, mast cells release a variety of pro-inflammatory mediators that can interact with the ENS cells. It has not been explored whether CPT-11 is able to alter the enteric glial and neuronal cell, and the role of mast cells in this effect. Therefore, this study was conducted to investigate the effect of CPT-11 on the enteric glial and neuronal cells, as well as to study the role of mast cells in the CPT-11-induced intestinal mucositis. METHODS: Intestinal mucositis was induced in Swiss mice by the injection of CPT-11 (60 mg/kg, i.p.) once a day for 4 days following by euthanasia on the fifth day. To investigate the role of mast cells, the mice were pretreated with compound 48/80 for 4 days (first day, 0.6 mg/kg; second day, 1.0 mg/kg; third day, 1.2 mg/kg; fourth day, 2.4 mg/kg) to induce mast cell degranulation before the CPT-11 treatment. RESULTS: Here, we show that CPT-11 increased glial fibrillary acidic protein (GFAP) and S100ß gene and S100ß protein expressions and decreased HuC/D protein expression in the small intestine segments. Concomitantly, CPT-11 enhanced tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) levels and inducible nitric oxide synthase (iNOS) gene expression, associated with an increase in the total number macrophages (positive cells for ionized calcium-binding adapter molecule, Iba-1) and degranulated mast cells in the small intestine segments and caused significant weight loss. The pretreatment with compound 48/80, an inductor of mast cells degranulation, significantly prevented these CPT-11-induced effects. CONCLUSIONS: Our data suggests the participation of mast cells on the CPT-11-induced intestinal mucositis, macrophages activation, enteric reactive gliosis, and neuron loss.


Subject(s)
Camptothecin/analogs & derivatives , Enteric Nervous System/metabolism , Gliosis/chemically induced , Gliosis/metabolism , Mast Cells/metabolism , Neurons/metabolism , Animals , Antineoplastic Agents, Phytogenic/toxicity , Camptothecin/toxicity , Cell Count , Enteric Nervous System/drug effects , Enteric Nervous System/pathology , Gliosis/pathology , Intestine, Small/drug effects , Intestine, Small/metabolism , Intestine, Small/pathology , Irinotecan , Mast Cells/drug effects , Mast Cells/pathology , Mice , Neurons/drug effects , Neurons/pathology , Random Allocation
7.
PLoS One ; 10(8): e0133689, 2015.
Article in English | MEDLINE | ID: mdl-26241738

ABSTRACT

Connective-tissue growth factor (CTGF) is a modular secreted protein implicated in multiple cellular events such as chondrogenesis, skeletogenesis, angiogenesis and wound healing. CTGF contains four different structural modules. This modular organization is characteristic of members of the CCN family. The acronym was derived from the first three members discovered, cysteine-rich 61 (CYR61), CTGF and nephroblastoma overexpressed (NOV). CTGF is implicated as a mediator of important cell processes such as adhesion, migration, proliferation and differentiation. Extensive data have shown that CTGF interacts particularly with the TGFß, WNT and MAPK signaling pathways. The capacity of CTGF to interact with different growth factors lends it an important role during early and late development, especially in the anterior region of the embryo. ctgf knockout mice have several cranio-facial defects, and the skeletal system is also greatly affected due to an impairment of the vascular-system development during chondrogenesis. This study, for the first time, indicated that CTGF is a potent inductor of gliogenesis during development. Our results showed that in vitro addition of recombinant CTGF protein to an embryonic mouse neural precursor cell culture increased the number of GFAP- and GFAP/Nestin-positive cells. Surprisingly, CTGF also increased the number of Sox2-positive cells. Moreover, this induction seemed not to involve cell proliferation. In addition, exogenous CTGF activated p44/42 but not p38 or JNK MAPK signaling, and increased the expression and deposition of the fibronectin extracellular matrix protein. Finally, CTGF was also able to induce GFAP as well as Nestin expression in a human malignant glioma stem cell line, suggesting a possible role in the differentiation process of gliomas. These results implicate ctgf as a key gene for astrogenesis during development, and suggest that its mechanism may involve activation of p44/42 MAPK signaling. Additionally, CTGF-induced differentiation of glioblastoma stem cells into a less-tumorigenic state could increase the chances of successful intervention, since differentiated cells are more vulnerable to cancer treatments.


Subject(s)
Astrocytes/drug effects , Connective Tissue Growth Factor/pharmacology , Fibronectins/biosynthesis , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line, Tumor , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Fibronectins/genetics , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Glioblastoma/pathology , Humans , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nestin/analysis , Nestin/biosynthesis , Nestin/genetics , Phosphorylation/drug effects , Protein Processing, Post-Translational/drug effects , Recombinant Proteins/pharmacology , SOXB1 Transcription Factors/analysis , Xenopus Proteins/pharmacology
8.
Dev Biol ; 384(1): 13-25, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24099925

ABSTRACT

The neural crest (NC), an ectoderm-derived structure of the vertebrate embryo, gives rise to the melanocytes, most of the peripheral nervous system and the craniofacial mesenchymal tissues (i.e., connective, bone, cartilage and fat cells). In the trunk of Amniotes, no mesenchymal tissues are derived from the NC. In certain in vitro conditions however, avian and murine trunk NC cells (TNCCs) displayed a limited mesenchymal differentiation capacity. Whether this capacity originates from committed precursors or from multipotent TNCCs was unknown. Here, we further investigated the potential of TNCCs to develop into mesenchymal cell types in vitro. We found that, in fact, quail TNCCs exhibit a high ability to differentiate into myofibroblasts, chondrocytes, lipid-laden adipocytes and mineralizing osteoblasts. In single cell cultures, both mesenchymal and neural cell types coexisted in TNCC clonal progeny: 78% of single cells yielded osteoblasts together with glial cells and neurons; moreover, TNCCs generated heterogenous clones with adipocytes, myofibroblasts, melanocytes and/or glial cells. Therefore, alike cephalic NCCs, early migratory TNCCs comprised multipotent progenitors able to generate both mesenchymal and melanocytic/neural derivatives, suggesting a continuum in NC developmental potentials along the neural axis. The skeletogenic capacity of the TNC, which was present in the exoskeletal armor of the extinct basal forms of Vertebrates and which persisted in the distal fin rays of extant teleost fish, thus did not totally disappear during vertebrate evolution. Mesenchymal potentials of the TNC, although not fulfilled during development, are still present in a dormant state in Amniotes and can be disclosed in in vitro culture. Whether these potentials are not expressed in vivo due to the presence of inhibitory cues or to the lack of permissive factors in the trunk environment remains to be understood.


Subject(s)
Multipotent Stem Cells/cytology , Neural Crest/cytology , Quail/metabolism , 3T3 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , Cell Differentiation , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Feeder Cells/cytology , Gene-Environment Interaction , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Multipotent Stem Cells/metabolism , Neural Crest/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Quail/embryology
9.
Mol Cell ; 51(5): 632-46, 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24034695

ABSTRACT

The neurotrophin receptor TrkC was recently identified as a dependence receptor, and, as such, it triggers apoptosis in the absence of its ligand, NT-3. The molecular mechanism for apoptotic engagement involves the double cleavage of the receptor's intracellular domain, leading to the formation of a proapoptotic "killer" fragment (TrkC KF). Here, we show that TrkC KF interacts with Cobra1, a putative cofactor of BRCA1, and that Cobra1 is required for TrkC-induced apoptosis. We also show that, in the developing chick neural tube, NT-3 silencing is associated with neuroepithelial cell death that is rescued by Cobra1 silencing. Cobra1 shuttles TrkC KF to the mitochondria, where it promotes Bax activation, cytochrome c release, and apoptosome-dependent apoptosis. Thus, we propose that, in the absence of NT-3, the proteolytic cleavage of TrkC leads to the release of a killer fragment that triggers mitochondria-dependent apoptosis via the recruitment of Cobra1.


Subject(s)
Apoptosis/physiology , Mitochondria/metabolism , Nuclear Proteins/metabolism , Receptor, trkC/metabolism , Animals , Chick Embryo/metabolism , Cytochromes c/metabolism , Cytosol/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Gene Silencing , Humans , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Neurons/metabolism , Neurotrophin 3/metabolism , Neurotrophin 3/pharmacology , Nuclear Proteins/genetics , Peptide Fragments/metabolism , RNA-Binding Proteins , Receptor, trkC/genetics , bcl-2-Associated X Protein/metabolism
10.
PLoS One ; 8(1): e55605, 2013.
Article in English | MEDLINE | ID: mdl-23383241

ABSTRACT

Connective-tissue growth factor (CTGF/CCN2) is a matricellular-secreted protein involved in complex processes such as wound healing, angiogenesis, fibrosis and metastasis, in the regulation of cell proliferation, migration and extracellular matrix remodeling. Glioblastoma (GBM) is the major malignant primary brain tumor and its adaptation to the central nervous system microenvironment requires the production and remodeling of the extracellular matrix. Previously, we published an in vitro approach to test if neurons can influence the expression of the GBM extracellular matrix. We demonstrated that neurons remodeled glioma cell laminin. The present study shows that neurons are also able to modulate CTGF expression in GBM. CTGF immnoreactivity and mRNA levels in GBM cells are dramatically decreased when these cells are co-cultured with neonatal neurons. As proof of particular neuron effects, neonatal neurons co-cultured onto GBM cells also inhibit the reporter luciferase activity under control of the CTGF promoter, suggesting inhibition at the transcription level. This inhibition seems to be contact-mediated, since conditioned media from embryonic or neonatal neurons do not affect CTGF expression in GBM cells. Furthermore, the inhibition of CTGF expression in GBM/neuronal co-cultures seems to affect the two main signaling pathways related to CTGF. We observed inhibition of TGFß luciferase reporter assay; however phopho-SMAD2 levels did not change in these co-cultures. In addition levels of phospho-p44/42 MAPK were decreased in co-cultured GBM cells. Finally, in transwell migration assay, CTGF siRNA transfected GBM cells or GBM cells co-cultured with neurons showed a decrease in the migration rate compared to controls. Previous data regarding laminin and these results demonstrating that CTGF is down-regulated in GBM cells co-cultured with neonatal neurons points out an interesting view in the understanding of the tumor and cerebral microenvironment interactions and could open up new strategies as well as suggest a new target in GBM control.


Subject(s)
Cell Communication , Connective Tissue Growth Factor/metabolism , Glioblastoma/metabolism , Neurons/metabolism , Animals , Cell Line, Tumor , Cell Movement , Coculture Techniques , Connective Tissue Growth Factor/genetics , Gene Expression , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Primary Cell Culture , Promoter Regions, Genetic , Rats , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transcriptional Activation , Transforming Growth Factor beta/metabolism
11.
Cytometry A ; 83(1): 38-47, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22837061

ABSTRACT

A wide array of neural and non-neural cell types arises from the neural crest during vertebrate embryogenesis. The neural crest forms transiently in the dorsal neural primordium to yield migratory cells that will invade nearly all tissues and later, differentiate into bones and cartilages, vascular smooth muscle cells, connective tissues, neurons and glial cells of the peripheral nervous system, endocrine cells, and melanocytes. Due to the amazingly diversified array of cell types they generate, the neural crest cells represent an attractive model in the stem cell field. We review here in vivo and in vitro studies of individual cells, which led to the discovery and characterization of neural crest progenitors endowed with multipotency and stem cell properties. We also present an overview of the diverse types, marker expression, and locations of the neural crest-derived stem cells identified in the vertebrate body, with emphasis on those evidenced recently in mammalian adult tissues.


Subject(s)
Cell Differentiation/physiology , Neural Crest/cytology , Neural Stem Cells/cytology , Animals , Cell Movement/physiology , Cell Separation/methods , Embryonic Development/physiology , Gene Expression Regulation, Developmental/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL