Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Neurol Neuroimmunol Neuroinflamm ; 11(3): e200230, 2024 May.
Article in English | MEDLINE | ID: mdl-38669615

ABSTRACT

BACKGROUND AND OBJECTIVES: The aim of this study was to identify novel biomarkers for multiple sclerosis (MS) diagnosis and prognosis, addressing the critical need for specific and prognostically valuable markers in the field. METHODS: We conducted an extensive proteomic investigation, combining analysis of (1) CSF proteome from symptomatic controls, fast and slow converters after clinically isolated syndromes, and patients with relapsing-remitting MS (n = 10 per group) using label-free quantitative proteomics and (2) oligodendrocyte secretome changes under proinflammatory or proapoptotic conditions using stable isotope labeling by amino acids in cell culture. Proteins exhibiting differential abundance in both proteomic analyses were combined with other putative MS biomarkers, yielding a comprehensive list of 87 proteins that underwent quantification through parallel reaction monitoring (PRM) in a novel cohort, comprising symptomatic controls, inflammatory neurologic disease controls, and patients with MS at various disease stages (n = 10 per group). The 11 proteins that passed this qualification step were subjected to a new PRM assay within an expanded cohort comprising 158 patients with either MS at different disease stages or other inflammatory or noninflammatory neurologic disease controls. RESULTS: This study unveiled a promising biomarker signature for MS, including previously established candidates, such as chitinase 3-like protein 1, chitinase 3-like protein 2, chitotriosidase, immunoglobulin kappa chain region C, neutrophil gelatinase-associated lipocalin, and CD27. In addition, we identified novel markers, namely cat eye syndrome critical region protein 1 (adenosine deaminase 2, a therapeutic target in multiple sclerosis) and syndecan-1, a proteoglycan, also known as plasma cell surface marker CD138 and acting as chitinase 3-like protein 1 receptor implicated in inflammation and cancer signaling. CD138 exhibited good diagnostic accuracy in distinguishing MS from inflammatory neurologic disorders (area under the curve [AUC] = 0.85, CI 0.75-0.95). CD138 immunostaining was also observed in the brains of patients with MS and cultured oligodendrocyte precursor cells but was absent in astrocytes. DISCUSSION: These findings identify CD138 as a specific CSF biomarker for MS and suggest the selective activation of the chitinase 3-like protein 1/CD138 pathway within the oligodendrocyte lineage in MS. They offer promising prospects for improving MS diagnosis and prognosis by providing much-needed specificity and clinical utility. CLASSIFICATION OF EVIDENCE: This study provides Class II evidence that CD138 distinguishes multiple sclerosis from other inflammatory neurologic disorders with an AUC of 0.85 (95% CI 0.75-0.95).


Subject(s)
Biomarkers , Multiple Sclerosis, Relapsing-Remitting , Syndecan-1 , Humans , Biomarkers/cerebrospinal fluid , Adult , Female , Male , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Middle Aged , Syndecan-1/cerebrospinal fluid , Cohort Studies , Proteomics , Multiple Sclerosis/cerebrospinal fluid , Multiple Sclerosis/diagnosis , Oligodendroglia/metabolism
3.
Cell Mol Life Sci ; 80(7): 181, 2023 Jun 17.
Article in English | MEDLINE | ID: mdl-37329342

ABSTRACT

Ependymal cells lining the central canal of the spinal cord play a crucial role in providing a physical barrier and in the circulation of cerebrospinal fluid. These cells express the FOXJ1 and SOX2 transcription factors in mice and are derived from various neural tube populations, including embryonic roof and floor plate cells. They exhibit a dorsal-ventral expression pattern of spinal cord developmental transcription factors (such as MSX1, PAX6, ARX, and FOXA2), resembling an embryonic-like organization. Although this ependymal region is present in young humans, it appears to be lost with age. To re-examine this issue, we collected 17 fresh spinal cords from organ donors aged 37-83 years and performed immunohistochemistry on lightly fixed tissues. We observed cells expressing FOXJ1 in the central region in all cases, which co-expressed SOX2 and PAX6 as well as RFX2 and ARL13B, two proteins involved in ciliogenesis and cilia-mediated sonic hedgehog signaling, respectively. Half of the cases exhibited a lumen and some presented portions of the spinal cord with closed and open central canals. Co-staining of FOXJ1 with other neurodevelopmental transcription factors (ARX, FOXA2, MSX1) and NESTIN revealed heterogeneity of the ependymal cells. Interestingly, three donors aged > 75 years exhibited a fetal-like regionalization of neurodevelopmental transcription factors, with dorsal and ventral ependymal cells expressing MSX1, ARX, and FOXA2. These results provide new evidence for the persistence of ependymal cells expressing neurodevelopmental genes throughout human life and highlight the importance of further investigation of these cells.


Subject(s)
Hedgehog Proteins , Spinal Cord , Humans , Mice , Animals , Hedgehog Proteins/genetics , Spinal Cord/metabolism , Neuroglia/metabolism , Transcription Factors/metabolism , Ependyma/metabolism , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism
4.
Cells ; 10(12)2021 11 27.
Article in English | MEDLINE | ID: mdl-34943841

ABSTRACT

Ependymal cells reside in the adult spinal cord and display stem cell properties in vitro. They proliferate after spinal cord injury and produce neurons in lower vertebrates but predominantly astrocytes in mammals. The mechanisms underlying this glial-biased differentiation remain ill-defined. We addressed this issue by generating a molecular resource through RNA profiling of ependymal cells before and after injury. We found that these cells activate STAT3 and ERK/MAPK signaling post injury and downregulate cilia-associated genes and FOXJ1, a central transcription factor in ciliogenesis. Conversely, they upregulate 510 genes, seven of them more than 20-fold, namely Crym, Ecm1, Ifi202b, Nupr1, Rbp1, Thbs2 and Osmr-the receptor for oncostatin, a microglia-specific cytokine which too is strongly upregulated after injury. We studied the regulation and role of Osmr using neurospheres derived from the adult spinal cord. We found that oncostatin induced strong Osmr and p-STAT3 expression in these cells which is associated with reduction of proliferation and promotion of astrocytic versus oligodendrocytic differentiation. Microglial cells are apposed to ependymal cells in vivo and co-culture experiments showed that these cells upregulate Osmr in neurosphere cultures. Collectively, these results support the notion that microglial cells and Osmr/Oncostatin pathway may regulate the astrocytic fate of ependymal cells in spinal cord injury.


Subject(s)
Cell Lineage , Ependyma/metabolism , Gene Expression Profiling , Gene Expression Regulation , Oncostatin M/metabolism , RNA/genetics , Spinal Cord Injuries/genetics , Stem Cells/pathology , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cilia/genetics , Down-Regulation/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , Oncostatin M Receptor beta Subunit , RNA/metabolism , Spheroids, Cellular/metabolism , Spinal Cord/pathology , Up-Regulation/genetics
5.
Stem Cell Reports ; 12(5): 1159-1177, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31031189

ABSTRACT

Anamniotes, rodents, and young humans maintain neural stem cells in the ependymal zone (EZ) around the central canal of the spinal cord, representing a possible endogenous source for repair in mammalian lesions. Cell diversity and genes specific for this region are ill defined. A cellular and molecular resource is provided here for the mouse and human EZ based on RNA profiling, immunostaining, and fluorescent transgenic mice. This uncovered the conserved expression of 1,200 genes including 120 transcription factors. Unexpectedly the EZ maintains an embryonic-like dorsal-ventral pattern of expression of spinal cord developmental transcription factors (ARX, FOXA2, MSX1, and PAX6). In mice, dorsal and ventral EZ cells express Vegfr3 and are derived from the embryonic roof and floor plates. The dorsal EZ expresses a high level of Bmp6 and Gdf10 genes and harbors a subpopulation of radial quiescent cells expressing MSX1 and ID4 transcription factors.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental , RNA/genetics , Spinal Cord/metabolism , Stem Cells/metabolism , Animals , Embryonic Stem Cells/cytology , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Female , Humans , MSX1 Transcription Factor/genetics , MSX1 Transcription Factor/metabolism , Male , Mice , Mice, Transgenic , Microscopy, Fluorescence , Middle Aged , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , RNA/metabolism , Spinal Cord/cytology , Stem Cell Niche , Stem Cells/cytology , Young Adult
6.
Mol Ther ; 27(6): 1101-1113, 2019 06 05.
Article in English | MEDLINE | ID: mdl-31005598

ABSTRACT

Most cases of sensorineural deafness are caused by degeneration of hair cells. Although stem/progenitor cell therapy is becoming a promising treatment strategy in a variety of organ systems, cell engraftment in the adult mammalian cochlea has not yet been demonstrated. In this study, we generated human otic progenitor cells (hOPCs) from induced pluripotent stem cells (iPSCs) in vitro and identified these cells by the expression of known otic markers. We showed successful cell transplantation of iPSC-derived-hOPCs in an in vivo adult guinea pig model of ototoxicity. The delivered hOPCs migrated throughout the cochlea, engrafted in non-sensory regions, and survived up to 4 weeks post-transplantation. Some of the engrafted hOPCs responded to environmental cues within the cochlear sensory epithelium and displayed molecular features of early sensory differentiation. We confirmed these results with hair cell progenitors derived from Atoh1-GFP mice as donor cells. These mouse otic progenitors transplanted using the same in vivo delivery system migrated into damaged cochlear sensory epithelium and adopted a partial sensory cell fate. This is the first report of the survival and differentiation of hOPCs in ototoxic-injured mature cochlear epithelium, and it should stimulate further research into cell-based therapies for treatment of deafness.


Subject(s)
Cell Enlargement , Hair Cells, Auditory/drug effects , Hearing Loss/surgery , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/transplantation , Ototoxicity/surgery , Stem Cell Transplantation/methods , Amikacin/adverse effects , Amikacin/pharmacology , Animals , Auditory Threshold/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cyclosporine/pharmacology , Disease Models, Animal , Fibroblast Growth Factor 10/pharmacology , Fibroblast Growth Factor 3/pharmacology , Guinea Pigs , Hair Cells, Auditory/immunology , Hair Cells, Auditory/metabolism , Hearing Loss/chemically induced , Humans , Immunosuppressive Agents/pharmacology , Induced Pluripotent Stem Cells/immunology , Living Donors
7.
Aging (Albany NY) ; 10(6): 1442-1453, 2018 Jun 19.
Article in English | MEDLINE | ID: mdl-29920476

ABSTRACT

Progressive loss of tissue homeostasis is a hallmark of numerous age-related pathologies, including osteoarthritis (OA). Accumulation of senescent chondrocytes in joints contributes to the age-dependent cartilage loss of functions through the production of hypertrophy-associated catabolic matrix-remodeling enzymes and pro-inflammatory cytokines. Here, we evaluated the effects of the secreted variant of the anti-aging hormone α-Klotho on cartilage homeostasis during both cartilage formation and OA development. First, we found that α-Klotho expression was detected during mouse limb development, and transiently expressed during in vitro chondrogenic differentiation of bone marrow-derived mesenchymal stem cells. Genome-wide gene array analysis of chondrocytes from OA patients revealed that incubation with recombinant secreted α-Klotho repressed expression of the NOS2 and ZIP8/MMP13 catabolic remodeling axis. Accordingly, α-Klotho expression was reduced in chronically IL1ß-treated chondrocytes and in cartilage of an OA mouse model. Finally, in vivo intra-articular secreted α-Kotho gene transfer delays cartilage degradation in the OA mouse model. Altogether, our results reveal a new tissue homeostatic function for this anti-aging hormone in protecting against OA onset and progression.


Subject(s)
Cation Transport Proteins/metabolism , Glucuronidase/metabolism , Homeostasis/physiology , Matrix Metalloproteinase 13/metabolism , Nitric Oxide Synthase Type II/metabolism , Animals , Cartilage/growth & development , Cartilage/metabolism , Cation Transport Proteins/genetics , Chondrocytes/metabolism , Embryo, Mammalian/metabolism , Gene Expression Regulation/physiology , Glucuronidase/genetics , Humans , Klotho Proteins , Matrix Metalloproteinase 13/genetics , Mice
8.
Stem Cells ; 36(2): 192-205, 2018 02.
Article in English | MEDLINE | ID: mdl-29044892

ABSTRACT

One strategy for stem cell-based therapy of the cerebral cortex involves the generation and transplantation of functional, histocompatible cortical-like neurons from embryonic stem cells (ESCs). Diploid parthenogenetic Pg-ESCs have recently emerged as a promising source of histocompatible ESC derivatives for organ regeneration but their utility for cerebral cortex therapy is unknown. A major concern with Pg-ESCs is genomic imprinting. In contrast with biparental Bp-ESCs derived from fertilized oocytes, Pg-ESCs harbor two maternal genomes but no sperm-derived genome. Pg-ESCs are therefore expected to have aberrant expression levels of maternally expressed (MEGs) and paternally expressed (PEGs) imprinted genes. Given the roles of imprinted genes in brain development, tissue homeostasis and cancer, their deregulation in Pg-ESCs might be incompatible with therapy. Here, we report that, unexpectedly, only one gene out of 7 MEGs and 12 PEGs was differentially expressed between Pg-ESCs and Bp-ESCs while 13 were differentially expressed between androgenetic Ag-ESCs and Bp-ESCs, indicating that Pg-ESCs but not Ag-ESCs, have a Bp-like imprinting compatible with therapy. In vitro, Pg-ESCs generated cortical-like progenitors and electrophysiologically active glutamatergic neurons that maintained the Bp-like expression levels for most imprinted genes. In vivo, Pg-ESCs participated to the cortical lineage in fetal chimeras. Finally, transplanted Pg-ESC derivatives integrated into the injured adult cortex and sent axonal projections in the host brain. In conclusion, mouse Pg-ESCs generate functional cortical-like neurons with Bp-like imprinting and their derivatives properly integrate into both the embryonic cortex and the injured adult cortex. Collectively, our data support the utility of Pg-ESCs for cortical therapy. Stem Cells 2018;36:192-205.


Subject(s)
Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Animals , DNA Methylation/genetics , DNA Methylation/physiology , Electrophysiology , Genomic Imprinting/genetics , Genomic Imprinting/physiology , Mice , Mice, Inbred C57BL , Neurons/cytology , Neurons/metabolism , Parthenogenesis/genetics , Parthenogenesis/physiology
9.
J Biotechnol ; 236: 35-44, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27497759

ABSTRACT

Fluorescence expression tools for stable and innocuous whole mycoplasma cell labelling have been developed. A Tn4001-derivative mini-transposon affording unmarked, stable mutagenesis in mycoplasmas was modified to allow the constitutive, high-level expression of mCherry, mKO2 and mNeonGreen. These tools were used to introduce the respective fluorescent proteins as chromosomal tags in the phylogenetically distant species Mycoplasma mycoides subsp. mycoides and Mycoplasma bovis. The production, selection and characterisation of fluorescent clones were straightforward and resulted in the unprecedented observation of red and green fluorescent mycoplasma colonies in the two species, with no apparent cytotoxicity. Equivalent fluorescence expression levels were quantified by flow cytometry in both species, suggesting that these tools can be broadly applied in mycoplasmas. A macrophage infection assay was performed to assess the usefulness of mNeonGreen-expressing strains for monitoring mycoplasma infections, and notably cell invasion. The presence of fluorescent mycoplasmas inside live phagocytic cells was detected and quantified by flow cytometry and corroborated by confocal microscopy, which allowed the identification of individual mycoplasmas in the cytoplasm of infected cells. The fluorescence expression tools developed in this study are suitable for host-pathogen interaction studies and offer innumerable perspectives for the functional analysis of mycoplasmas both in vitro and in vivo.


Subject(s)
Host-Pathogen Interactions/physiology , Molecular Imaging/methods , Mycoplasma/chemistry , Mycoplasma/cytology , Spectrometry, Fluorescence/methods , Animals , Cattle , Cells, Cultured , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mycoplasma/metabolism , Mycoplasma/pathogenicity , Mycoplasma Infections/microbiology , Mycoplasma Infections/physiopathology , Phagocytes/cytology , Phagocytes/microbiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Reproducibility of Results
10.
PLoS One ; 11(3): e0151274, 2016.
Article in English | MEDLINE | ID: mdl-26953813

ABSTRACT

Asymmetric division (AD) is a fundamental mechanism whereby unequal inheritance of various cellular compounds during mitosis generates unequal fate in the two daughter cells. Unequal repartitions of transcription factors, receptors as well as mRNA have been abundantly described in AD. In contrast, the involvement of intermediate filaments in this process is still largely unknown. AD occurs in stem cells during development but was also recently observed in cancer stem cells. Here, we demonstrate the asymmetric distribution of the main astrocytic intermediate filament, namely the glial fibrillary acid protein (GFAP), in mitotic glioma multipotent cells isolated from glioblastoma (GBM), the most frequent type of brain tumor. Unequal mitotic repartition of GFAP was also observed in mice non-tumoral neural stem cells indicating that this process occurs across species and is not restricted to cancerous cells. Immunofluorescence and videomicroscopy were used to capture these rare and transient events. Considering the role of intermediate filaments in cytoplasm organization and cell signaling, we propose that asymmetric distribution of GFAP could possibly participate in the regulation of normal and cancerous neural stem cell fate.


Subject(s)
Asymmetric Cell Division , Glial Fibrillary Acidic Protein/metabolism , Glioma/metabolism , Multipotent Stem Cells/metabolism , Neoplastic Stem Cells/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Gene Expression , Genes, Reporter , Glial Fibrillary Acidic Protein/genetics , Humans , Mice , Mitosis , Neural Stem Cells/metabolism , Protein Transport
11.
PLoS One ; 10(4): e0122337, 2015.
Article in English | MEDLINE | ID: mdl-25875008

ABSTRACT

The proliferation and differentiation of neural stem cells are tightly controlled by intrinsic and extrinsic cues. Cell adhesion molecules are increasingly recognized as regulators of these processes. Here we report the expression of the olfactory cell adhesion molecule (OCAM/NCAM2/RNCAM) during mouse spinal cord development and in neural stem cells cultured as neurospheres. OCAM is also weakly expressed in the dormant adult stem cell niche around the central canal and is overexpressed after spinal cord injury. Both transmembrane (TM) and glycosylphosphatidylinositol (GPI)-linked isoforms are present in neurospheres. Electron microscopy and internalisation experiments revealed a dynamic trafficking of OCAM between the membrane and intracellular compartments. After differentiation, OCAM remains in neurons and oligodendrocytes whereas no expression is detected in astrocytes. Using OCAM knockout (KO) mice, we found that mutant spinal cord stem cells showed an increased proliferation and self-renewal rates although no effect on differentiation was observed. This effect was reversed by lentivirus-mediated re-introduction of OCAM. Mechanistically, we identified the ErbB2/Neu/HER2 protein as being implicated in the enhanced proliferation of mutant cells. ErbB2 protein expression and phosphorylation level were significantly increased in KO cells whereas no difference was observed at the mRNA level. Overexpression of ErbB2 in wild-type and mutant cells also increased their growth while reintroduction of OCAM in mutant cells reduced the level of phosphorylated ErbB2. These results indicate that OCAM exerts a posttranscriptional control on the ErbB2 signalling in spinal cord stem cells. This study adds further support for considering cell adhesion molecules as regulators of the ErbB signalling.


Subject(s)
Embryonic Stem Cells/metabolism , Neural Cell Adhesion Molecules/biosynthesis , Receptor, ErbB-2/biosynthesis , Spinal Cord/metabolism , Animals , Cell Adhesion/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Neural Cell Adhesion Molecules/genetics , RNA, Messenger/biosynthesis , Receptor, ErbB-2/genetics , Signal Transduction/genetics , Spinal Cord/growth & development
12.
Glia ; 61(2): 225-39, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23047160

ABSTRACT

Glioblastoma multiform (GBM) are devastating brain tumors containing a fraction of multipotent stem-like cells which are highly tumorigenic. These cells are resistant to treatments and are likely to be responsible for tumor recurrence. One approach to eliminate GBM stem-like cells would be to force their terminal differentiation. During development, neurons formation is controlled by neurogenic transcription factors such as Ngn1/2 and NeuroD1. We found that in comparison with oligodendrogenic genes, the expression of these neurogenic genes is low or absent in GBM tumors and derived cultures. We thus explored the effect of overexpressing these neurogenic genes in three CD133(+) Sox2(+) GBM stem-like cell cultures and the U87 glioma line. Introduction of Ngn2 in CD133(+) cultures induced massive cell death, proliferation arrest and a drastic reduction of neurosphere formation. Similar effects were observed with NeuroD1. Importantly, Ngn2 effects were accompanied by the downregulation of Olig2, Myc, Shh and upregulation of Dcx and NeuroD1 expression. The few surviving cells adopted a typical neuronal morphology and some of them generated action potentials. These cells appeared to be produced at the expense of GFAP(+) cells which were radically reduced after differentiation with Ngn2. In vivo, Ngn2-expressing cells were unable to form orthotopic tumors. In the U87 glioma line, Ngn2 could not induce neuronal differentiation although proliferation in vitro and tumoral growth in vivo were strongly reduced. By inducing cell death, cell cycle arrest or differentiation, this work supports further exploration of neurogenic proteins to oppose GBM stem-like and non-stem-like cell growth.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/pharmacology , Brain Neoplasms/pathology , Cell Differentiation , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Transcription Factors/pharmacology , AC133 Antigen , Antigens, CD/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Death , Flow Cytometry , Glial Fibrillary Acidic Protein/metabolism , Glycoproteins/metabolism , Hedgehog Proteins/metabolism , Humans , Neoplastic Stem Cells/drug effects , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/pharmacology , Oligodendrocyte Transcription Factor 2 , Oncogene Protein p55(v-myc)/metabolism , Peptides/metabolism , SOXB1 Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured
13.
BMC Neurosci ; 12: 99, 2011 Oct 10.
Article in English | MEDLINE | ID: mdl-21985235

ABSTRACT

BACKGROUND: The adult central nervous system (CNS) contains different populations of immature cells that could possibly be used to repair brain and spinal cord lesions. The diversity and the properties of these cells in the human adult CNS remain to be fully explored. We previously isolated Nestin+ Sox2+ neural multipotential cells from the adult human spinal cord using the neurosphere method (i.e. non adherent conditions and defined medium). RESULTS: Here we report the isolation and long term propagation of another population of Nestin+ cells from this tissue using adherent culture conditions and serum. QPCR and immunofluorescence indicated that these cells had mesenchymal features as evidenced by the expression of Snai2 and Twist1 and lack of expression of neural markers such as Sox2, Olig2 or GFAP. Indeed, these cells expressed markers typical of smooth muscle vascular cells such as Calponin, Caldesmone and Acta2 (Smooth muscle actin). These cells could not differentiate into chondrocytes, adipocytes, neuronal and glial cells, however they readily mineralized when placed in osteogenic conditions. Further characterization allowed us to identify the Nkx6.1 transcription factor as a marker for these cells. Nkx6.1 was expressed in vivo by CNS vascular muscular cells located in the parenchyma and the meninges. CONCLUSION: Smooth muscle cells expressing Nestin and Nkx6.1 is the main cell population derived from culturing human spinal cord cells in adherent conditions with serum. Mineralization of these cells in vitro could represent a valuable model for studying calcifications of CNS vessels which are observed in pathological situations or as part of the normal aging. In addition, long term propagation of these cells will allow the study of their interaction with other CNS cells and their implication in scar formation during spinal cord injury.


Subject(s)
Calcification, Physiologic/physiology , Homeodomain Proteins/metabolism , Intermediate Filament Proteins/metabolism , Myocytes, Smooth Muscle/metabolism , Nerve Tissue Proteins/metabolism , Primary Cell Culture/methods , Spinal Cord/blood supply , Spinal Cord/metabolism , Adult , Cell Adhesion/physiology , Cell Separation/methods , Homeodomain Proteins/blood , Humans , Intermediate Filament Proteins/blood , Myocytes, Smooth Muscle/cytology , Nerve Tissue Proteins/blood , Nestin , Spinal Cord/cytology
14.
Stem Cells ; 27(11): 2722-33, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19785035

ABSTRACT

In humans and rodents the adult spinal cord harbors neural stem cells located around the central canal. Their identity, precise location, and specific signaling are still ill-defined and controversial. We report here on a detailed analysis of this niche. Using microdissection and glial fibrillary acidic protein (GFAP)-green fluorescent protein (GFP) transgenic mice, we demonstrate that neural stem cells are mostly dorsally located GFAP(+) cells lying ependymally and subependymally that extend radial processes toward the pial surface. The niche also harbors doublecortin protein (Dcx)(+) Nkx6.1(+) neurons sending processes into the lumen. Cervical and lumbar spinal cord neural stem cells maintain expression of specific rostro-caudal Hox gene combinations and the niche shows high levels of signaling proteins (CD15, Jagged1, Hes1, differential screening-selected gene aberrative in neuroblastoma [DAN]). More surprisingly, the niche displays mesenchymal traits such as expression of epithelial-mesenchymal-transition zinc finger E-box-binding protein 1 (ZEB1) transcription factor and smooth muscle actin. We found ZEB1 to be essential for neural stem cell survival in vitro. Proliferation within the niche progressively ceases around 13 weeks when the spinal cord reaches its final size, suggesting an active role in postnatal development. In addition to hippocampus and subventricular zone niches, adult spinal cord constitutes a third central nervous system stem cell niche with specific signaling, cellular, and structural characteristics that could possibly be manipulated to alleviate spinal cord traumatic and degenerative diseases.


Subject(s)
Glial Fibrillary Acidic Protein/metabolism , Homeodomain Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , Stem Cell Niche/cytology , Stem Cell Niche/metabolism , Stem Cells/cytology , Actins/metabolism , Animals , Cell Proliferation , Doublecortin Protein , Gene Expression Regulation, Developmental , Glial Fibrillary Acidic Protein/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mice , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , Stem Cells/metabolism , Zinc Finger E-box-Binding Homeobox 1
15.
Neuroreport ; 18(15): 1539-42, 2007 Oct 08.
Article in English | MEDLINE | ID: mdl-17885597

ABSTRACT

Neurosphere cultures provide a useful model to study neural stem/progenitor cells (NSC/NPCs). The degree to which neurospheres (NS) retain their regional identity in vitro has, however, been questioned. Here, NS obtained from mouse embryonic cortex, striatum or spinal cord were compared after differentiation. Neurons from cortical NS formed well ordered clusters containing astrocytes, those from striatal NS formed an external ring at the borderof the astrocyte layer, whereas those from spinal cord NS spread radially like the astrocytes. Such in-vitro neural behaviour was region-specific and persisted in clonal conditions, providing evidence of the maintenance of positional cues in NS cultures.


Subject(s)
Embryonic Stem Cells/physiology , Neurons/physiology , Animals , Astrocytes/physiology , Cell Differentiation/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Clone Cells , Cluster Analysis , Female , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Neostriatum/cytology , Neostriatum/embryology , Nerve Fibers/physiology , Pregnancy , Spinal Cord/cytology , Spinal Cord/embryology
16.
Stem Cells ; 25(2): 340-53, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17053213

ABSTRACT

Neural stem cells cultured with fibroblast growth factor 2 (FGF2)/epidermal growth factor (EGF) generate clonal expansions called neurospheres (NS), which are widely used for therapy in animal models. However, their cellular composition is still poorly defined. Here, we report that NS derived from several embryonic and adult central nervous system (CNS) regions are composed mainly of remarkable cells coexpressing radial glia markers (BLBP, RC2, GLAST), oligodendrogenic/neurogenic factors (Mash1, Olig2, Nkx2.2), and markers that in vivo are typical of the oligodendrocyte lineage (NG2, A2B5, PDGFR-alpha). On NS differentiation, the latter remain mostly expressed in neurons, together with Olig2 and Mash1. Using cytometry, we show that in growing NS the small population of multipotential self-renewing NS-forming cells are A2B5(+) and NG2(+). Additionally, we demonstrate that these NS-forming cells in the embryonic spinal cord were initially NG2(-) and rapidly acquired NG2 in vitro. NG2 and Olig2 were found to be rapidly induced by cell culture conditions in spinal cord neural precursor cells. Olig2 expression was also induced in astrocytes and embryonic peripheral nervous system (PNS) cells in culture after EGF/FGF treatment. These data provide new evidence for profound phenotypic modifications in CNS and PNS neural precursor cells induced by culture conditions.


Subject(s)
Antigens/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Central Nervous System/cytology , Nerve Tissue Proteins/metabolism , Neurons/cytology , Peripheral Nervous System/cytology , Phenotype , Proteoglycans/metabolism , Stem Cells/cytology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation , Cells, Cultured , Embryo, Mammalian/cytology , Gangliosides/metabolism , Gene Expression Regulation , High Mobility Group Proteins/metabolism , Homeobox Protein Nkx-2.2 , Mice , Models, Biological , Nerve Tissue Proteins/genetics , Neuroglia/cytology , Neuroglia/metabolism , Neurons/metabolism , Oligodendrocyte Transcription Factor 2 , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , SOX9 Transcription Factor , Spinal Cord/cytology , Spinal Cord/embryology , Stem Cells/metabolism , Transcription Factors/metabolism
17.
Brain Res Mol Brain Res ; 105(1-2): 98-107, 2002 Sep 30.
Article in English | MEDLINE | ID: mdl-12399112

ABSTRACT

The functioning of the mammalian cochlea is entirely based on its mechanical properties, which are supported by a highly complex tissue architecture resulting from the precise arrangement of sensory hair cells and non-sensory supporting cells. Growing evidence indicates that evolutionary conserved signaling pathways are involved in inner ear development and in the differentiation of its diverse cell types. We investigated whether members of the Wnt and Frizzled gene families, which play key roles in a wide variety of cellular and developmental processes, are expressed in the postnatal rat cochlea. A PCR screening of a rat cochlea cDNA library performed with degenerate primers allowed us to isolate five members of the Wnt gene family (RWnt-2B, -4, -5A, -5B, and -7A) and six members of the Frizzled gene family (Rfz1, Rfz2, Rfz3, Rfz4, Rfz6, Rfz9). In situ hybridization and immunocytochemistry experiments demonstrated that RWnt-4, -5B, -7A have distinct, although partly overlapping, expression patterns in the juvenile rat cochlea. These results suggest that the Wnt-Frizzled signaling pathway could be involved in several aspects of late cochlear differentiation and/or auditory function.


Subject(s)
Cell Differentiation/genetics , Cochlea/growth & development , Gene Expression Regulation, Developmental/genetics , Proto-Oncogene Proteins/genetics , Receptors, Neurotransmitter/genetics , Signal Transduction/genetics , Zebrafish Proteins , Aging/genetics , Aging/metabolism , Animals , Animals, Newborn , Cochlea/cytology , Cochlea/metabolism , DNA, Complementary/analysis , DNA, Complementary/genetics , Frizzled Receptors , Gene Library , Genetic Testing , Glycoproteins/genetics , Glycoproteins/metabolism , Immunohistochemistry , Labyrinth Supporting Cells/cytology , Labyrinth Supporting Cells/metabolism , Proto-Oncogene Proteins/metabolism , RNA, Messenger/metabolism , Rats , Receptors, G-Protein-Coupled , Receptors, Neurotransmitter/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Wnt Proteins , Wnt4 Protein
18.
J Comp Neurol ; 442(1): 6-22, 2002 Jan 01.
Article in English | MEDLINE | ID: mdl-11754363

ABSTRACT

Hair cell losses in the mammalian cochlea following an ototoxic insult are irreversible. However, past studies have shown that amikacin treatment in rat cochleae resulted in the transient presence of atypical Deiters' cells (ACs) in the damaged organ of Corti. These ACs arise through a transformation of Deiters' cells, which produce, at their apical pole, densely packed microvilli reminiscent of early-differentiating stereociliary bundles. The ACs do not, however, express typical hair cell markers such as parvalbumin or calbindin. The present study was designed to determine whether specific growth factors could influence the survival and differentiation of these ACs and stimulate hair cell regeneration processes in vitro. Apical-medial segments of organ of Corti of juvenile amikacin-treated rats were established as organotypic cultures, and the effects of epidermal growth factor (EGF), insulin-like growth factor 1 (IGF-1), transforming growth factor-alpha (TGFalpha), and retinoic acid were studied using morphological and molecular approaches. Our results indicate that TGFalpha supports the survival of the damaged organ of Corti and influences ACs differentiation in vitro, possibly acting through reorganization of the actin cytoskeleton. These effects could be directly mediated through activation of the EGF receptor, which is expressed by supporting cells in the mature organ of Corti. TGFalpha does not, however, allow the ACs to progress towards a hair cell phenotype.


Subject(s)
Cell Differentiation/drug effects , Cell Survival/drug effects , Hair Cells, Auditory/drug effects , Nerve Degeneration/drug therapy , Nerve Regeneration/drug effects , Neurotoxins/toxicity , Transforming Growth Factor alpha/pharmacology , Actins/drug effects , Actins/metabolism , Amikacin/toxicity , Animals , Anti-Bacterial Agents/toxicity , Bromodeoxyuridine , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Survival/physiology , Epidermal Growth Factor/metabolism , Epidermal Growth Factor/pharmacology , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/ultrastructure , Immunohistochemistry , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacology , Microscopy, Electron , Microscopy, Electron, Scanning , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Nerve Regeneration/physiology , Neuroglia/drug effects , Neuroglia/metabolism , Neuroglia/ultrastructure , Organ Culture Techniques , Rats , Rats, Wistar , Transforming Growth Factor alpha/metabolism , Tretinoin/metabolism , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...