Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Oncotarget ; 10(24): 2397-2415, 2019 Mar 22.
Article in English | MEDLINE | ID: mdl-31040930

ABSTRACT

Glioblastoma resists chemotherapy then recurs as a fatal space-occupying lesion. To improve the prognosis, the issues of chemoresistance and tumor size should be addressed. Glioma stem cell (GSC) populations, a heterogeneous power-law coded population in glioblastoma, are believed to be responsible for the recurrence and progressive expansion of tumors. Thus, we propose a therapeutic strategy of reducing the initial size and controlling the regrowth of GSC populations which directly facilitates initial and long-term control of glioblastoma recurrence. In this study, we administered an anti-glioma/GSC drug temozolomide (TMZ) and OTS964, an inhibitor for T-Lak cell originated protein kinase, in combination (T&O), investigating whether together they efficiently and substantially shrink the initial size of power-law coded GSC populations and slow the long-term re-growth of drug-resistant GSC populations. We employed a detailed quantitative approach using clonal glioma sphere (GS) cultures, measuring sphere survivability and changes to growth during the self-renewal. T&O eliminated self-renewing GS clones and suppressed their growth. We also addressed whether T&O reduced the size of self-renewed GS populations. T&O quickly reduced the size of GS populations via efficient elimination of GS clones. The growth of the surviving T&O-resistant GS populations was continuously disturbed, leading to substantial long-term shrinkage of the self-renewed GS populations. Thus, T&O reduced the initial size of GS populations and suppressed their later regrowth. A combination therapy of TMZ and OTS964 would represent a novel therapeutic paradigm with the potential for long-term control of glioblastoma recurrence via immediate and sustained shrinkage of power-law coded heterogeneous GSC populations.

2.
Oncotarget ; 9(3): 3043-3059, 2018 Jan 09.
Article in English | MEDLINE | ID: mdl-29423027

ABSTRACT

Glioblastoma resists chemoradiotherapy, then, recurs to be a fatal space-occupying lesion. The recurrence is caused by re-growing cell populations such as glioma stem cells (GSCs), suggesting that GSC populations should be targeted. This study addressed whether a novel anti-cancer drug, OTS964, an inhibitor for T-LAK cell originated protein kinase (TOPK), is effective in reducing the size of the heterogeneous GSC populations, a power-law coded heterogeneous GSC populations consisting of glioma sphere (GS) clones, by detailing quantitative growth properties. We found that OTS964 killed GS clones while suppressing the growth of surviving GS clones, thus identifying clone-eliminating and growth-disturbing efficacies of OTS964. The efficacies led to a significant size reduction in GS populations in a dose-dependent manner. The surviving GS clones reconstructed GS populations in the following generations; the recovery of GS populations fits a recurrence after the chemotherapy. The recovering GS clones resisted the clone-eliminating effect of OTS964 in sequential exposure during the growth recovery. However, surprisingly, the resistant properties of the recovered-GS clones had been plastically canceled during self-renewal, and then the GS clones had become re-sensitive to OTS964. Thus, OTS964 targets GSCs to eliminate them or suppress their growth, resulting in shrinkage of the power-law coded GSC populations. We propose a therapy focusing on long-term control in recurrence of glioblastoma via reducing the size of the GSC populations by OTS964.

3.
Neuropathology ; 38(2): 144-153, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29271010

ABSTRACT

This study aimed to evaluate the biological features of T-lymphokine-activated killer cell-originating protein kinase (TOPK) in vitro and to assess clinical impact of TOPK on the outcome in patients with malignant glioma. TOPK protein level and TOPK mRNA and protein levels in six glioma cell lines were examined using Western blot and reverse transcription-polymerase chain reaction (RT-PCR), respectively. Immunohistochemistry was performed to examine their subcellular localization of TOPK. Using surgical specimens from 57 patients with gliomas, TOPK and Ki-67 expressions were examined by immunohistochemistry. Their co-localization was also examined with double immunofluorescence immunohistochemistry. Impacts of TOPK/Ki-67 expression on the overall survival (OS) and progression-free survival (PFS) in 32 patients with glioblastoma multiforme (GBM) were examined, using Kaplan-Meier and Cox proportion hazard models. Immunohistochemistry revealed that approximately 20-30% of glioma cells were positive for TOPK in vitro. TOPK mRNA was identified in all glioma cell lines on RT-PCR. The value of TOPK/GAPDH was 0.27 ± 0.11. TOPK and Ki-67 expressions were significantly higher in GBM patients than in non-GBM patients. A majority of TOPK-positive cells were also positive for Ki-67 and vice versa. Multivariate analysis revealed that a low TOPK expression (≤ 12.7%) was an independent predictor of longer OS (P = 0.0372), and that gross total removal and a low TOPK expression (≤ 12.7%) were independent predictors of longer PFS (P = 0.0470 and P = 0.0189, respectively). The findings strongly suggest biological and clinical importance of TOPK expression in gliomas, indicating a novel therapeutic potential of TOPK inhibitors to treat malignant gliomas.


Subject(s)
Brain Neoplasms/diagnosis , Glioma/diagnosis , Mitogen-Activated Protein Kinase Kinases/metabolism , Adolescent , Adult , Aged , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Cell Line, Tumor , Female , Glioblastoma/diagnosis , Glioblastoma/metabolism , Glioma/enzymology , Glioma/genetics , Humans , Ki-67 Antigen/metabolism , Male , Middle Aged , Mitogen-Activated Protein Kinase Kinases/genetics , Prognosis , Proportional Hazards Models , Young Adult
4.
Sci Rep ; 7(1): 3855, 2017 06 20.
Article in English | MEDLINE | ID: mdl-28634350

ABSTRACT

Pericytes are believed to originate from either mesenchymal or neural crest cells. It has recently been reported that pericytes play important roles in the central nervous system (CNS) by regulating blood-brain barrier homeostasis and blood flow at the capillary level. However, the origin of CNS microvascular pericytes and the mechanism of their recruitment remain unknown. Here, we show a new source of cerebrovascular pericytes during neurogenesis. In the CNS of embryonic day 10.5 mouse embryos, CD31+F4/80+ hematopoietic lineage cells were observed in the avascular region around the dorsal midline of the developing midbrain. These cells expressed additional macrophage markers such as CD206 and CD11b. Moreover, the CD31+F4/80+ cells phagocytosed apoptotic cells as functionally matured macrophages, adhered to the newly formed subventricular vascular plexus, and then divided into daughter cells. Eventually, these CD31+F4/80+ cells transdifferentiated into NG2/PDGFRß/desmin-expressing cerebrovascular pericytes, enwrapping and associating with vascular endothelial cells. These data indicate that a subset of cerebrovascular pericytes derive from mature macrophages in the very early phase of CNS vascular development, which in turn are recruited from sites of embryonic hematopoiesis such as the yolk sac by way of blood flow.


Subject(s)
Central Nervous System/blood supply , Macrophages/cytology , Macrophages/metabolism , Pericytes/cytology , Pericytes/metabolism , Animals , Biomarkers , Capillaries/embryology , Cell Tracking , Cell Transdifferentiation , Mice , Mice, Knockout , Phenotype
5.
PLoS One ; 10(8): e0135760, 2015.
Article in English | MEDLINE | ID: mdl-26284929

ABSTRACT

BACKGROUND: Accumulating evidence indicates that cancer stem cells (CSCs) drive tumorigenesis. This suggests that CSCs should make ideal therapeutic targets. However, because CSC populations in tumors appear heterogeneous, it remains unclear how CSCs might be effectively targeted. To investigate the mechanisms by which CSC populations maintain heterogeneity during self-renewal, we established a glioma sphere (GS) forming model, to generate a population in which glioma stem cells (GSCs) become enriched. We hypothesized, based on the clonal evolution concept, that with each passage in culture, heterogeneous clonal sublines of GSs are generated that progressively show increased proliferative ability. METHODOLOGY/PRINCIPAL FINDINGS: To test this hypothesis, we determined whether, with each passage, glioma neurosphere culture generated from four different glioma cell lines become progressively proliferative (i.e., enriched in large spheres). Rather than monitoring self-renewal, we measured heterogeneity based on neurosphere clone sizes (#cells/clone). Log-log plots of distributions of clone sizes yielded a good fit (r>0.90) to a straight line (log(% total clones) = k*log(#cells/clone)) indicating that the system follows a power-law (y = xk) with a specific degree exponent (k = -1.42). Repeated passaging of the total GS population showed that the same power-law was maintained over six passages (CV = -1.01 to -1.17). Surprisingly, passage of either isolated small or large subclones generated fully heterogeneous populations that retained the original power-law-dependent heterogeneity. The anti-GSC agent Temozolomide, which is well known as a standard therapy for glioblastoma multiforme (GBM), suppressed the self-renewal of clones, but it never disrupted the power-law behavior of a GS population. CONCLUSIONS/SIGNIFICANCE: Although the data above did not support the stated hypothesis, they did strongly suggest a novel mechanism that underlies CSC heterogeneity. They indicate that power-law growth governs the self-renewal of heterogeneous glioma stem cell populations. That the data always fit a power-law suggests that: (i) clone sizes follow continuous, non-random, and scale-free hierarchy; (ii) precise biologic rules that reflect self-organizing emergent behaviors govern the generation of neurospheres. That the power-law behavior and the original GS heterogeneity are maintained over multiple passages indicates that these rules are invariant. These self-organizing mechanisms very likely underlie tumor heterogeneity during tumor growth. Discovery of this power-law behavior provides a mechanism that could be targeted in the development of new, more effective, anti-cancer agents.


Subject(s)
Cell Self Renewal , Glioma/pathology , Neoplastic Stem Cells/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Cell Size , Clone Cells/drug effects , Clone Cells/pathology , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Glioma/drug therapy , Humans , Models, Biological , Molecular Targeted Therapy , Neoplastic Stem Cells/drug effects , Temozolomide
6.
Biochem Biophys Res Commun ; 432(1): 66-72, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23376073

ABSTRACT

Trichohyalin-like 1 (TCHHL1) protein is a novel member of the fused-type S100 protein gene family. The deduced amino acid sequence of TCHHL1 contains an EF-hand domain in the N-terminus, one trans-membrane domain and a nuclear localization signal. We generated specific antibodies against the C-terminus of the TCHHL1 protein and examined the expression of TCHHL1 proteins in normal and pathological human skin. An immunohistochemical study showed that TCHHL1 proteins were expressed in the basal layer of the normal epidermis. In addition, signals of TCHHL1 proteins were observed around the nuclei of cultured growing keratinocytes. Accordingly, TCHHL1 mRNA has been detected in normal skin and cultured growing keratinocytes. Furthermore, TCHHL1 proteins were strongly expressed in the peripheral areas of tumor nests in basal cell carcinomas and squamous cell carcinomas. A dramatic increase in the number of Ki67 positive cells was observed in TCHHL1-expressing areas. The expression of TCHHL1 proteins also increased in non-cancerous hyperproliferative epidermal tissues such as those of psoriasis vulgaris and lichen planus. These findings highlight the possibility that TCHHL1 proteins are expressed in growing keratinocytes of the epidermis and might be associated with the proliferation of keratinocytes.


Subject(s)
Lichen Planus/metabolism , Psoriasis/metabolism , S100 Proteins/metabolism , Skin/metabolism , Amino Acid Sequence , Cell Proliferation , Cells, Cultured , Epidermis/metabolism , Epidermis/pathology , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Molecular Sequence Data , S100 Proteins/analysis , S100 Proteins/genetics , Skin/pathology
7.
Hippocampus ; 21(5): 565-74, 2011 May.
Article in English | MEDLINE | ID: mdl-20169538

ABSTRACT

Recording evoked local field potentials (LFPs) in the hippocampus in vivo has yielded us useful information about the neural mechanisms of learning and memory. Although this technique has been used in studies of the hippocampus of rodents, lagomorphs, and felines, it has not yet been applied to the primate hippocampus. Here, we report a method for recording evoked LFPs in the hippocampus of monkeys. A stimulation electrode and a recording electrode were implanted in the perforant pathway and dentate gyrus, respectively, under the guidance of electrophysiological recording. With a low stimulus intensity just above the threshold, the potential appeared as a slow positive-wave component, which was regarded as field excitatory postsynaptic potential (putative fEPSP); as stimulation intensity increased, the fEPSP amplitude increased, followed by a sharp negative component which was regarded as putative population spike. When the coordinates of the recording or stimulation electrode were moved stepwise, we observed a systematic change in the waveforms of evoked LFPs; this change corresponded to the structural arrangement through which the electrode passed. In a test for short-term synaptic plasticity by paired-pulse stimulation, potentials evoked by the second pulse were influenced by the first one in a manner dependent on interpulse intervals. In a test for long-term synaptic plasticity by high-frequency stimulation, the slopes of the fEPSPs and the area of population spikes were increased for more than 1 h. These results indicate that the method developed in the present study is useful for testing theories of hippocampal functions in primates.


Subject(s)
Dentate Gyrus/physiology , Electrophysiology/methods , Evoked Potentials/physiology , Excitatory Postsynaptic Potentials/physiology , Neuronal Plasticity/physiology , Animals , Electrodes, Implanted/standards , Female , Macaca , Neurons/physiology , Perforant Pathway/physiology
8.
J Neurosci ; 30(45): 15085-96, 2010 Nov 10.
Article in English | MEDLINE | ID: mdl-21068314

ABSTRACT

To investigate the neural basis of the associative aspects of facial identification, we recorded neuronal activity from the ventral, anterior inferior temporal cortex (AITv) of macaque monkeys during the performance of an asymmetrical paired-association (APA) task that required associative pairing between an abstract pattern and five different facial views of a single person. In the APA task, after one element of a pair (either an abstract pattern or a face) was presented as a sample cue, the reward-seeking monkey correctly identified the other element of the pair among various repeatedly presented test stimuli (faces or patterns) that were temporally separated by interstimulus delays. The results revealed that a substantial number of AITv neurons responded both to faces and abstract patterns, and the majority of these neurons responded selectively to a particular associative pair. It was demonstrated that in addition to the view-invariant identity of faces used in the APA task, the population of AITv neurons was also able to represent the associative pairing between faces and abstract patterns, which was acquired by training in the APA task. It also appeared that the effect of associative pairing was not so strong that the abstract pattern could be treated in a manner similar to a series of faces belonging to a unique identity. Together, these findings indicate that the AITv plays a crucial role in both facial identification and semantic associations with facial identities.


Subject(s)
Association Learning/physiology , Neurons/physiology , Recognition, Psychology/physiology , Temporal Lobe/physiology , Analysis of Variance , Animals , Cues , Electrodes, Implanted , Electrophysiology , Female , Macaca , Photic Stimulation
9.
Development ; 135(7): 1271-81, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18287202

ABSTRACT

Development of oligodendrocytes, myelin-forming glia in the central nervous system (CNS), proceeds on a protracted schedule. Specification of oligodendrocyte progenitors (OLPs) begins early in development, whereas their terminal differentiation occurs at late embryonic and postnatal periods. How these distinct steps are controlled remains unclear. Our previous study demonstrated an important role of the helix-loop-helix (HLH) transcription factor Ascl1 in early generation of OLPs in the developing spinal cord. Here, we show that Ascl1 is also involved in terminal differentiation of oligodendrocytes late in development. Ascl1-/- mutant mice showed a deficiency in differentiation of myelin-expressing oligodendrocytes at birth. In vitro culture studies demonstrate that the induction and maintenance of co-expression of Olig2 and Nkx2-2 in OLPs, and thyroid hormone-responsive induction of myelin proteins are impaired in Ascl1-/- mutants. Gain-of-function studies further showed that Ascl1 collaborates with Olig2 and Nkx2-2 in promoting differentiation of OLPs into oligodendrocytes in vitro. Overexpression of Ascl1, Olig2 and Nkx2-2 alone stimulated the specification of OLPs, but the combinatorial action of Ascl1 and Olig2 or Nkx2-2 was required for further promoting their differentiation into oligodendrocytes. Thus, Ascl1 regulates multiple aspects of oligodendrocyte development in the spinal cord.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Oligodendroglia/cytology , Spinal Cord/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cells, Cultured , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Mice , Mice, Mutant Strains , Models, Biological , Nerve Tissue Proteins/metabolism , Oligodendrocyte Transcription Factor 2 , Rats , Rats, Sprague-Dawley , Spinal Cord/cytology , Spinal Cord/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Transcription Factors/metabolism , Zebrafish Proteins
10.
J Neurosci ; 27(16): 4233-42, 2007 Apr 18.
Article in English | MEDLINE | ID: mdl-17442807

ABSTRACT

The bHLH (basic helix-loop-helix) transcription factor Mash1 is best known for its role in the regulation of neurogenesis. However, Mash1 is also expressed in oligodendrocyte precursors and has recently been shown to promote the generation of oligodendrocytes in cell culture, suggesting that it may regulate oligodendrogenesis as well. Here, we show that in the developing ventral forebrain, Mash1 is expressed by a subset of oligodendrocyte precursors (OPCs) as soon as they are generated in the ventricular zone. Using reporter mice, we demonstrate that a subset of OPCs in both the embryonic and postnatal forebrain originate from Mash1-positive progenitors, including a large fraction of adult NG2-positive OPCs. Using Mash1 null mutant mice, we show that Mash1 is required for the generation of an early population of OPCs in the ventral forebrain between embryonic day 11.5 (E11.5) and E13.5, whereas OPCs generated later in embryonic development are not affected. Overexpression of Mash1 in the dorsal telencephalon induces expression of PDGFRalpha (platelet-derived growth factor receptor alpha) but not other OPC markers, suggesting that Mash1 specifies oligodendrogenesis in cooperation with other factors. Analysis of double-mutant mice suggests that Olig2 is one of the factors that cooperate with Mash1 for generation of OPCs. Together, our results show for the first time that Mash1 cooperates in vivo with Olig2 in oligodendrocyte specification, demonstrating an essential role for Mash1 in the generation of a subset of oligodendrocytes and revealing a genetic heterogeneity of oligodendrocyte lineages in the mouse forebrain.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Lineage/genetics , Nerve Tissue Proteins/metabolism , Oligodendroglia/classification , Telencephalon/metabolism , Animals , Immunohistochemistry , Mice , Mice, Transgenic , Oligodendrocyte Transcription Factor 2 , Stem Cells/metabolism , Telencephalon/embryology
11.
Development ; 134(8): 1617-29, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17344230

ABSTRACT

During development, the three major neural cell lineages, neurons, oligodendrocytes and astrocytes, differentiate in specific temporal orders at topologically defined positions. How the timing and position of their generation are coordinately regulated remains poorly understood. Here, we provide evidence that the transcription factors Pax6, Olig2 and Nkx2.2 (Nkx2-2), which define the positional identity of multipotent progenitors early in development, also play crucial roles in controlling the timing of neurogenesis and gliogenesis in the developing ventral spinal cord. We show that each of these factors has a unique ability to either enhance or inhibit the activities of the proneural helix-loop-helix (HLH) factors Ngn1 (Neurog1), Ngn2 (Neurog2), Ngn3 (Neurog3) and Mash1 (Ascl1), and the inhibitory HLH factors Id1 and Hes1, thereby regulating both the timing of differentiation of multipotent progenitors and their fate. Consistent with this, dynamic changes in their co-expression pattern in vivo are closely correlated to stage- and domain-specific generation of three neural cell lineages. We also show that genetic manipulations of their temporal expression patterns in mice alter the timing of differentiation of neurons and glia. We propose a molecular code model whereby the combinatorial actions of two classes of transcription factors coordinately regulate the domain-specific temporal sequence of neurogenesis and gliogenesis in the developing spinal cord.


Subject(s)
Helix-Loop-Helix Motifs , Multipotent Stem Cells/cytology , Neurons/cytology , Spinal Cord/embryology , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.2 , Mice , Mice, Mutant Strains , Multipotent Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Spinal Cord/cytology , Spinal Cord/metabolism , Tissue Culture Techniques
12.
Mol Cell Biol ; 27(11): 3982-94, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17371842

ABSTRACT

Precise control of proliferation and differentiation of multipotent neural stem cells (NSCs) is crucial for proper development of the nervous system. Although signaling through the cell surface receptor Notch has been implicated in many aspects of neural development, its role in NSCs remains elusive. Here we examined how the Notch pathway cross talks with signaling for growth factors and cytokines in controlling the self-renewal and differentiation of NSCs. Both Notch and growth factors were required for active proliferation of NSCs, but each of these signals was sufficient and independent of the other to inhibit differentiation of neurons and glia. Moreover, Notch signals could support the clonal self-renewing growth of NSCs in the absence of growth factors. This growth factor-independent action of Notch involved the regulation of the cell cycle and cell-cell interactions. During differentiation of NSCs, Notch signals promoted the generation of astrocytes in collaboration with ciliary neurotrophic factor and growth factors. Their cooperative actions were likely through synergistic phosphorylation of signal transducer and activator of transcription 3 on tyrosine at position 705 and serine at position 727. Our data suggest that distinct intracellular signaling pathways operate downstream of Notch for the self-renewal of NSCs and stimulation of astrogenesis.


Subject(s)
Cell Differentiation/physiology , Cytokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neurons/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Stem Cells/physiology , Animals , Cell Adhesion , Cell Cycle , Cell Proliferation , Cells, Cultured , Ciliary Neurotrophic Factor/metabolism , Female , Genes, Reporter , Neurons/cytology , Pregnancy , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism , Stem Cells/cytology
13.
Exp Neurol ; 203(2): 394-405, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17034791

ABSTRACT

The production of dopamine (DA) neurons from neural progenitor cells (NPC) is of particular interest as these neurons degenerate in Parkinson's disease. Here, we report that the characteristics of NPC from the ventral midbrain (NPC(VM)) and the striatum (NPC(STR)) are intrinsically determined. A detailed analysis of the VM during development revealed Ngn2 and Mash1 expression in a DA progenitor domain. Interestingly, over-expression of either Ngn2 or Mash1 induced neurogenesis from expanded NPC(VM). Whereas Ngn2 inhibited cell division and the production of neurons even in the presence of mitogens, Mash1 allowed the progenitors to divide while retaining neurogenic potential. However, none of the new neurons derived by over-expressing Ngn2 or Mash1 were positive for DA neuronal markers such as tyrosine hydroxylase. Nurr1 over-expression increased TH levels in a dose-dependant manner within both neurons and glia, suggesting a non-neuronal-specific activation of this enzyme by Nurr1. Double infection with Nurr1 and either Ngn2 or Mash1 resulted in the production of small numbers of TH+ neurons, which were larger in size when derived from NPC(VM) compared to NPC(STR). These data provide proof of concept that over-expression of multiple transcription factors can drive the fate of NPC first towards neurons, and then towards the DA phenotype. However, further factors may be required to generate fully functional DA neurons.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , DNA-Binding Proteins/physiology , Neurons/metabolism , Stem Cells/enzymology , Transcription Factors/physiology , Tyrosine 3-Monooxygenase/biosynthesis , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Division/physiology , Cell Proliferation , Cloning, Molecular , Culture Media , Female , Green Fluorescent Proteins/metabolism , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/pharmacology , Mesencephalon/cytology , Nuclear Receptor Subfamily 4, Group A, Member 2 , Pregnancy , Rats , Rats, Inbred Lew , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
14.
J Neurosci ; 26(46): 11948-60, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17108169

ABSTRACT

Neurons and oligodendrocytes are highly vulnerable to various insults, and their spontaneous replacement occurs to only a limited extent after damage in the adult spinal cord. The environment of injured tissue is thus thought to restrict the regenerative capacity of endogenous neural stem/progenitor cells; strategies for overcoming such restrictions remain to be developed. Here, we combined growth factor treatment and genetic manipulation to stimulate neurogenesis and oligodendrogenesis by endogenous progenitors in vivo. The recombinant retrovirus pMXIG, which was designed to coexpress green fluorescent proteins (GFPs) and a neurogenic/gliogenic transcription factor, was directly injected into the injured spinal cord parenchyma to manipulate proliferative cells in situ. We found that cells expressing Olig2, Nkx2.2, and NG2 were enriched among virus-infected, GFP-positive (GFP+) cells. Moreover, a fraction of GFP+ cells formed neurospheres and differentiated into neurons, astrocytes, and oligodendrocytes in vitro, demonstrating that GFP retroviruses indeed infected endogenous neural progenitors in vivo. Neuronal differentiation of control virus-infected cells did not occur at a detectable level in the injured spinal cord. We found, however, that direct administration of fibroblast growth factor 2 and epidermal growth factor into lesioned tissue could induce a significant fraction of GFP-labeled cells to express immature neuronal markers. Moreover, retrovirus-mediated overexpression of the basic helix-loop-helix transcription factors Neurogenin2 and Mash1, together with growth factor treatment, enhanced the production and maturation of new neurons and oligodendrocytes, respectively. These results demonstrate that endogenous neural progenitors can be manipulated to replace neurons and oligodendrocytes lost to insults in the injured spinal cord.


Subject(s)
Cell Differentiation/drug effects , Genetic Vectors/therapeutic use , Intercellular Signaling Peptides and Proteins/pharmacology , Spinal Cord Injuries/therapy , Spinal Cord/drug effects , Stem Cells/drug effects , Animals , Antigens/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Cells, Cultured , Disease Models, Animal , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Genetic Therapy/methods , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/therapeutic use , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/cytology , Oligodendroglia/metabolism , Proteoglycans/metabolism , Rats , Rats, Sprague-Dawley , Retroviridae/genetics , Spinal Cord/cytology , Spinal Cord/physiology , Spinal Cord Injuries/physiopathology , Stem Cells/cytology , Stem Cells/physiology , Transcription Factors/metabolism , Treatment Outcome , Zebrafish Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...