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










Publication year range
1.
Interv Neuroradiol ; : 15910199231169597, 2023 May 08.
Article in English | MEDLINE | ID: mdl-37157800

ABSTRACT

BACKGROUND: Several translational animal models have been described assessing intra-arterial (IA) treatments for malignant gliomas. We describe the first endovascular animal model that allows testing of IA drug delivery as a first-line treatment, which is difficult to do in actual patients. We report a unique protocol for vascular access and IA delivery in the rat model that, unlike prior reports, does not require direct puncture and opening of proximal cerebrovasculature which carries risk of ischemia in the animal brain post-delivery. METHODS: Wistar rats underwent left femoral artery catherization with a Balt Magic 1.2F catheter or Marathon Flow directed 1.5F Microcatheter with an Asahi Chikai 0.008 micro-guidewire which was navigated to the left internal carotid artery under x-ray. 25% mannitol was administered to test blood brain barrier breakdown (BBBB). Additional rats were implanted with C6 glioma cells in the left frontal lobe. C6 Glioma-Implanted Rats (C6GRs) were monitored for overall survival and tumor growth. Tumor volumes from MRI images were calculated utilizing 3D slicer. Additional rats underwent femoral artery catheterization with Bevacizumab, carboplatin, or irinotecan injected into the left internal carotid artery to test feasibility and safety. RESULTS: A successful endovascular access and BBBB protocol was established. BBBB was confirmed with positive Evans blue staining. 10 rats were successfully implanted with C6 gliomas with confirmed growths on MRI. Overall survival was 19.75 ± 2.21 days. 5 rats were utilized for the development of our femoral catheterization protocol and BBBB testing. With regards to IA chemotherapy dosage testing, control rats tolerated targeted 10 mg/kg of bevascizumab, 2.4 mg/kg of carboplatin, and 15 mg/kg of irinotecan IA ICA injections without any complications. CONCLUSIONS: We present the first endovascular IA rat glioma model that allows selective catheterization of the intracranial vasculature and assessment of IA therapies for gliomas without need for access and sacrifice of proximal cerebrovasculature.

2.
Nat Commun ; 12(1): 1923, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33772011

ABSTRACT

Chronic demyelination in the human CNS is characterized by an inhibitory microenvironment that impairs recruitment and differentiation of oligodendrocyte progenitor cells (OPCs) leading to failed remyelination and axonal atrophy. By network-based transcriptomics, we identified sulfatase 2 (Sulf2) mRNA in activated human primary OPCs. Sulf2, an extracellular endosulfatase, modulates the signaling microenvironment by editing the pattern of sulfation on heparan sulfate proteoglycans. We found that Sulf2 was increased in demyelinating lesions in multiple sclerosis and was actively secreted by human OPCs. In experimental demyelination, elevated OPC Sulf1/2 expression directly impaired progenitor recruitment and subsequent generation of oligodendrocytes thereby limiting remyelination. Sulf1/2 potentiates the inhibitory microenvironment by promoting BMP and WNT signaling in OPCs. Importantly, pharmacological sulfatase inhibition using PI-88 accelerated oligodendrocyte recruitment and remyelination by blocking OPC-expressed sulfatases. Our findings define an important inhibitory role of Sulf1/2 and highlight the potential for modulation of the heparanome in the treatment of chronic demyelinating disease.


Subject(s)
Cell Differentiation/genetics , Cellular Microenvironment/genetics , Demyelinating Diseases/genetics , Gene Expression Profiling/methods , Oligodendrocyte Precursor Cells/metabolism , Remyelination/genetics , Animals , Axons/metabolism , Cells, Cultured , Demyelinating Diseases/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Multiple Sclerosis/genetics , Multiple Sclerosis/metabolism , Oligodendrocyte Precursor Cells/cytology , Sulfatases/genetics , Sulfatases/metabolism , Sulfotransferases/genetics , Sulfotransferases/metabolism
3.
Cell Rep ; 25(12): 3435-3450.e6, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30566868

ABSTRACT

Human oligodendrocyte progenitor cells (hOPCs) persist into adulthood as an abundant precursor population capable of division and differentiation. The transcriptional mechanisms that regulate hOPC homeostasis remain poorly defined. Herein, we identify paired related homeobox protein 1 (PRRX1) in primary PDGFαR+ hOPCs. We show that enforced PRRX1 expression results in reversible G1/0 arrest. While both PRRX1 splice variants reduce hOPC proliferation, only PRRX1a abrogates migration. hOPC engraftment into hypomyelinated shiverer/rag2 mouse brain is severely impaired by PRRX1a, characterized by reduced cell proliferation and migration. PRRX1 induces a gene expression signature characteristic of stem cell quiescence. Both IFN-γ and BMP signaling upregulate PRRX1 and induce quiescence. PRRX1 knockdown modulates IFN-γ-induced quiescence. In mouse brain, PRRX1 mRNA was detected in non-dividing OPCs and is upregulated in OPCs following demyelination. Together, these data identify PRRX1 as a regulator of quiescence in hOPCs and as a potential regulator of pathological quiescence.


Subject(s)
Cell Cycle , Homeodomain Proteins/metabolism , Oligodendrocyte Precursor Cells/cytology , Oligodendrocyte Precursor Cells/metabolism , Animals , Bone Morphogenetic Proteins/pharmacology , Cell Cycle/drug effects , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Gene Expression Regulation/drug effects , Humans , Interferon-gamma/pharmacology , Ki-67 Antigen/metabolism , Mice , Myelin Sheath/metabolism , Oligodendrocyte Precursor Cells/transplantation , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation/drug effects , White Matter/metabolism , White Matter/pathology
4.
Stem Cell Reports ; 9(2): 710-723, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28793249

ABSTRACT

Impaired human oligodendrocyte progenitor cell (hOPC) differentiation likely contributes to failed remyelination in multiple sclerosis. The characterization of molecular pathways that regulate hOPC differentiation will provide means to induce remyelination. In this study, we determined the gene expression profile of PDGFαR+ hOPCs during initial oligodendrocyte commitment. Weighted gene coexpression network analysis was used to define progenitor and differentiation-specific gene expression modules and functionally important hub genes. These modules were compared with rodent OPC and oligodendrocyte data to determine the extent of species conservation. These analyses identified G-protein ß4 (GNB4), which was associated with hOPC commitment. Lentiviral GNB4 overexpression rapidly induced human oligodendrocyte differentiation. Following xenograft in hypomyelinating shiverer/rag2 mice, GNB4 overexpression augmented myelin synthesis and the ability of hOPCs to ensheath host axons, establishing GNB4 as functionally important in human myelination. As such, network analysis of hOPC gene expression accurately predicts genes that influence human oligodendrocyte differentiation in vivo.


Subject(s)
Cell Differentiation/genetics , Computational Biology/methods , Gene Expression Regulation , Gene Regulatory Networks , Genomics , Oligodendrocyte Precursor Cells/cytology , Oligodendrocyte Precursor Cells/metabolism , Animals , Axons/metabolism , GTP-Binding Protein beta Subunits/genetics , Gene Expression Profiling , Genomics/methods , Humans , Oligodendroglia/cytology , Oligodendroglia/metabolism , Receptors, G-Protein-Coupled/metabolism , Rodentia , Signal Transduction , Transcriptome
5.
Exp Neurol ; 283(Pt B): 489-500, 2016 09.
Article in English | MEDLINE | ID: mdl-27001544

ABSTRACT

Oligodendrocyte development has been studied for several decades, and has served as a model system for both neurodevelopmental and stem/progenitor cell biology. Until recently, the vast majority of studies have been conducted in lower species, especially those focused on rodent development and remyelination. In humans, the process of myelination requires the generation of vastly more myelinating glia, occurring over a period of years rather than weeks. Furthermore, as evidenced by the presence of chronic demyelination in a variety of human neurologic diseases, it appears likely that the mechanisms that regulate development and become dysfunctional in disease may be, in key ways, divergent across species. Improvements in isolation techniques, applied to primary human neural and oligodendrocyte progenitors from both fetal and adult brain, as well as advancements in the derivation of defined progenitors from human pluripotent stem cells, have begun to reveal the extent of both species-conserved signaling pathways and potential key differences at cellular and molecular levels. In this article, we will review the commonalities and differences in myelin development between rodents and man, describing the approaches used to study human oligodendrocyte differentiation and myelination, as well as heterogeneity within targetable progenitor pools, and discuss the advances made in determining which conserved pathways may be both modeled in rodents and translate into viable therapeutic strategies to promote myelin repair.


Subject(s)
Cell Transplantation/methods , Demyelinating Diseases/surgery , Oligodendroglia/physiology , Stem Cells/physiology , Animals , Cell Differentiation , Humans , Regeneration/physiology
6.
J Neurosci ; 35(8): 3676-88, 2015 Feb 25.
Article in English | MEDLINE | ID: mdl-25716865

ABSTRACT

Therapeutic repair of myelin disorders may be limited by the relatively slow rate of human oligodendrocyte differentiation. To identify appropriate pharmacological targets with which to accelerate differentiation of human oligodendrocyte progenitors (hOPCs) directly, we used CD140a/O4-based FACS of human forebrain and microarray to hOPC-specific receptors. Among these, we identified CHRM3, a M3R muscarinic acetylcholine receptor, as being restricted to oligodendrocyte-biased CD140a(+)O4(+) cells. Muscarinic agonist treatment of hOPCs resulted in a specific and dose-dependent blockade of oligodendrocyte commitment. Conversely, when hOPCs were cocultured with human neurons, M3R antagonist treatment stimulated oligodendrocytic differentiation. Systemic treatment with solifenacin, an FDA-approved muscarinic receptor antagonist, increased oligodendrocyte differentiation of transplanted hOPCs in hypomyelinated shiverer/rag2 brain. Importantly, solifenacin treatment of engrafted animals reduced auditory brainstem response interpeak latency, indicative of increased conduction velocity and thereby enhanced functional repair. Therefore, solifenacin and other selective muscarinic antagonists represent new adjunct approaches to accelerate repair by engrafted human progenitors.


Subject(s)
Fetal Stem Cells/cytology , Muscarinic Antagonists/pharmacology , Myelin Sheath/metabolism , Oligodendroglia/cytology , Quinuclidines/pharmacology , Regeneration , Tetrahydroisoquinolines/pharmacology , Animals , Brain Stem/cytology , Brain Stem/physiology , Cells, Cultured , DNA-Binding Proteins/genetics , Evoked Potentials, Auditory, Brain Stem , Female , Fetal Stem Cells/drug effects , Fetal Stem Cells/metabolism , Fetal Stem Cells/transplantation , Humans , Male , Mice , Muscarinic Agonists/pharmacology , Myelin Sheath/genetics , Neurogenesis , O Antigens/genetics , O Antigens/metabolism , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Oligodendroglia/transplantation , Prosencephalon/cytology , Prosencephalon/embryology , Receptor, Muscarinic M3 , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptors, Muscarinic/genetics , Receptors, Muscarinic/metabolism , Solifenacin Succinate
7.
Proc Natl Acad Sci U S A ; 111(28): E2885-94, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24982138

ABSTRACT

Human oligodendrocyte progenitor cell (OPC) specification and differentiation occurs slowly and limits the potential for cell-based treatment of demyelinating disease. In this study, using FACS-based isolation and microarray analysis, we identified a set of transcription factors expressed by human primary CD140a(+)O4(+) OPCs relative to CD133(+)CD140a(-) neural stem/progenitor cells (NPCs). Among these, lentiviral overexpression of transcription factors ASCL1, SOX10, and NKX2.2 in NPCs was sufficient to induce Sox10 enhancer activity, OPC mRNA, and protein expression consistent with OPC fate; however, unlike ASCL1 and NKX2.2, only the transcriptome of SOX10-infected NPCs was induced to a human OPC gene expression signature. Furthermore, only SOX10 promoted oligodendrocyte commitment, and did so at quantitatively equivalent levels to native OPCs. In xenografts of shiverer/rag2 animals, SOX10 increased the rate of mature oligodendrocyte differentiation and axon ensheathment. Thus, SOX10 appears to be the principle and rate-limiting regulator of myelinogenic fate from human NPCs.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Enhancer Elements, Genetic , Heterografts , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Humans , Lentivirus , Mice , Neural Stem Cells/cytology , Nuclear Proteins , Oligodendroglia/cytology , Stem Cell Transplantation , Transcription Factors/genetics , Transcriptome , Transduction, Genetic
8.
Exp Neurol ; 247: 694-702, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23507034

ABSTRACT

In this study, we sought to establish a novel method to prospectively and dynamically identify live human oligodendrocyte precursor cells (OPCs) and oligodendrocyte lineage cells from brain dissociates and pluripotent stem cell culture. We selected a highly conserved enhancer element of the Sox10 gene, known as MCS5, which directs reporter expression to oligodendrocyte lineage cells in mouse and zebrafish. We demonstrate that lentiviral Sox10-MCS5 induced expression of GFP at high levels in a subpopulation of human CD140a/PDGFαR-sorted OPCs as well as their immature oligodendrocyte progeny. Furthermore, we show that almost all Sox10-MCS5:GFP(high) cells expressed OPC antigen CD140a and human OPCs expressing SOX10, OLIG2, and PDGFRA mRNAs could be prospectively identified using GFP based fluorescence activated cells sorting alone. Additionally, we established a human induced pluripotent cell (iPSC) line transduced with the Sox10-MCS5:GFP reporter using a Rex-Neo cassette. Similar to human primary cells, GFP expression was restricted to embryoid bodies containing both oligodendrocyte progenitor and oligodendrocyte cells and co-localized with NG2 and O4-positive cells respectively. As such, we have developed a novel reporter system that can track oligodendrocyte commitment in human cells, establishing a valuable tool to improve our understanding and efficiency of human oligodendrocyte derivation.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/physiology , Enhancer Elements, Genetic/genetics , Oligodendroglia/metabolism , SOXE Transcription Factors/metabolism , Antigens/metabolism , Cells, Cultured , Fetus , Flow Cytometry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , O Antigens/metabolism , Proteoglycans/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , SOXE Transcription Factors/genetics
9.
Stem Cells Dev ; 22(15): 2121-31, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23488628

ABSTRACT

The mechanisms underlying the specification of oligodendrocyte fate from multipotent neural progenitor cells (NPCs) in developing human brain are unknown. In this study, we sought to identify antigens sufficient to distinguish NPCs free from oligodendrocyte progenitor cells (OPCs). We investigated the potential overlap of NPC and OPC antigens using multicolor fluorescence-activated cell sorting (FACS) for CD133/PROM1, A2B5, and CD140a/PDGFαR antigens. Surprisingly, we found that CD133, but not A2B5, was capable of enriching for OLIG2 expression, Sox10 enhancer activity, and oligodendrocyte potential. As a subpopulation of CD133-positive cells expressed CD140a, we asked whether CD133 enriched bone fide NPCs regardless of CD140a expression. We found that CD133(+)CD140a(-) cells were highly enriched for neurosphere initiating cells and were multipotent. Importantly, when analyzed immediately following isolation, CD133(+)CD140a(-) NPCs lacked the capacity to generate oligodendrocytes. In contrast, CD133(+)CD140a(+) cells were OLIG2-expressing OPCs capable of oligodendrocyte differentiation, but formed neurospheres with lower efficiency and were largely restricted to glial fate. Gene expression analysis further confirmed the stem cell nature of CD133(+)CD140a(-) cells. As human CD133(+) cells comprised both NPCs and OPCs, CD133 expression alone cannot be considered a specific marker of the stem cell phenotype, but rather comprises a heterogeneous mix of glial restricted as well as multipotent neural precursors. In contrast, CD133/CD140a-based FACS permits the separation of defined progenitor populations and the study of neural stem and oligodendrocyte fate specification in the human brain.


Subject(s)
Antigens, CD/metabolism , Glycoproteins/metabolism , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Peptides/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , AC133 Antigen , Biomarkers/metabolism , Cell Separation , Cells, Cultured , Flow Cytometry , Humans , Oligonucleotide Array Sequence Analysis , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Transcriptome
10.
Glia ; 60(12): 1944-53, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22927334

ABSTRACT

The molecular mechanisms controlling human oligodendrocyte development are poorly characterized. Microarray analysis of human oligodendrocyte progenitor cells (OPCs) and immature oligodendrocytes revealed that specific-class I histone deacetylase (HDAC) target genes were actively repressed during oligodendrocyte commitment. Although epigenetic regulation of oligodendrocyte differentiation has been established in rodent development, the role of HDACs in human OPCs remains undefined. We used HDAC inhibitors (HDACi) trichostatin A (TSA) and sodium butyrate to determine the importance of HDAC activity in human primary OPC differentiation. Treatment with either drug resulted in significant dose-dependent inhibition of O4(+) oligodendrocyte cell differentiation, reduction of oligodendrocyte morphological maturation, and downregulation of myelin basic protein mRNA. High dose TSA treatment was also associated with reduction in OPC proliferation. HDACi treatment prevented downregulation of SOX2, ID4, and TCF7L2 mRNAs but did not regulate HES5, suggesting that targets of HDAC repression may differ between species. These results predict that HDACi treatment would impair proliferation and differentiation by parenchymal oligodendrocyte progenitors, and thereby degrade their potential for endogenous repair in human demyelinating disease. © 2012 Wiley Periodicals, Inc.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/enzymology , Histone Deacetylase Inhibitors , Histone Deacetylases/metabolism , Oligodendroglia/enzymology , Cell Differentiation/drug effects , Cells, Cultured , Down-Regulation/drug effects , Embryonic Stem Cells/drug effects , Enzyme Activation/drug effects , Enzyme Activation/physiology , Fetus/cytology , Fetus/drug effects , Fetus/enzymology , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/physiology , Humans , Oligodendroglia/drug effects , Oligonucleotide Array Sequence Analysis/methods , Stem Cells/drug effects , Stem Cells/enzymology
11.
Plant Signal Behav ; 3(6): 398-400, 2008 Jun.
Article in English | MEDLINE | ID: mdl-19704578

ABSTRACT

Development of sessile organisms requires adaptation to an ever-changing environment. In order to respond quickly to these challenges, complex signaling mechanisms have evolved to facilitate cellular modifications. The importance of phospholipid-based signaling pathways in plants, as well as animals, has recently been gaining attention. Both the PLD and PLC pathways produce the signaling molecule PA, which modulates MTs, F-actin and endomembrane trafficking. We have examined the roles of the PLD signaling pathway during development of the marine brown alga Silvetia compressa. Zygotes were treated with 1- and 2-butanol, both of which activate the PLD enzyme. However, only 1-butanol competes with water as a transphosphatidylation substrate, at the expense of PA production. Interestingly, we found that 1- and 2-butanol both disrupted MT organization and thereby cell division, with 1-butanol being more potent. These findings question whether the effects of butyl alcohol treatment are due to lowered PA levels or activation of the PLD enzyme. Additionally, preliminary results show that inhibition of DAGK results in loss of centrosomal MTs and formation of cortical MT cages that are strikingly similar to those formed following 1-butanol treatment. These data suggest that perturbation of the PLD or PLC pathway leads to cortical stabilization and/or nucleation of MT arrays.

SELECTION OF CITATIONS
SEARCH DETAIL
...