UB Vice President for Research and Economic Development markup
Fraser J. Sim UB Profile
Pharmacology and Toxicology
Associate Professor
Neuroscience Program Director
Specialty/Research Focus: Genomics and proteomics; Neurobiology; Neurodegenerative disorders
955 Main Street
Room 3256
Buffalo NY 14203
716-829-2151
Export
Publication Types
Co-Authors
  • More Coauthors +
 
Authors: Everett J Sinibaldi, Fraser J Sim, Jennifer K Lang
Journal-article: Circulation Research, vol. 125, 2019
Authors: Suyog U Pol, Jessie J Polanco, Richard A Seidman, Melanie A O'Bara, Hani J Shayya, Karen C Dietz, Fraser J Sim
Keywords: oligodendrocyte progenitor species conservation
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 PDGFR+ 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 4 (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 invivo.
PubMedId: 28793249
Journal-article: Stem cell reports, vol. 9, 2017
Authors: Jennifer A Martin, Aaron Caccamise, Craig T Werner, Rathipriya Viswanathan, Jessie J Polanco, Andrew F Stewart, Shruthi A Thomas, Fraser J Sim, David M Dietz
Abstract: Opiate abuse and addiction have become a worldwide epidemic with great societal and financial burdens, highlighting a critical need to understand the neurobiology of opiate addiction. While several studies have focused on drug-dependent changes in neurons, the role of glia in opiate addiction remains largely unstudied. RNA sequencing pathway analysis from the prefrontal cortex of male rats revealed changes in several genes associated with oligodendrocyte differentiation and maturation following heroin self-administration. Among these genes changed was Sox10, which is regulated, in part, by the chromatin remodeler BRG1/SMARCA4. To directly test the functional role of Sox10 in mediating heroin-induced behavioral plasticity, we selectively overexpressed Sox10 and BRG1 in the prefrontal cortex. Overexpression of either Sox10 or BRG1 decreased the motivation to obtain heroin infusions in a progressive ratio test without altering the acquisition or maintenance of heroin self-administration. These data demonstrate a critical, and perhaps compensatory, role of Sox10 and BRG1 in oligodendrocytes in regulating the motivation for heroin.Neuropsychopharmacology accepted article preview online, 20 December 2017. doi:10.1038/npp.2017.303.
PubMedId: 29260792
Journal-article: Neuropsychopharmacology, 2017
Authors: Zi-Jun Wang, Jennifer A Martin, Amy M Gancarz, Danielle N Adank, Fraser J Sim, David M Dietz
Abstract: Drug addiction is a long-lasting disease characterized by compulsive drug intake mediated in part by neuronal and biological adaptations in key brain areas, such as the nucleus accumbens (NAc). While we previously demonstrated involvement of the activin 2a receptor in drug taking, the role of its ligand, activin A, in cocaine relapse is unknown. Activin A levels in the NAc were assessed via ELISA and immunohistochemistry (in neurons, astrocytes, and microglia) following a cocaine binge paradigm. Cocaine exposure significantly increased the levels of activin A in the NAc of animals that had self-administered cocaine prior to the 14-day withdrawal compared with levels in saline controls. This was accompanied by an increase in the proportion of IBA1+ microglia in the NAc that were immunopositive for activin A. In contrast, the proportions of NeuN+ neurons and GFAP+ astrocytes that were immunopositive for activin A remained unaltered. In conclusion, these data suggest that increased secretion of activin A, particularly from microglia, in the NAc represents a novel potential target for the treatment of cocaine relapse.
PubMedId: 28272550
Journal-article: Scientific reports, vol. 7, 2016
Authors: Kavitha Abiraman, Suyog U Pol, Melanie A O'Bara, Guang-Di Chen, Zainab M Khaku, Jing Wang, David Thorn, Bansi H Vedia, Ezinne C Ekwegbalu, Jun-Xu Li, Richard J Salvi, Fraser J Sim
Keywords: glia human microarray oligodendrocyte
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.
PubMedId: 25716865
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 35, 2015
Authors: Yannick Poitelon, Camila Lopez-Anido, Kathleen Catignas, Caterina Berti, Marilena Palmisano, Courtney Williamson, Dominique Ameroso, Kansho Abiko, Yoonchan Hwang, Alex Gregorieff, Jeffrey L Wrana, Mohammadnabi Asmani, Ruogang Zhao, Fraser James Sim, Lawrence Wrabetz, John Svaren, Maria Laura Feltri
Abstract: Myelination is essential for nervous system function. Schwann cells interact with neurons and the basal lamina to myelinate axons using known receptors, signals and transcription factors. In contrast, the transcriptional control of axonal sorting and the role of mechanotransduction in myelination are largely unknown. Yap and Taz are effectors of the Hippo pathway that integrate chemical and mechanical signals in cells. We describe a previously unknown role for the Hippo pathway in myelination. Using conditional mutagenesis in mice, we show that Taz is required in Schwann cells for radial sorting and myelination and that Yap is redundant with Taz. Yap and Taz are activated in Schwann cells by mechanical stimuli and regulate Schwann cell proliferation and transcription of basal lamina receptor genes, both necessary for radial sorting of axons and subsequent myelination. These data link transcriptional effectors of the Hippo pathway and of mechanotransduction to myelin formation in Schwann cells.
PubMedId: 27273766
Journal-article: Nature neuroscience, vol. 19, 2015
Authors: Karen C Dietz, Jessie J Polanco, Suyog U Pol, Fraser J Sim
Keywords: Human cell implantation Oligodendrocyte progenitor Remyelination
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.
PubMedId: 27001544
Journal-article: Experimental neurology, vol. 283, 2015
Authors: Bo Zhou, Jason M. Osinski, Juan L. Mateo, Ben Martynoga, Fraser J. Sim, Christine E. Campbell, Francois Guillemot, Michael Piper, Richard M. Gronostajski
Abstract: Murine postnatal neural stem cells (NSCs) give rise to neurons, astrocytes, or oligodendrocytes (OLs); however, our knowledge of the genes that control this lineage specification is incomplete. In this study, we show that nuclear factor I X (NFIX), a transcription factor known to regulate NSC quiescence, also suppresses oligodendrogenesis (ODG) from NSCs. Immunostaining reveals little or no expression of NFIX in OL lineage cells both in vivo and in vitro. Loss of NFIX from subventricular zone (SVZ) NSCs results in enhanced ODG both in vivo and in vitro, while forced expression of NFIX blocks NSC differentiation into OLs in vitro. RNA-seq analysis shows that genes previously shown to be differentially expressed in OL progenitors are significantly enriched in RNA from Nfix(-/-) versus wild-type NSCs. These data indicate that NFIX influences the lineage specification of postnatal SVZ NSCs, specifically suppressing ODG.
PubMedId: 26083238
Journal-article: Stem Cells and Development, vol. 24, pp. 2114-2126, 2015
Authors: Panagiotis Douvaras, Jing Wang, Matthew Zimmer, Stephanie Hanchuk, Melanie A O'Bara, Saud Sadiq, Fraser J Sim, James Goldman, Valentina Fossati
Abstract: Multiple sclerosis (MS) is a chronic demyelinating disease of unknown etiology that affects the CNS. While current therapies are primarily directed against the immune system, the new challenge is to address progressive MS with remyelinating and neuroprotective strategies. Here, we develop a highly reproducible protocol to efficiently derive oligodendrocyte progenitor cells (OPCs) and mature oligodendrocytes from induced pluripotent stem cells (iPSCs). Key elements of our protocol include adherent cultures, dual SMAD inhibition, and addition of retinoids from the beginning of differentiation, which lead to increased yields of OLIG2 progenitors and high numbers of OPCs within 75 days. Furthermore, we show the generation of viral and integration-free iPSCs from primary progressive MS (PPMS) patients and their efficient differentiation to oligodendrocytes. PPMS OPCs are functional, as demonstrated by in vivo myelination in the shiverer mouse. These results provide encouraging advances toward the development of autologous cell therapies using iPSCs.
PubMedId: 25254339
Journal-article: Stem cell reports, vol. 3, 2014
Authors: Jing Wang, Suyog U Pol, Alexa K Haberman, Chunming Wang, Melanie A O'Bara, Fraser J Sim
Keywords: neural precursor cell reprogramming transplantation
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.
PubMedId: 24982138
Journal-article: Proceedings of the National Academy of Sciences of the United States of America, vol. 111, 2014
Authors: Jing Wang, Melanie A O'Bara, Suyog U Pol, Fraser J Sim
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/PDGFR 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.
PubMedId: 23488628
Journal-article: Stem cells and development, vol. 22, 2013
Authors: Jennifer M Dolan, Hui Meng, Fraser J Sim, John Kolega
Keywords: high wall shear stress intracranial aneurysm initiation microarray spatial gradient vascular remodeling
Abstract: Flow impingement at arterial bifurcations causes high frictional force [or wall shear stress (WSS)], and flow acceleration and deceleration in the branches create positive and negative streamwise gradients in WSS (WSSG), respectively. Intracranial aneurysms tend to form in regions with high WSS and positive WSSG. However, little is known about the responses of endothelial cells (ECs) to either positive or negative WSSG under high WSS conditions. We used cDNA microarrays to profile gene expression in cultured ECs exposed to positive or negative WSSG for 24 h in a flow chamber where WSS varied between 3.5 and 28.4 Pa. Gene ontology and biological pathway analysis indicated that positive WSSG favored proliferation, apoptosis, and extracellular matrix processing while decreasing expression of proinflammatory genes. To determine if similar responses occur in vivo, we examined EC proliferation and expression of the matrix metalloproteinase ADAMTS1 under high WSS and WSSG created at the basilar terminus of rabbits after bilateral carotid ligation. Precise hemodynamic conditions were determined by computational fluid dynamic simulations from three-dimensional angiography and mapped on immunofluorescence staining for the proliferation marker Ki-67 and ADAMTS1. Both proliferation and ADAMTS1 were significantly higher in ECs under positive WSSG than in adjacent regions of negative WSSG. Our results indicate that WSSG elicits distinct EC gene expression profiles and particular biological pathways including increased cell proliferation and matrix processing. Such EC responses may be important in understanding the mechanisms of intracranial aneurysm initiation at regions of high WSS and positive WSSG.
PubMedId: 23885059
Journal-article: American journal of physiology. Cell physiology, vol. 305, 2013
Authors: Marianna Bugiani, Nienke Postma, Emiel Polder, Nikki Dieleman, Peter G. Scheffer, Fraser J. Sim, Marjo S. van der Knaap, Ilja Boor
Journal-article: Tijdschrift voor Kindergeneeskunde, vol. 81, pp. 28-28, 2013
Authors: Romane??M. Auvergne, Fraser??J. Sim, Su Wang, Devin Chandler-Militello, Jaclyn Burch, Yazan Al??Fanek, Danielle Davis, Abdellatif Benraiss, Kevin Walter, Pragathi Achanta, Mahlon Johnson, Alfredo Quinones-Hinojosa, Sridaran Natesan, Heide??L. Ford, Steven??A. Goldman
Journal-article: Cell Reports, vol. 4, pp. 402, 2013
Authors: Marianna Bugiani, Nienke Postma, Emiel Polder, Nikki Dieleman, Peter G Scheffer, Fraser J Sim, Marjo S van der Knaap, Ilja Boor
Abstract: Vanishing white matter disease is a genetic leukoencephalopathy caused by mutations in eukaryotic translation initiation factor 2B. Patients experience a slowly progressive neurological deterioration with episodes of rapid clinical worsening triggered by stress. The disease may occur at any age and leads to early death. Characteristic neuropathological findings include cystic degeneration of the white matter with feeble, if any, reactive gliosis, dysmorphic astrocytes and paucity of myelin despite an increase in oligodendrocytic density. These features have been linked to a maturation defect of astrocytes and oligodendrocytes. However, the nature of the link between glial immaturity and the observed neuropathological features is unclear. We hypothesized that the defects in maturation and function of astrocytes and oligodendrocytes are related. Brain tissue of seven patients with genetically proven vanishing white matter disease was investigated using immunohistochemistry, western blotting, quantitative polymerase chain reaction and size exclusion chromatography. The results were compared with those obtained from normal brain tissue of age-matched controls, from chronic demyelinated multiple sclerosis lesions and from other genetic and acquired white matter disorders. We found that the white matter of patients with vanishing white matter disease is enriched in CD44-expressing astrocyte precursor cells and accumulates the glycosaminoglycan hyaluronan. Hyaluronan is a major component of the extracellular matrix, and CD44 is a hyaluronan receptor. We found that a high molecular weight form of hyaluronan is overabundant, especially in the most severely affected areas. Comparison between the more severely affected frontal white matter and the relatively spared cerebellum confirms that high molecular weight hyaluronan accumulation is more pronounced in the frontal white matter than in the cerebellum. High molecular weight hyaluronan is known to inhibit astrocyte and oligodendrocyte precursor maturation and can explain the arrested glial progenitor maturation observed in vanishing white matter disease. In conclusion, high molecular weight species of hyaluronan accumulate in the white matter of patients with vanishing white matter disease, and by inhibiting glial maturation and proper function, they may be a major determinant of the white matter pathology and lack of repair.
PubMedId: 23365098
Journal-article: Brain : a journal of neurology, vol. 136, 2013
Authors: Suyog U Pol, Jennifer K Lang, Melanie A O'Bara, Thomas R Cimato, Andrew S McCallion, Fraser J Sim
Keywords: Enhancer Lentivirus Pluripotent SOX10
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/PDGFR-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.
PubMedId: 23507034
Journal-article: Experimental neurology, vol. 247, 2012
Authors: Romane M Auvergne, Fraser J Sim, Su Wang, Devin Chandler-Militello, Jaclyn Burch, Yazan Al Fanek, Danielle Davis, Abdellatif Benraiss, Kevin Walter, Pragathi Achanta, Mahlon Johnson, Alfredo Quinones-Hinojosa, Sridaran Natesan, Heide L Ford, Steven A Goldman
Abstract: Glial progenitor cells (GPCs) are a potential source of malignant gliomas. We used A2B5-based sorting to extract tumorigenic GPCs from human gliomas spanning World Health Organization grades II-IV. Messenger RNA profiling identified a cohort of genes that distinguished A2B5+ glioma tumor progenitor cells (TPCs) from A2B5+ GPCs isolated from normal white matter. A core set of genes and pathways was substantially dysregulated in A2B5+ TPCs, which included the transcription factor SIX1 and its principal cofactors, EYA1 and DACH2. Small hairpin RNAi silencing of SIX1 inhibited the expansion of glioma TPCs in vitro and in vivo, suggesting a critical and unrecognized role of the SIX1-EYA1-DACH2 system in glioma genesis or progression. By comparing the expression patterns of glioma TPCs with those of normal GPCs, we have identified a discrete set of pathways by which glial tumorigenesis may be better understood and more specifically targeted.
PubMedId: 23727239
Journal-article: Cell reports, vol. 3, 2012
Authors: Fraser J. Sim, Steven A. Goldman
Book-chapter: Neuroglia, pp. 358-374, 2012
Authors: Jennifer M Dolan, Fraser J Sim, Hui Meng, John Kolega
Abstract: Chronic high flow can induce arterial remodeling, and this effect is mediated by endothelial cells (ECs) responding to wall shear stress (WSS). To assess how WSS above physiological normal levels affects ECs, we used DNA microarrays to profile EC gene expression under various flow conditions. Cultured bovine aortic ECs were exposed to no-flow (0 Pa), normal WSS (2 Pa), and very high WSS (10 Pa) for 24 h. Very high WSS induced a distinct expression profile compared with both no-flow and normal WSS. Gene ontology and biological pathway analysis revealed that high WSS modulated gene expression in ways that promote an anti-coagulant, anti-inflammatory, proliferative, and promatrix remodeling phenotype. A subset of characteristic genes was validated using quantitative polymerase chain reaction: very high WSS upregulated ADAMTS1 (a disintegrin and metalloproteinase with thrombospondin motif-1), PLAU (urokinase plasminogen activator), PLAT (tissue plasminogen activator), and TIMP3, all of which are involved in extracellular matrix processing, with PLAT and PLAU also contributing to fibrinolysis. Downregulated genes included CXCL5 and IL-8 and the adhesive glycoprotein THBS1 (thrombospondin-1). Expressions of ADAMTS1 and uPA proteins were assessed by immunhistochemistry in rabbit basilar arteries experiencing increased flow after bilateral carotid artery ligation. Both proteins were significantly increased when WSS was elevated compared with sham control animals. Our results indicate that very high WSS elicits a unique transcriptional profile in ECs that favors particular cell functions and pathways that are important in vessel homeostasis under increased flow. In addition, we identify specific molecular targets that are likely to contribute to adaptive remodeling under elevated flow conditions.
PubMedId: 22173868
Journal-article: American journal of physiology. Cell physiology, vol. 302, 2011
Authors: Fraser J Sim, Crystal R McClain, Steven J Schanz, Tricia L Protack, Martha S Windrem, Steven A Goldman
Abstract: Experimental animals with myelin disorders can be treated by transplanting oligodendrocyte progenitor cells (OPCs) into the affected brain or spinal cord. OPCs have been isolated by their expression of gangliosides recognized by mAb A2B5, but this marker also identifies lineage-restricted astrocytes and immature neurons. To establish a more efficient means of isolating myelinogenic OPCs, we sorted fetal human forebrain cells for CD140a, an epitope of platelet derived growth factor receptor (PDGFR), which is differentially expressed by OPCs. CD140a(+) cells were isolated as mitotic bipotential progenitors that initially expressed neither mature neuronal nor astrocytic phenotypic markers, yet could be instructed to either oligodendrocyte or astrocyte fate in vitro. Transplanted CD140a(+) cells were highly migratory and robustly myelinated the hypomyelinated shiverer mouse brain more rapidly and efficiently than did A2B5(+)cells. Microarray analysis of CD140a(+) cells revealed overexpression of the oligodendroglial marker CD9, suggesting that CD9(+)/CD140a(+) cells may constitute an even more highly enriched population of myelinogenic progenitor cells.
PubMedId: 21947029
Journal-article: Nature biotechnology, vol. 29, 2011
Authors: Gregory D Conway, Melanie A O'Bara, Bansi H Vedia, Suyog U Pol, Fraser J Sim
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.
PubMedId: 22927334
Journal-article: Glia, vol. 60, 2011
Authors: Steven A. Goldman, Fraser J. Sim
PubMedId: 21989674
Journal-article: Neurotherapeutics, vol. 8, pp. 537-538, 2011
Authors: Mahlon D. Johnson, Aubie K. Shaw, Mary J. O???Connell, Fraser J. Sim, Harold L. Moses
Journal-article: Journal of Neuro-Oncology, vol. 103, pp. 277-285, 2010
Authors: Su Wang, Devin Chandler-Militello, Gang Lu, Neeta S Roy, Alex Zielke, Romane Auvergne, Nancy Stanwood, Daniel Geschwind, Giovanni Coppola, Silvia K Nicolis, Fraser J Sim, Steven A Goldman
Abstract: Sox2 is expressed by neural stem and progenitor cells, and a sox2 enhancer identifies these cells in the forebrains of both fetal and adult transgenic mouse reporters. We found that an adenovirus encoding EGFP placed under the regulatory control of a 0.4 kb sox2 core enhancer selectively identified multipotential and self-renewing neural progenitor cells in dissociates of human fetal forebrain. Upon EGFP-based fluorescence-activated cell sorting (FACS), the E/sox2:EGFP(+) isolates were propagable for up to 1 year in vitro, and remained multilineage competent throughout. E/sox2:EGFP(+) cells expressed more telomerase enzymatic activity than matched E/sox2:EGFP-depleted populations, and maintained their telomeric lengths with successive passage. Gene expression analysis of E/sox2:EGFP-sorted neural progenitor cells, normalized to the unsorted forebrain dissociates from which they derived, revealed marked overexpression of genes within the notch and wnt pathways, and identified multiple elements of each pathway that appear selective to human neural progenitors. Sox2 enhancer-based FACS thus permits the prospective identification and direct isolation of a telomerase-active population of neural stem cells from the human fetal forebrain, and the elucidation of both the transcriptome and dominant signaling pathways of these critically important cells.
PubMedId: 21048121
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 30, 2010
Authors: Fraser J Sim, Martha S Windrem, Steven A Goldman
Abstract: Glial progenitor cells (GPCs) comprise the most abundant population of progenitor cells in the adult human brain. They are responsible for central nervous system (CNS) remyelination, and likely contribute to the astrogliotic response to brain injury and degeneration as well. Adult human GPCs are biased to differentiate as oligodendrocytes and elaborate new myelin, and yet they retain multilineage plasticity, and can give rise to neurons as well as astrocytes and oligodendrocytes once removed from the adult parenchymal environment. GPCs retain strong mechanisms for cell-autonomous self-renewal, and yet both their phenotype and fate may be dictated by their microenvironment. Using the transcriptional profiles of acutely isolated GPCs, we have begun to understand the operative ligand-receptor interactions involved in these processes, and have identified several key signaling pathways by which adult human GPCs may be reliably instructed to either oligodendrocytic or astrocytic fate. In addition, we have noted significant differences between the expressed genes and dominant signaling pathways of fetal and adult human GPCs, as well as between rodent and human GPCs. The latter data in particular call into question therapeutic strategies predicated solely upon data obtained using rodents, while perhaps highlighting the extent to which evolution has been attended by the phylogenetic modification of glial phenotype and function.
PubMedId: 19807941
Journal-article: Neuron glia biology, vol. 5, 2009
Authors: Anders I Persson, Claudia Petritsch, Fredrik J Swartling, Melissa Itsara, Fraser J Sim, Romane Auvergne, David D Goldenberg, Scott R Vandenberg, Kim N Nguyen, Stanislava Yakovenko, Jennifer Ayers-Ringler, Akiko Nishiyama, William B Stallcup, Mitchel S Berger, Gabriele Bergers, Tracy R McKnight, Steven A Goldman, William A Weiss
Abstract: Malignant astrocytic brain tumors are among the most lethal cancers. Quiescent and therapy-resistant neural stem cell (NSC)-like cells in astrocytomas are likely to contribute to poor outcome. Malignant oligodendroglial brain tumors, in contrast, are therapy sensitive. Using magnetic resonance imaging (MRI) and detailed developmental analyses, we demonstrated that murine oligodendroglioma cells show characteristics of oligodendrocyte progenitor cells (OPCs) and are therapy sensitive, and that OPC rather than NSC markers enriched for tumor formation. MRI of human oligodendroglioma also suggested a white matter (WM) origin, with markers for OPCs rather than NSCs similarly enriching for tumor formation. Our results suggest that oligodendroglioma cells show hallmarks of OPCs, and that a progenitor rather than a NSC origin underlies improved prognosis in patients with this tumor.
PubMedId: 21156288
Journal-article: Cancer cell, vol. 18, 2009
Authors: Fraser J Sim, Jennifer K Lang, Tracy A Ali, Neeta S Roy, G Edward Vates, Webster H Pilcher, Steven A Goldman
Abstract: The statins have been proposed as possible therapeutic agents for a variety of autoimmune disorders, including multiple sclerosis. In a genomic screen, we found that glial progenitor cells (GPCs) of the adult human white matter expressed significant levels of the principal statin target, HMG-CoA reductase, as well as additional downstream members of the sterol synthesis pathway. We therefore asked if statin treatment might influence the differentiated fate of adult glial progenitor cells. To assess the functional importance of the sterol synthesis pathway to adult human glial progenitors, we used simvastatin or pravastatin to inhibit HMG-CoA reductase, and then assessed the phenotypic differentiation of the progenitors, as well as the molecular concomitants thereof. We found that both statins induced a dose-dependent induction of oligodendrocyte phenotype, and concomitant reduction in progenitor number. Oligodendrocyte commitment was associated with induction of the sterol-regulated nuclear co-receptor PPARgamma, and could be blocked by the specific PPARgamma antagonist GW9662. Thus, statins may promote oligodendrocyte lineage commitment by parenchymal glial progenitor cells; this might reduce the available progenitor pool, and hence degrade the long-term regenerative competence of the adult white matter.
PubMedId: 18383345
Journal-article: Glia, vol. 56, 2008
Authors: Ditte Lovatt, Ursula Sonnewald, Helle S Waagepetersen, Arne Schousboe, Wei He, Jane H-C Lin, Xiaoning Han, Takahiro Takano, Su Wang, Fraser J Sim, Steven A Goldman, Maiken Nedergaard
Abstract: Protoplasmic astrocytes are critically important to energy metabolism in the CNS. Our current understanding of the metabolic interactions between neurons and glia is based on studies using cultured cells, from which mainly inferential conclusions have been drawn as to the relative roles of neurons and glia in brain metabolism. In this study, we used functional genomics to establish the relative compartmentalization of neuronal and astrocytic metabolic pathways in the adult brain. To this end, fluorescence-activated cell sorting was used to directly isolate neurons and protoplasmic astrocytes from the cortex of adult mice. Microarray analysis showed that astrocytes and neurons each express transcripts predicting individual self-sufficiency in both glycolysis and oxidative metabolism. Surprisingly, most enzymes in the tricarboxylic acid (TCA) cycle were expressed at higher relative levels in astrocytes than in neurons. Mass spectrometric analysis of the TCA cycle intermediates confirmed that freshly isolated adult astrocytes maintained an active TCA cycle, whereas immuno-electron microscopy revealed that fine astrocytic processes encompassing synapses contained a higher density of mitochondria than surrounding cells. These observations indicate that astrocytes exhibit robust oxidative metabolism in the intact adult brain and suggest a prominent contribution of astrocytic metabolism to functional brain imaging, including BOLD (blood-oxygen level-dependent) functional magnetic resonance imaging signals.
PubMedId: 17989291
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 27, 2007
Authors: Fraser J. Sim, Jennifer K. Lang, Ben Waldau, Neeta S. Roy, Theodore E. Schwartz, Webster H. Pilcher, Karen J. Chandross, Sridaran Natesan, Jean E. Merrill, Steven A. Goldman
Journal-article: Annals of Neurology, vol. 59, pp. 990-990, 2006
Authors: Steven A. Goldman, Fraser J. Sim
Book-chapter: Gene Therapy of the Central Nervous System, pp. 53-65, 2006
Authors: Fraser J Sim, Jennifer K Lang, Ben Waldau, Neeta S Roy, Theodore E Schwartz, Webster H Pilcher, Karen J Chandross, Sridaran Natesan, Jean E Merrill, Steven A Goldman, Steven A Goldmanm
Abstract: Glial progenitor cells are abundant in adult human white matter. This study was designed to identify signaling pathways regulating their self-renewal and fate. We compared the transcriptional profiles of freshly sorted adult human white matter progenitor cells (WMPCs), purified by A2B5-based immunomagnetic sorting, with those of the white matter from which they derived. We identified 132 genes differentially expressed by WMPCs; these included principal components of five receptor-defined signaling pathways, represented by platelet derived growth factor receptor alpha (PDGFRA) and type 3 fibroblast growth factor receptor (FGFR3), receptor tyrosine phosphatase-beta/zeta (RTPZ), notch, and syndecan3. WMPCs also differentially expressed the bone morphogenetic protein 4 (BMP4) inhibitors neuralin and BAMBI (BMP and activin membrane-bound inhibitor), suggesting tonic defense against BMP signaling. Differential overexpression of RTPZ was accompanied by that of its modulators pleiotrophin, NrCAM, tenascin, and the chondroitin sulfate proteoglycans, suggesting the importance of RTPZ signaling to WMPCs. When exposed to the RTPZ inhibitor bpV(phen), or lentiviral-shRNAi against RTPZ, WMPCs differentiated as oligodendrocytes. Conversely, when neuralin and BAMBI were antagonized by BMP4, astrocytic differentiation was induced, which was reversible by noggin. The RTPZ and BMP pathways regulate the self-maintenance of adult human WMPCs, and can be modulated to induce their oligodendrocytic or astrocytic differentiation. As such, they provide targets by which to productively mobilize resident progenitor cells of the adult human brain.
PubMedId: 16634042
Journal-article: Annals of neurology, vol. 59, 2006
Authors: Fraser J Sim, H Michael Keyoung, James E Goldman, Dong Kyu Kim, Hee-Won Jung, Neeta S Roy, Steven A Goldman
Abstract: Central neurocytoma (CN) is a rare periventricular tumor, whose derivation, lineage potential, and molecular regulation have been mostly unexplored. We noted that CN cells exhibited an antigenic profile typical of neuronal progenitor cells in vivo, yet in vitro generated neurospheres, divided in response to bFGF (basic fibroblast growth factor), activated the neuroepithelial enhancer of the nestin gene, and gave rise to both neuron-like cells and astrocytes. When CN gene expression was compared with that of both normal adult VZ (ventricular zone) and E/nestin:GFP (green fluorescent protein)-sorted native neuronal progenitors, significant overlap was noted. Marker analysis suggested that the gene expression pattern of CN was that of a proneuronal population; glial markers were conspicuously absent, suggesting that the emergence of astroglia from CN occurred only with passage. The expression pattern of CN was distinguished from that of native progenitor cells by a cohort of differentially expressed genes potentially involved in both the oncogenesis and phenotypic restriction of neurocytoma. These included both IGF2 and several components of its signaling pathway, whose sharp overexpression implicated dysregulated autocrine IGF2 signaling in CN oncogenesis. Both receptors and effectors of canonical wnt signaling, as well as GDF8 (growth differentiation factor 8), PDGF-D, and neuregulin, were differentially overexpressed by CN, suggesting that CN is characterized by the concurrent overactivation of these pathways, which may serve to drive neurocytoma expansion while restricting tumor progenitor phenotype. This strategy of comparing the gene expression of tumor cells to that of the purified native progenitors from which they derive may provide a focused approach to identifying transcripts important to stem and progenitor cell oncogenesis.
PubMedId: 17135416
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 26, 2006
Authors: C. Ibanez, S.A. Shields, M. El-Etr, E. Leonelli, V. Magnaghi, W.-W. Li, F.J. Sim, E.-E. Baulieu, R.C. Melcangi, M. Schumacher, R.J.M. Franklin
Journal-article: Progress in Neurobiology, vol. 71, pp. 49-56, 2003
Authors: C Ibanez, S A Shields, M El-Etr, E Leonelli, V Magnaghi, W-W Li, F J Sim, E-E Baulieu, R C Melcangi, M Schumacher, R J M Franklin
PubMedId: 14611867
Comparative Study: Progress in neurobiology, vol. 71, 2003
Authors: Fraser J Sim, Chao Zhao, Wen-Wu Li, Andras Lakatos, Robin J M Franklin
PubMedId: 12213447
Journal Article: Molecular and cellular neurosciences, vol. 20, 2002
Authors: P. M. Smith, F. J. Sim, R. J. M. Franklin
Journal-article: Neuropathology and Applied Neurobiology, vol. 28, pp. 154-154, 2002
Authors: Fraser J. Sim, Chao Zhao, Jacques Penderis, Robin J. M. Franklin
Journal-article: The Journal of Neuroscience, vol. 22, pp. 2451-2459, 2002
Authors: Robin J M Franklin, Chao Zhao, Fraser J Sim
PubMedId: 12004191
Journal Article: Neuroreport, vol. 13, 2002
Authors: Robin J. M. Franklin, Chao Zhao, Fraser J. Sim
Journal-article: NeuroReport, vol. 13, pp. 923-928, 2002
Authors: F Sim
Journal-article: Molecular and Cellular Neuroscience, vol. 20, pp. 669-682, 2002
Authors: Fraser J Sim, Chao Zhao, Jacques Penderis, Robin J M Franklin
Abstract: The age-associated decrease in the efficiency of CNS remyelination has clear implications for recovery from demyelinating diseases such as multiple sclerosis (MS) that may last for several decades. Developing strategies to reverse the age-associated decline requires the identification of how the regenerative process is impaired. We addressed whether remyelination becomes slower because of an impairment of recruitment of oligodendrocyte progenitors (OPs) or, as is the case in some MS lesions, an impairment of OP differentiation into remyelinating oligodendrocytes. The OP response during remyelination of focal, toxin-induced CNS demyelination in young and old rats was compared by in situ hybridization using probes to two OP-expressed mRNA species: platelet-derived growth factor-alpha receptor and the OP transcription factor myelin transcription factor 1 (MyT1). We found that the expression patterns for the two OP markers are very similar and reveal a delay in the colonization of the demyelinated focus with OPs in the old animals compared with the young animals. By comparing the mRNA expression pattern of MyT1 with that of the myelin proteins myelin basic protein and Gtx, we found that in the old animals there is also a delay in OP differentiation that increases with longer survival times. These results indicate that the age-associated decrease in remyelination efficiency occurs because of an impairment of OP recruitment and the subsequent differentiation of the OPs into remyelinating oligodendrocytes, and that strategies aimed at ameliorating the age-associated decline in remyelination efficiency will therefore need to promote both components of the regenerative process.
PubMedId: 11923409
Journal-article: The Journal of neuroscience : the official journal of the Society for Neuroscience, vol. 22, 2002
Authors: P.M. Smith, F.J. Sim, S.C. Barnett, Robin J.M. Franklin
Journal-article: Glia, vol. 36, pp. 342-353, 2001
Authors: P M Smith, F J Sim, S C Barnett, R J Franklin
PubMedId: 11746771
Journal Article: Glia, vol. 36, 2001
Authors: Mark R. Kotter, Anna Setzu, Fraser J. Sim, Nico Van Rooijen, Robin J.M. Franklin
Journal-article: Glia, vol. 35, pp. 204-212, 2001
Authors: M R Kotter, A Setzu, F J Sim, N Van Rooijen, R J Franklin
PubMedId: 11494411
Journal Article: Glia, vol. 35, 2001
Authors: F.J Sim, G.L Hinks, R.J.M Franklin
Journal-article: Neuroscience, vol. 100, pp. 131-139, 2000
Authors: F J Sim, G L Hinks, R J Franklin
PubMedId: 10996464
Journal Article: Neuroscience, vol. 100, 2000
Authors: F. J. Sim, S. A. Goldman
Book-chapter: Opportunities and Challenges of the Therapies Targeting CNS Regeneration, pp. 61-81,
Authors: S. A. Goldman, J. Lang, N. Roy, S. J. Schanz, F. S. Sim, S. Wang, V. Washco, M. S. Windrem
Book-chapter: Ernst Schering Research Foundation Workshop, pp. 195-213,
Authors: Steven A. Goldman, Fraser Sim
Book-chapter: Stem Cells: Nuclear Reprogramming and Therapeutic Applications, pp. 66-91,
Presented by CUBS © 2024