199 results on '"Vescovi A. L."'
Search Results
152. Lineage restriction of neuroepithelial precursor cells from fetal human spinal cord
- Author
-
Quinn, Sean M., primary, Walters, Winston M., additional, Vescovi, Angelo L., additional, and Whittemore, Scott R., additional
- Published
- 1999
- Full Text
- View/download PDF
153. Isolation and Intracerebral Grafting of Nontransformed Multipotential Embryonic Human CNS Stem Cells
- Author
-
VESCOVI, ANGELO L., primary, GRITTI, ANGELA, additional, GALLI, ROSSELLA, additional, and PARATI, EUGENIO A., additional
- Published
- 1999
- Full Text
- View/download PDF
154. Establishment and Properties of Neural Stem Cell Clones: Plasticity In Vitro and In Vivo
- Author
-
Vescovi, Angelo L., primary and Snyder, Evan Y., additional
- Published
- 1999
- Full Text
- View/download PDF
155. Epidermal and Fibroblast Growth Factors Behave as Mitogenic Regulators for a Single Multipotent Stem Cell-Like Population from the Subventricular Region of the Adult Mouse Forebrain
- Author
-
Gritti, Angela, primary, Frölichsthal-Schoeller, Paola, additional, Galli, Rossella, additional, Parati, Eugenio A., additional, Cova, Lidia, additional, Pagano, Stefano F., additional, Bjornson, Christopher R., additional, and Vescovi, Angelo L., additional
- Published
- 1999
- Full Text
- View/download PDF
156. Basic fibroblast growth factor supports the proliferation of epidermal growth factor-generated neuronal precursor cells of the adult mouse CNS
- Author
-
Gritti, Angela, primary, Cova, Lidia, additional, Parati, Eugenio A., additional, Galli, Rossella, additional, and Vescovi, Angelo L., additional
- Published
- 1995
- Full Text
- View/download PDF
157. Differential effects of CGRP on adenylyl cyclase in adult and embryonic rat brain
- Author
-
Villa, Isabella, primary, Colangelo, Anna Maria, additional, Vescovi, Angelo L, additional, Gritti, Angela, additional, Schinelli, Sergio, additional, Pagani, Francesca, additional, and Guidobono, Francesca, additional
- Published
- 1994
- Full Text
- View/download PDF
158. Murine neural stem cells model Hunter disease in vitro: glial cell-mediated neurodegeneration as a possible mechanism involved.
- Author
-
Poli, E. Fusar, Zalfa, C., D'Avanzo, F., Tomanin, R., Carlessi, L., Bossi, M., Nodari, L. Rota, Binda, E., Marmiroli, P., Scarpa, M., Delia, D., Vescovi, A. L., and De Filippis, L.
- Published
- 2013
- Full Text
- View/download PDF
159. bFGF regulates the proliferative fate of unipotent (neuronal) and bipotent (neuronal/astroglial) EGF-generated CNS progenitor cells
- Author
-
Vescovi, Angelo L., primary, Reynolds, Brent A., additional, Fraser, Douglas D., additional, and Weiss, Samuel, additional
- Published
- 1993
- Full Text
- View/download PDF
160. NOTCH Pathway Blockade Depletes CD133-Positive Glioblastoma Cells and Inhibits Growth of Tumor Neurospheres and Xenografts.
- Author
-
XING FAN, KHAKI, LEILA, ZHU, THANT S., SOULES, MARY E., TALSMA, CAROLINE E., GUL, NAHEED, KOH, CHERYL, JIANGYANG ZHANG, YUE-MING LI, MACIACZYK, JAREK, NIKKHAH, GUIDO, DIMECO, FRANCESCO, PICCIRILLO, SARA, VESCOVI, ANGELO L., and EBERHART, CHARLES G.
- Subjects
TUMOR growth ,BRAIN tumors ,XENOGRAFTS ,CANCER cells ,GLIOMAS ,DIMETHYL sulfoxide ,PHOSPHORYLATION - Abstract
Cancer stem cells (CSCs) are thought to be critical for the engraftment and long-term growth of many tumors, including glioblastoma (GBM). The cells are at least partially spared by traditional chemotherapies and radiation therapies, and finding new treatments that can target CSCs may be critical for improving patient survival. It has been shown that the NOTCH signaling pathway regulates normal stem cells in the brain, and that GBMs contain stemlike cells with higher NOTCH activity. We therefore used low-passage and established GBM-derived neurosphere cultures to examine the overall requirement for NOTCH activity, and also examined the effects on tumor cells expressing stem cell markers. NOTCH blockade by γ-secretase inhibitors (GSIs) reduced neurosphere growth and clonogenicity in vitro, whereas expression of an active form of NOTCH2 increased tumor growth. The putative CSC markers CD133, NESTIN, BMI1, and OLIG2 were reduced following NOTCH blockade. When equal numbers of viable cells pretreated with either vehicle (dimethyl sulfoxide) or GSI were injected subcutaneously into nude mice, the former always formed tumors, whereas the latter did not. In vivo delivery of GSI by implantation of drug-impregnated polymer beads also effectively blocked tumor growth, and significantly prolonged survival, albeit in a relatively small cohort of animals. We found that NOTCH pathway inhibition appears to deplete stem-like cancer cells through reduced proliferation and increased apoptosis associated with decreased AKT and STAT3 phosphorylation. In summary, we demonstrate that NOTCH pathway blockade depletes stem-like cells in GBMs, suggesting that GSIs may be useful as chemotherapeutic reagents to target CSCs in malignant gliomas. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
161. DNER, an Epigenetically Modulated Gene, Regulates Glioblastoma-Derived Neurosphere Cell Differentiation and Tumor Propagation.
- Author
-
Peng Sun, Shuli Xia, Lal, Bachchu, Eberhart, Charles G., Quinones-Hinojosa, Alfredo, Maciaczyk, Jarek, Matsui, William, Dimeco, Francesco, Piccirillo, Sara M., Vescovi, Angelo L., and Laterra, John
- Subjects
GLIOBLASTOMA multiforme ,MORPHOGENESIS ,TUMORS ,ANTINEOPLASTIC antibiotics ,BASIC proteins - Abstract
Neurospheres derived from glioblastoma (GBM) and other solid malignancies contain neoplastic stem-like cells that efficiently propagate tumor growth and resist cytotoxic therapeutics. The primary objective of this study was to use histone-modifying agents to elucidate mechanisms by which the phenotype and tumor-promoting capacity of GBM-derived neoplastic stem-like cells are regulated. Using established GBM-derived neurosphere lines and low passage primary GBM-derived neurospheres, we show that histone deacetylase (HDAC) inhibitors inhibit growth, induce differentiation, and induce apoptosis of neoplastic neurosphere cells. A specific gene product induced by HDAC inhibition, Delta/Notch-like epidermal growth factor-related receptor (DNER), inhibited the growth of GBM-derived neurospheres, induced their differentiation in vivo and in vitro, and inhibited their engraftment and growth as tumor xenografts. The differentiating and tumor suppressive effects of DNER, a noncanonical Notch ligand, contrast with the previously established tumor-promoting effects of canonical Notch signaling in brain cancer stem-like cells. Our findings are the first to implicate noncanonical Notch signaling in the regulation of neoplastic stem-like cells and suggest novel neoplastic stem cell targeting treatment strategies for GBM and potentially other solid malignancies. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
162. Efficient In Vitro Labeling of Human Neural Precursor Cells with Superparamagnetic Iron Oxide Particles: Relevance for In Vivo Cell Tracking.
- Author
-
Neri, Margherita, Maderna, Claudio, Cavazzin, Chiara, Deidda-Vigoriti, Vivien, Politi, Letterio S., Scotti, Giuseppe, Marzola, Pasquina, Sbarbati, Andrea, Vescovi, Angelo L., and Gritti, Angela
- Subjects
STEM cells ,EMBRYONIC stem cells ,HEMATOPOIETIC stem cells ,BLOOD cells ,BONE marrow cells ,DIAGNOSTIC imaging ,CONFOCAL microscopy - Abstract
Recent studies have raised appealing possibilities of replacing damaged or lost neural cells by transplanting in vitro-expanded neural precursor cells (NPCs) and/or their progeny. Magnetic resonance (MR) tracking of superparamagnetic iron oxide (SPIO)-labeled cells is a noninvasive technique to track transplanted cells in longitudinal studies on living animals. Murine NPCs and human mesenchymal or hematopoietic stem cells can be efficiently labeled by SPIOs. However, the validation of SPIO-based protocols to label human neural precursor cells (hNPCs) has not been extensively addressed. Here, we report the development and validation of optimized protocols using two SPIOs (Sinerem and Endorem) to label human hNPCs that display bona fide stem cell features in vitro. A careful titration of both SPIOs was required to set the conditions resulting in efficient cell labeling without impairment of cell survival, proliferation, self-renewal, and multipotency. In vivo magnetic resonance imaging (MRI) combined with histology and confocal microscopy indicated that low numbers (5 × 10
3 to 1 × 104 ) of viable SPIO-labeled hNPCs could be efficiently detected in the short term after transplantation in the adult murine brain and could be tracked for at least 1 month in longitudinal studies. By using this approach, we also clarified the impact of donor cell death to the MR signal. This study describes a simple protocol to label NPCs of human origin using SPIOs at optimized low dosages and demonstrates the feasibility of noninvasive imaging of labeled cells after transplantation in the brain; it also evidentiates potential limitations of the technique that have to be considered, particularly in the perspective of neural cell-based clinical applications. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
163. Storage of Mutant Human SOD1 in Non-Neural Cells from the Type-1 Amyotrophic Lateral Sclerosis rat G93A Model Correlated with the Lysosomes' Dysfunction.
- Author
-
Bicchi, Ilaria, Morena, Francesco, Argentati, Chiara, Nodari, Laura Rota, Emiliani, Carla, Gelati, Maurizio, Vescovi, Angelo L., and Martino, Sabata
- Subjects
AMYOTROPHIC lateral sclerosis ,LYSOSOMES ,SUPEROXIDE dismutase ,HOMEOSTASIS ,METABOLIC disorders - Abstract
Herein, we explored the impact of the lysosome dysfunction during the progression of Amyotrophic Lateral Sclerosis type-1 (ALS1). We conducted the study in non-neural cells, primary fibroblasts (rFFFs), and bone marrow-mesenchymal stem cells (rBM-MSCs), isolated from the animal model rat
G93A for ALS1 at two stages of the disease: Pre-symptomatic-stage (ALS1-PreS) and Terminal-stage (ALS1-EndS). We documented the storage of human mutant Superoxide Dismutase 1, SOD1G93A (SOD1*) in the lysosomes of ALS1-rFFFs and ALS1-rBM-MSCs and demonstrated the hallmarks of the disease in non-neural cells as in ratG93A -ALS1-tissues. We showed that the SOD1* storage is associated with the altered glycohydrolases and proteases levels in tissues and both cell types from ALS1-PreS to ALS1-EndS. Only in ALS1-rFFFs, the lysosomes lost homeostasis, enlarge drastically, and contribute to the cell metabolic damage. Contrariwise, in ALS1-rBM-MSCs, we found a negligible metabolic dysfunction, which makes these cells' status similar to WT. We addressed this phenomenon to a safety mechanism perhaps associated with an enhanced lysosomal autophagic activity in ALS1-rBM-MSCs compared to ALS1-rFFFs, in which the lysosomal level of LC3-II/LC3I was comparable to that of WT-rFFFs. We suggested that the autophagic machinery could balance the storage of SOD1* aggregates and the lysosomal enzyme dysfunction even in ALS1-EndS-stem cells. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
164. Emx2 regulates the proliferation of stem cells of the adult mammalian central nervous system
- Author
-
Galli, Rossella, Fiocco, Roberta, De Filippis, Lidia, Muzio, Luca, Gritti, Angela, Mercurio, Sara, Broccoli, Vania, Pellegrini, Massimo, Mallamaci, Antonello, and Vescovi, Angelo L.
- Abstract
The appropriate control of proliferation of neural precursors has fundamental implications for the development of the central nervous system and for cell homeostasis/replacement within specific brain regions throughout adulthood. The role of genetic determinants in this process is largely unknown. We report the expression of the homeobox transcription factor Emx2 within the periventricular region of the adult telencephalon. This neurogenetic area displays a large number of multipotent stem cells. Adult neural stem cells isolated from this region do express Emx2 and down-regulate it significantly upon differentiation into neurons and glia. Abolishing or, increasing Emx2 expression in adult neural stem cells greatly enhances or reduces their rate of proliferation, respectively. We determined that altering the expression of Emx2 affects neither the cell cycle length of adult neural stem cells nor their ability to generate neurons and glia. Rather, when Emx2 expression is abolished, the frequency of symmetric divisions that generate two stem cells increases, whereas it decreases when Emx2 expression is enhanced.
- Published
- 2002
- Full Text
- View/download PDF
165. The neural stem cells and their transdifferentiation capacity
- Author
-
Vescovi, A. L., Galli, R., and Gritti, A.
- Published
- 2001
- Full Text
- View/download PDF
166. Sox2 regulatory sequences direct expression of a (beta)-geo transgene to telencephalic neural stem cells and precursors of the mouse embryo, revealing regionalization of gene expression in CNS stem cells.
- Author
-
Zappone, M V, Galli, R, Catena, R, Meani, N, De Biasi, S, Mattei, E, Tiveron, C, Vescovi, A L, Lovell-Badge, R, Ottolenghi, S, and Nicolis, S K
- Abstract
Sox2 is one of the earliest known transcription factors expressed in the developing neural tube. Although it is expressed throughout the early neuroepithelium, we show that its later expression must depend on the activity of more than one regionally restricted enhancer element. Thus, by using transgenic assays and by homologous recombination-mediated deletion, we identify a region upstream of Sox2 (-5.7 to -3.3 kb) which can not only drive expression of a (beta)-geo transgene to the developing dorsal telencephalon, but which is required to do so in the context of the endogenous gene. The critical enhancer can be further delimited to an 800 bp fragment of DNA surrounding a nuclease hypersensitive site within this region, as this is sufficient to confer telencephalic expression to a 3.3 kb fragment including the Sox2 promoter, which is otherwise inactive in the CNS. Expression of the 5.7 kb Sox2(beta)-geo transgene localizes to the neural plate and later to the telencephalic ventricular zone. We show, by in vitro clonogenic assays, that transgene-expressing (and thus G418-resistant) ventricular zone cells include cells displaying functional properties of stem cells, i.e. self-renewal and multipotentiality. We further show that the majority of telencephalic stem cells express the transgene, and this expression is largely maintained over two months in culture (more than 40 cell divisions) in the absence of G418 selective pressure. In contrast, stem cells grown in parallel from the spinal cord never express the transgene, and die in G418. Expression of endogenous telencephalic genes was similarly observed in long-term cultures derived from the dorsal telencephalon, but not in spinal cord-derived cultures. Thus, neural stem cells of the midgestation embryo are endowed with region-specific gene expression (at least with respect to some networks of transcription factors, such as that driving telencephalic expression of the Sox2 transgene), which can be inherited through multiple divisions outside the embryonic environment.
- Published
- 2000
167. Sox2 regulatory sequences direct expression of a β-geo transgene to telencephalic neural stem cells and precursors of the mouse embryo, revealing regionalization of gene expression in CNS stem cells
- Author
-
Zappone, Massimo V., Galli, Rossella, Catena, Raffaella, Meani, Natalia, Biasi, Silvia De, Mattei, Elisabetta, Tiveron, Cecilia, Vescovi, Angelo L., Lovell-Badge, Robin, Ottolenghi, Sergio, and Nicolis, Silvia K.
- Abstract
Sox2 is one of the earliest known transcription factors expressed in the developing neural tube. Although it is expressed throughout the early neuroepithelium, we show that its later expression must depend on the activity of more than one regionally restricted enhancer element. Thus, by using transgenic assays and by homologous recombination-mediated deletion, we identify a region upstream of Sox2 (−5.7 to −3.3 kb) which can not only drive expression of a β-geo transgene to the developing dorsal telencephalon, but which is required to do so in the context of the endogenous gene. The critical enhancer can be further delimited to an 800 bp fragment of DNA surrounding a nuclease hypersensitive site within this region, as this is sufficient to confer telencephalic expression to a 3.3 kb fragment including the Sox2 promoter, which is otherwise inactive in the CNS. Expression of the 5.7 kb Sox2β-geo transgene localizes to the neural plate and later to the telencephalic ventricular zone. We show, by in vitro clonogenic assays, that transgene-expressing (and thus G418-resistant) ventricular zone cells include cells displaying functional properties of stem cells, i.e. self-renewal and multipotentiality. We further show that the majority of telencephalic stem cells express the transgene, and this expression is largely maintained over two months in culture (more than 40 cell divisions) in the absence of G418 selective pressure. In contrast, stem cells grown in parallel from the spinal cord never express the transgene, and die in G418. Expression of endogenous telencephalic genes was similarly observed in long-term cultures derived from the dorsal telencephalon, but not in spinal cord-derived cultures. Thus, neural stem cells of the midgestation embryo are endowed with region-specific gene expression (at least with respect to some networks of transcription factors, such as that driving telencephalic expression of the Sox2 transgene), which can be inherited through multiple divisions outside the embryonic environment.
- Published
- 2000
- Full Text
- View/download PDF
168. Establishment and Properties of Neural Stem Cell Clones: Plasticity In Vitroand In Vivo
- Author
-
Vescovi, Angelo L. and Snyder, Evan Y.
- Abstract
The study of the basic physiology of the neural precursors generated during brain development is driven by two inextricably linked goals. First, such knowledge is instrumental to our understanding of how the high degree of cellular complexity of the mature central nervous system (CNS) is generated, and how to dissect the steps of proliferation, fate commitment, and differentiation that lead early pluripotent neural progenitors to give rise to mature CNS cells. Second, it is hoped that the isolation, propagation, and manipulation of brain precursors and, particularly, of multipotent neural stem cells (NSCs), will lead to therapeutic applications in neurological disorders. The debate is still open concerning the most appropriate definition of a stem cell and on how it is best identified, characterized, and manipulated. By adopting an operational definition of NSCs, we review some of the basic findings in this area and elaborate on their potential threapeutic applications. Further, we discuss recent evidence from our two groups that describe, based on that rigorous definition, the isolation and propagation of clones of NSCs from the human fetal brain and illustrate how they have begun to show promise for neural cell replacement and molecular support therapy in models of degenerative CNS diseases. The extensive propagation and engraftment potential of human CNS stem cells may, in the not‐too‐distant future, be directed towards genuine clinical therapeutic ends, and may open novel and multifaceted strategies for redressing a variety of heretofore untreatable CNS dysfunctions.
- Published
- 1999
- Full Text
- View/download PDF
169. Epidermal and fibroblast growth factors behave as mitogenic regulators for a single multipotent stem cell-like population from the subventricular region of the adult mouse forebrain
- Author
-
Gritti, A., Frölichsthal-Schoeller, P., Galli, R., Parati, E. A., Lidia Cova, Pagano, S. F., Bjornson, C. R., and Vescovi, A. L.
170. NEURAL PRECURSOR CELL CULTURES FROM GM2-GANGLIOSIDOSIS MURINE MODELS RECAPITULATE THE BIOCHEMICAL HALLMARKS OF THE BRAIN PATHOLOGY
- Author
-
Gritti, A., Di Girolamo, I., Cavazzin, C., Tiribuzi, R., Rivaroli, A., Sandro Sonnino, Vescovi, A. L., Orlacchio, A., and Martino, S.
171. Electrospun microfiber tubes and self-assembling peptides stimulate neural regeneration in rat sciatic nerve transections
- Author
-
Gelain, F., Silvia Panseri, Modica, L., Taraballi, F., Lowery, J. L., and Vescovi, A. L.
172. Beta sheet forming peptides functionalization with biologically active motifs may alter their self-assembling propensity
- Author
-
Taraballi, F., Campione, M., Sassella, A., Vescovi, A. L., Hwang, W., Paleari, A., and Fabrizio Gelain
173. Inorganic mercury changes the fate of murine CNS stem cells
- Author
-
Cedrola, S., Guzzi, G., Ferrari, D., Gritti, A., Vescovi, A. L., Pendergrass, J. C., and caterina la porta
174. Neurosphere Cultures
- Author
-
Vescovi, Angelo L., primary
- Full Text
- View/download PDF
175. Different states of stemness of glioblastoma stem cells sustain glioblastoma subtypes indicating novel clinical biomarkers and high-efficacy customized therapies.
- Author
-
Visioli, Alberto, Trivieri, Nadia, Mencarelli, Gandino, Giani, Fabrizio, Copetti, Massimiliano, Palumbo, Orazio, Pracella, Riccardo, Cariglia, Maria Grazia, Barile, Chiara, Mischitelli, Luigi, Soriano, Amata Amy, Palumbo, Pietro, Legnani, Federico, DiMeco, Francesco, Gorgoglione, Leonardo, Pesole, Graziano, Vescovi, Angelo L., and Binda, Elena
- Abstract
Background: Glioblastoma (GBM) is the most malignant among gliomas with an inevitable lethal outcome. The elucidation of the physiology and regulation of this tumor is mandatory to unravel novel target and effective therapeutics. Emerging concepts show that the minor subset of glioblastoma stem cells (GSCs) accounts for tumorigenicity, representing the true target for innovative therapies in GBM. Methods: Here, we isolated and established functionally stable and steadily expanding GSCs lines from a large cohort of GBM patients. The molecular, functional and antigenic landscape of GBM tissues and their derivative GSCs was highlited in a side-by-side comprehensive genomic and transcriptomic characterization by ANOVA and Fisher’s exact tests. GSCs’ physio-pathological hallmarks were delineated by comparing over time in vitro and in vivo their expansion, self-renewal and tumorigenic ability with hierarchical linear models for repeated measurements and Kaplan–Meier method. Candidate biomarkers performance in discriminating GBM patients’ classification emerged by classification tree and patients’ survival analysis. Results: Here, distinct biomarker signatures together with aberrant functional programs were shown to stratify GBM patients as well as their sibling GSCs population into TCGA clusters. Of importance, GSCs cells were demonstrated to fully resemble over time the molecular features of their patient of origin. Furthermore, we pointed out the existence of distinct GSCs subsets within GBM classification, inherently endowed with different self-renewal and tumorigenic potential. Particularly, classical GSCs were identified by more undifferentiated biological hallmarks, enhanced expansion and clonal capacity as compared to the more mature, relatively slow-propagating mesenchymal and proneural cells, likely endowed with a higher potential for infiltration either ex vivo or in vivo. Importantly, the combination of DCX and EGFR markers, selectively enriched among GSCs pools, almost exactly predicted GBM patients’ clusters together with their survival and drug response. Conclusions: In this study we report that an inherent enrichment of distinct GSCs pools underpin the functional inter-cluster variances displayed by GBM patients. We uncover two selectively represented novel functional biomarkers capable of discriminating GBM patients’ stratification, survival and drug response, setting the stage for the determination of patient-tailored diagnostic and prognostic strategies and, mostly, for the design of appropriate, patient-selective treatment protocols. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
176. Reply: Quantitative evaluation of the human subventricular zone.
- Author
-
van den Berge, Simone A., van Strien, Miriam E., Korecka, Joanna A., Dijkstra, Anke A., Sluijs, Jacqueline A., Kooijman, Lieneke, Eggers, Ruben, De Filippis, Lidia, Vescovi, Angelo L., Verhaagen, Joost, van de Berg, Wilma D.J., and Hol, Elly M.
- Subjects
QUANTITATIVE research ,PARKINSON'S disease ,NEURAL stem cells ,CELL lines ,CELL differentiation ,GLIAL fibrillary acidic protein ,BROMODEOXYURIDINE - Published
- 2012
- Full Text
- View/download PDF
177. Ground-breaking stem-cell work has been reproduced.
- Author
-
Vescovi, Angelo L., Reynolds, Brent A., Rietze, Rodney L., and Bjornson, Christopher
- Subjects
- *
LETTERS to the editor , *STEM cell research - Abstract
A letter to the editor is presented in response to the article "The hard copy," published in the previous issue.
- Published
- 2007
- Full Text
- View/download PDF
178. CD90 is Identified as a Candidate Marker for Cancer Stem Cells in Primary High-Grade Gliomas Using Tissue Microarrays*
- Author
-
He, Jintang, Liu, Yashu, Zhu, Thant, Zhu, Jianhui, DiMeco, Francesco, Vescovi, Angelo L., Heth, Jason A., Muraszko, Karin M., Fan, Xing, and Lubman, David M.
- Abstract
Although CD90 has been identified as a marker for various kinds of stem cells including liver cancer stem cells (CSCs) that are responsible for tumorigenesis, the potential role of CD90 as a marker for CSCs in gliomas has not been characterized. To address the issue, we investigated the expression of CD90 in tissue microarrays containing 15 glioblastoma multiformes (GBMs), 19 WHO grade III astrocytomas, 13 WHO grade II astrocytomas, 3 WHO grade I astrocytomas and 8 normal brain tissues. Immunohistochemical analysis showed that CD90 was expressed at a medium to high level in all tested high-grade gliomas (grade III and GBM) whereas it was barely detectable in low-grade gliomas (grade I and grade II) and normal brains. Double immunofluorescence staining for CD90 and CD133 in GBM tissues revealed that CD133+CSCs are a subpopulation of CD90+cells in GBMs in vivo. Flow cytometry analysis of the expression of CD90 and CD133 in GBM-derived stem-like neurospheres further confirmed the conclusion in vitro. The expression levels of both CD90 and CD133 were reduced along with the loss of stem cells after differentiation. Furthermore, the limiting dilution assay demonstrated that the sphere formation ability was comparable between the CD90+/CD133+and the CD90+/CD133−populations of GBM neurospheres, which is much higher than that of the CD90−/CD133−population. We also performed double staining for CD90 and a vascular endothelial cell marker CD31 in tissue microarrays which revealed that the CD90+cells were clustered around the tumor vasculatures in high-grade glioma tissues. These findings suggest that CD90 is not only a potential prognostic marker for high-grade gliomas but also a marker for CSCs within gliomas, and it resides within endothelial niche and may also play a critical role in the generation of tumor vasculatures via differentiation into endothelial cells.
- Published
- 2012
- Full Text
- View/download PDF
179. AQP4-dependent glioma cell features affect the phenotype of surrounding cells via extracellular vesicles.
- Author
-
Simone, Laura, Pisani, Francesco, Binda, Elena, Frigeri, Antonio, Vescovi, Angelo L., Svelto, Maria, and Nicchia, Grazia P.
- Subjects
- *
EXTRACELLULAR vesicles , *GLIOMAS , *BRAIN tumors , *GLIOBLASTOMA multiforme , *ORTHOGONAL arrays - Abstract
Background: Extracellular vesicles (EVs) are membrane-enclosed particles released systemically by all cells, including tumours. Tumour EVs have been shown to manipulate their local environments as well as distal targets to sustain the tumour in a variety of tumours, including glioblastoma (GBM). We have previously demonstrated the dual role of the glial water channel aquaporin-4 (AQP4) protein in glioma progression or suppression depending on its aggregation state. However, its possible role in communication mechanisms in the microenvironment of malignant gliomas remains to be unveiled. Results: Here we show that in GBM cells AQP4 is released via EVs that are able to affect the GBM microenvironment. To explore this role, EVs derived from invasive GBM cells expressing AQP4-tetramers or apoptotic GBM cells expressing orthogonal arrays of particles (AQP4-OAPs) were isolated, using a differential ultracentrifugation method, and were added to pre-seeded GBM cells. Confocal microscopy analysis was used to visualize the interaction and uptake of AQP4-containing EVs by recipient cells. Chemoinvasion and Caspase3/7 activation assay, performed on recipient cells after EVs uptake, revealed that EVs produced by AQP4-tetramers expressing cells were able to drive surrounding tumour cells toward the migratory phenotype, whereas EVs produced by AQP4-OAPs expressing cells drive them toward the apoptosis pathway. Conclusion: This study demonstrates that the different GBM cell phenotypes can be transferred by AQP4-containing EVs able to influence tumour cell fate toward invasiveness or apoptosis. This study opens a new perspective on the role of AQP4 in the brain tumour microenvironment associated with the EV-dependent communication mechanism. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
180. Glioma stem cells: turpis omen in nomen? (the evil in the name?)
- Author
-
Binda, E, Reynolds, B A, and Vescovi, A L
- Abstract
High-grade gliomas remain incurable and lethal. Through the availability of the stem-like cells responsible for glioblastoma (GB) formation, expansion, resilience and recurrence, the discovery of glioma cancer stem cells (GCSCs) is revolutionizing this field. GCSCs provide an unprecedented opportunity to reproduce and study GB pathophysiology more accurately. This critically emphasizes our ability to unambiguously identify, isolate and investigate cells that do qualify as GCSCs, to use them as a potential model that is truly predictive of GBs and of their regulation and response to therapeutic agents. We review this concept against the background of key findings on somatic, neural and solid tumour stem cells (SCs), also taking into account the emerging phenomenon of phenotypic SC plasticity. We suggest that basic approaches in these areas can be imported into the GCSC field, so that the same functional method used to identify normal somatic SCs becomes the most appropriate to define GCSCs. This, combined with knowledge of the cellular and molecular basis of normal adult neurogenesis, promises to improve the identification of GCSCs and of selective markers, as well as the development of innovative, more specific and efficacious antiglioma strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
181. Growth factor independence underpins a paroxysmal, aggressive Wnt5aHigh/EphA2Low phenotype in glioblastoma stem cells, conducive to experimental combinatorial therapy.
- Author
-
Trivieri, Nadia, Visioli, Alberto, Mencarelli, Gandino, Cariglia, Maria Grazia, Marongiu, Laura, Pracella, Riccardo, Giani, Fabrizio, Soriano, Amata Amy, Barile, Chiara, Cajola, Laura, Copetti, Massimiliano, Palumbo, Orazio, Legnani, Federico, DiMeco, Francesco, Gorgoglione, Leonardo, Vescovi, Angelo L., and Binda, Elena
- Subjects
- *
BRAIN tumors , *GROWTH factors , *STEM cells , *TUMOR markers , *GLIOBLASTOMA multiforme , *PROGNOSIS , *FALSE discovery rate , *PROTEIN-tyrosine kinases - Abstract
Background: Glioblastoma multiforme (GBM) is an incurable tumor, with a median survival rate of only 14–15 months. Along with heterogeneity and unregulated growth, a central matter in dealing with GBMs is cell invasiveness. Thus, improving prognosis requires finding new agents to inhibit key multiple pathways, even simultaneously. A subset of GBM stem-like cells (GSCs) may account for tumorigenicity, representing, through their pathways, the proper cellular target in the therapeutics of glioblastomas. GSCs cells are routinely enriched and expanded due to continuous exposure to specific growth factors, which might alter some of their intrinsic characteristic and hide therapeutically relevant traits. Methods: By removing exogenous growth factors stimulation, here we isolated and characterized a subset of GSCs with a "mitogen-independent" phenotype (I-GSCs) from patient's tumor specimens. Differential side-by-side comparative functional and molecular analyses were performed either in vitro or in vivo on these cells versus their classical growth factor (GF)-dependent counterpart (D-GSCs) as well as their tissue of origin. This was performed to pinpoint the inherent GSCs' critical regulators, with particular emphasis on those involved in spreading and tumorigenic potential. Transcriptomic fingerprints were pointed out by ANOVA with Benjamini-Hochberg False Discovery Rate (FDR) and association of copy number alterations or somatic mutations was determined by comparing each subgroup with a two-tailed Fisher's exact test. The combined effects of interacting in vitro and in vivo with two emerging GSCs' key regulators, such as Wnt5a and EphA2, were then predicted under in vivo experimental settings that are conducive to clinical applications. In vivo comparisons were carried out in mouse-human xenografts GBM model by a hierarchical linear model for repeated measurements and Dunnett's multiple comparison test with the distribution of survival compared by Kaplan–Meier method. Results: Here, we assessed that a subset of GSCs from high-grade gliomas is self-sufficient in the activation of regulatory growth signaling. Furthermore, while constitutively present within the same GBM tissue, these GF-independent GSCs cells were endowed with a distinctive functional and molecular repertoire, defined by highly aggressive Wnt5aHigh/EphA2Low profile, as opposed to Wnt5aLow/EphA2High expression in sibling D-GSCs. Regardless of their GBM subtype of origin, I-GSCs, are endowed with a raised in vivo tumorigenic potential than matched D-GSCs, which were fast-growing ex-vivo but less lethal and invasive in vivo. Also, the malignant I-GSCs' transcriptomic fingerprint faithfully mirrored the original tumor, bringing into evidence key regulators of invasiveness, angiogenesis and immuno-modulators, which became candidates for glioma diagnostic/prognostic markers and therapeutic targets. Particularly, simultaneously counteracting the activity of the tissue invasive mediator Wnt5a and EphA2 tyrosine kinase receptor addictively hindered GSCs' tumorigenic and invasive ability, thus increasing survival. Conclusion: We show how the preservation of a mitogen-independent phenotype in GSCs plays a central role in determining the exacerbated tumorigenic and high mobility features distinctive of GBM. The exploitation of the I-GSCs' peculiar features shown here offers new ways to identify novel, GSCs-specific effectors, whose modulation can be used in order to identify novel, potential molecular therapeutic targets. Furthermore, we show how the combined use of PepA, the anti-Wnt5a drug, and of ephrinA1-Fc to can hinder GSCs' lethality in a clinically relevant xenogeneic in vivo model thus being conducive to perspective, novel combinatorial clinical application. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
182. BRAFV600E mutation impinges on gut microbial markers defining novel biomarkers for serrated colorectal cancer effective therapies.
- Author
-
Trivieri, Nadia, Pracella, Riccardo, Cariglia, Maria Grazia, Panebianco, Concetta, Parrella, Paola, Visioli, Alberto, Giani, Fabrizio, Soriano, Amata Amy, Barile, Chiara, Canistro, Giuseppe, Latiano, Tiziana Pia, Dimitri, Lucia, Bazzocchi, Francesca, Cassano, Dario, Vescovi, Angelo L., Pazienza, Valerio, and Binda, Elena
- Subjects
- *
COLORECTAL cancer , *BIOMARKERS , *EPITHELIAL-mesenchymal transition , *RANDOM forest algorithms , *FALSE discovery rate , *HEREDITARY nonpolyposis colorectal cancer - Abstract
Background: Colorectal cancer (CRC) harboring BRAFV600E mutation exhibits low response to conventional therapy and poorest prognosis. Due to the emerging correlation between gut microbiota and CRC carcinogenesis, we investigated in serrated BRAFV600E cases the existence of a peculiar fecal microbial fingerprint and specific bacterial markers, which might represent a tool for the development of more effective clinical strategies. Methods: By injecting human CRC stem-like cells isolated from BRAFV600E patients in immunocompromised mice, we described a new xenogeneic model of this subtype of CRC. By performing bacterial 16S rRNA sequencing, the fecal microbiota profile was then investigated either in CRC-carrying mice or in a cohort of human CRC subjects. The microbial communities' functional profile was also predicted. Data were compared with Mann-Whitney U, Welch's t-test for unequal variances and Kruskal-Wallis test with Benjamini–Hochberg false discovery rate (FDR) correction, extracted as potential BRAF class biomarkers and selected as model features. The obtained mean test prediction scores were subjected to Receiver Operating characteristic (ROC) analysis. To discriminate the BRAF status, a Random Forest classifier (RF) was employed. Results: A specific microbial signature distinctive for BRAF status emerged, being the BRAF-mutated cases closer to healthy controls than BRAF wild-type counterpart. In agreement, a considerable score of correlation was also pointed out between bacteria abundance from BRAF-mutated cases and the level of markers distinctive of BRAFV600E pathway, including those involved in inflammation, innate immune response and epithelial-mesenchymal transition. We provide evidence that two candidate bacterial markers, Prevotella enoeca and Ruthenibacterium lactatiformans, more abundant in BRAFV600E and BRAF wild-type subjects respectively, emerged as single factors with the best performance in distinguishing BRAF status (AUROC = 0.72 and 0.74, respectively, 95% confidence interval). Furthermore, the combination of the 10 differentially represented microorganisms between the two groups improved performance in discriminating serrated CRC driven by BRAF mutation from BRAF wild-type CRC cases (AUROC = 0.85, 95% confidence interval, 0.69–1.01). Conclusion: Overall, our results suggest that BRAFV600E mutation itself drives a distinctive gut microbiota signature and provide predictive CRC-associated bacterial biomarkers able to discriminate BRAF status in CRC patients and, thus, useful to devise non-invasive patient-selective diagnostic strategies and patient-tailored optimized therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
183. Multifunctionalized hydrogels foster hNSC maturation in 3D cultures and neural regeneration in spinal cord injuries.
- Author
-
Marchini, Amanda, Raspa, Andrea, Pugliese, Raffaele, Malek, Marina Abd El, Pastori, Valentina, Lecchi, Marzia, Vescovi, Angelo L., and Gelain, Fabrizio
- Subjects
- *
HYDROGELS , *DEVELOPMENTAL biology , *NERVOUS system regeneration , *SPINAL cord injuries , *CHOLINERGIC receptor research - Abstract
Three-dimensional cell cultures are leading the way to the fabrication of tissue-like constructs useful to developmental biology and pharmaceutical screenings. However, their reproducibility and translational potential have been limited by biomaterial and culture media compositions, as well as cellular sources. We developed a construct comprising synthetic multifunctionalized hydrogels, serum-free media, and densely seeded good manufacturing practice protocolgrade human neural stem cells (hNSC). We tracked hNSC proliferation, differentiation, and maturation into GABAergic, glutamatergic, and cholinergic neurons, showing entangled electrically active neural networks. The neuroregenerative potential of the “engineered tissue" was assessed in spinal cord injuries, where hNSC-derived progenitors and predifferentiated hNSC progeny, embedded in multifunctionalized hydrogels, were implanted. All implants decreased astrogliosis and lowered the immune response, but scaffolds with predifferentiated hNSCs showed higher percentages of neuronal markers, better hNSC engraftment, and improved behavioral recovery. Our hNSC-construct enables the formation of 3D functional neuronal networks in vitro, allowing novel strategies for hNSC therapies in vivo. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
184. Chronic exposure of astrocytes to interferon-α reveals molecular changes related to Aicardi–Goutières syndrome.
- Author
-
Cuadrado, Eloy, Jansen, Machiel H., Anink, Jasper, De Filippis, Lidia, Vescovi, Angelo L., Watts, Colin, Aronica, Eleonora, Hol, Elly M., and Kuijpers, Taco W.
- Subjects
- *
ASTROCYTES , *INTERFERONS , *AICARDI-Goutieres syndrome , *MICROCEPHALY , *PSYCHOMOTOR disorders , *INTELLECTUAL disabilities - Abstract
Aicardi–Goutières syndrome is a genetically determined infantile encephalopathy, manifesting as progressive microcephaly, psychomotor retardation, and in ∼25% of patients, death in early childhood. Aicardi–Goutières syndrome is caused by mutations in any of the genes encoding TREX1, RNASEH2-A, -B, -C and SAMHD1, with protein dysfunction hypothesized to result in the accumulation of nucleic acids within the cell, thus triggering an autoinflammatory response with increased interferon-α production. Astrocytes have been identified as a major source of interferon-α production in the brains of patients with Aicardi–Goutières syndrome. Here, we study the effect of interferon-α treatment on astrocytes derived from immortalized human neural stem cells. Chronic interferon-α treatment promoted astrocyte activation and a reduction in cell proliferation. Moreover, chronic exposure resulted in an alteration of genes and proteins involved in the stability of white matter (ATF4, eIF2Bα, cathepsin D, cystatin F), an increase of antigen-presenting genes (human leukocyte antigen class I) and downregulation of pro-angiogenic factors and other cytokines (vascular endothelial growth factor and IL-1). Interestingly, withdrawal of interferon-α for 7 days barely reversed these cellular alterations, demonstrating that the interferon-α mediated effects persist over time. We confirmed our in vitro findings using brain samples from patients with Aicardi–Goutières syndrome. Our results support the idea of interferon-α as a key factor in the pathogenesis of Aicardi–Goutières syndrome relating to the observed leukodystrophy and microangiopathy. Because of the sustained interferon-α effect, even after withdrawal, therapeutic targets for Aicardi–Goutières syndrome, and other interferon-α-mediated encephalopathies, may include downstream interferon-α signalling cascade effectors rather than interferon-α alone. [ABSTRACT FROM PUBLISHER]
- Published
- 2013
- Full Text
- View/download PDF
185. The EphA2 Receptor Drives Self-Renewal and Tumorigenicity in Stem-like Tumor-Propagating Cells from Human Glioblastomas
- Author
-
Binda, Elena, Visioli, Alberto, Giani, Fabrizio, Lamorte, Giuseppe, Copetti, Massimiliano, Pitter, Ken L., Huse, Jason T., Cajola, Laura, Zanetti, Nadia, DiMeco, Francesco, De Filippis, Lidia, Mangiola, Annunziato, Maira, Giulio, Anile, Carmelo, De Bonis, Pasquale, Reynolds, Brent A., Pasquale, Elena B., and Vescovi, Angelo L.
- Subjects
- *
EPHRIN receptors , *CELL proliferation , *CARCINOGENICITY , *STEM cells , *TUMOR growth , *GLIOBLASTOMA multiforme , *PROTEIN-tyrosine kinases , *GENE expression - Abstract
Summary: In human glioblastomas (hGBMs), tumor-propagating cells with stem-like characteristics (TPCs) represent a key therapeutic target. We found that the EphA2 receptor tyrosine kinase is overexpressed in hGBM TPCs. Cytofluorimetric sorting into EphA2High and EphA2Low populations demonstrated that EphA2 expression correlates with the size and tumor-propagating ability of the TPC pool in hGBMs. Both ephrinA1-Fc, which caused EphA2 downregulation in TPCs, and siRNA-mediated knockdown of EPHA2 expression suppressed TPCs self-renewal ex vivo and intracranial tumorigenicity, pointing to EphA2 downregulation as a causal event in the loss of TPCs tumorigenicity. Infusion of ephrinA1-Fc into intracranial xenografts elicited strong tumor-suppressing effects, suggestive of therapeutic applications. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
186. The proliferative capacity of the subventricular zone is maintained in the parkinsonian brain.
- Author
-
van den Berge, Simone A., van Strien, Miriam E., Korecka, Joanna A., Dijkstra, Anke A., Sluijs, Jacqueline A., Kooijman, Lieneke, Eggers, Ruben, De Filippis, Lidia, Vescovi, Angelo L., Verhaagen, Joost, van de Berg, Wilma D. J., and Hol, Elly M.
- Subjects
- *
PARKINSONIAN disorders , *NEURAL stem cells , *PARKINSON'S disease , *DEVELOPMENTAL neurobiology , *DOPAMINE , *CEREBRAL ventricles , *CELL proliferation - Abstract
There are many indications that neurogenesis is impaired in Parkinson's disease, which might be due to a lack of dopamine in the subventricular zone. An impairment in neurogenesis may have negative consequences for the development of new therapeutic approaches in Parkinson's disease, as neural stem cells are a potential source for endogenous repair. In this study, we examined the subventricular zone of 10 patients with Parkinson's disease and 10 age- and sex-matched controls for proliferation and neural stem cell numbers. We also included five cases with incidental Lewy body disease, which showed Parkinson's disease pathology but no clinical symptoms and thus did not receive dopaminergic treatment. We quantified the neural stem cell number and proliferative capacity in the subventricular zone of these three donor groups. We found subventricular neural stem cells in each donor, with a high variation in number. We did not observe significant differences in neural stem cell number or in proliferation between the groups. Additionally, we were able to culture neural stem cells from post-mortem brain of several patients with Parkinson's disease, confirming the presence of viable neural stem cells in these brains. We have also examined the subventricular zone of a chronic, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced Parkinson's disease mouse model, and again found no effect of dopaminergic denervation on precursor proliferation. Lastly, we investigated the proliferation capacity of two different human neural stem cell lines in response to dopamine. Both cell lines did not respond with a change in proliferation to treatment with dopamine agonists and an antagonist. In summary, the adult neural stem cell pool in the subventricular zone was not clearly affected in the human parkinsonian brain or a Parkinson's disease mouse model. Furthermore, we did not find evidence that dopamine has a direct effect on human neural stem cell proliferation in vitro. Thus, we conclude that the number of adult neural stem cells is probably not diminished in the parkinsonian brain and that dopamine depletion most likely has no effect on human neural stem cells. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
187. Brain cancer stem cells.
- Author
-
Piccirillo, Sara G. M., Binda, Elena, Fiocco, Roberta, Vescovi, Angelo L., and Shah, Khalid
- Subjects
- *
BRAIN cancer , *CANCER cell growth , *GLIOMAS , *ONCOLOGY , *TUMORS - Abstract
Cancers comprise heterogeneous cells, ranging from highly proliferative immature precursors to more differentiated cell lineages. In the last decade, several groups have demonstrated the existence of cancer stem cells in both nonsolid solid tumors, including some of the brain: glioblastoma multiforme (GBM), medulloblastoma, and ependymoma. These cells, like their normal counterpart in homologous tissues, are multipotent, undifferentiated, self-sustaining, yet transformed cells. In particular, glioblastoma-stem like cells (GBSCs) self-renew under clonal conditions and differentiate into neuron- and glia-like cells, with aberrant, mixed neuronal/astroglial phenotypes. Remarkably, upon subcutaneous and intracerebral transplantation in immunosuppressed mice, GBSCs are able to form secondary tumors that closely resemble the human pathology, a property retained also throughout serial transplantation. The search is up for the identification of the markers and the molecular mechanisms that underpin the tumorigenic potential of these cells. This is critical if we aim at defining new therapeutic approaches for the treatment of malignant brain tumors. Lately, it has been shown that some key regulatory system that plays pivotal roles in neural stem cell physiology can also regulate the tumorigenic ability of cancer stem cells in GBMs. This suggests that the study of cancer stem cells in brain tumors might help to identify new and more specific therapeutic molecular effectors, with the cancer stem cells themselves representing one of the main targets, in fact the Holy Grail, in cancer cell therapy. This review includes a summary review on brain cancer cells and their usefulness as emerging targets in cancer cell therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
188. Distinct pools of cancer stem-like cells coexist within human glioblastomas and display different tumorigenicity and independent genomic evolution.
- Author
-
Piccirillo, S. G. M., Combi, R., Cajola, L., Patrizi, A., Redaelli, S., Bentivegna, A., Baronchelli, S., Maira, G., Pollo, B., Mangiola, A., DiMeco, F., Dalprà, L., and Vescovi, A. L.
- Subjects
- *
STEM cells , *CANCER cells , *GLIOBLASTOMA multiforme , *CARCINOGENESIS , *CYTOGENETICS , *ONCOGENES , *GENOMICS - Abstract
Glioblastomas (GBMs) contain transformed, self-maintaining, multipotent, tumour-initiating cancer stem cells, whose identification has radically changed our perspective on the physiology of these tumours. Currently, it is unknown whether multiple types of transformed precursors, which display alternative sets of the complement of properties of true cancer stem cells, can be found in a GBM. If different subsets of such cancer stem-like cells (CSCs) do exist, they might represent distinct cell targets, with a differential therapeutic importance, also depending on their characteristics and lineage relationship. Here, we report the presence of two types of CSCs within different regions of the same human GBM. Cytogenetic and molecular analysis shows that the two types of CSCs bear quite diverse tumorigenic potential and distinct genetic anomalies, and, yet, derive from common ancestor cells. This provides critical information to unravel the development of CSCs and the key molecular/genetic components underpinning tumorigenicity in human GBMs.Oncogene (2009) 28, 1807–1811; doi:10.1038/onc.2009.27; published online 16 March 2009 [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
189. Bone morphogenetic proteins inhibit the tumorigenic potential of human brain tumour-initiating cells.
- Author
-
Piccirillo, S. G. M., Reynolds, B. A., Zanetti, N., Lamorte, G., Binda, E., Broggi, G., Brem, H., Olivi, A., Dimeco, F., and Vescovi, A. L.
- Subjects
- *
STEM cells , *GLIOBLASTOMA multiforme , *TUMOR growth , *BRAIN stem , *BRAIN , *CELL proliferation - Abstract
Transformed, oncogenic precursors, possessing both defining neural-stem-cell properties and the ability to initiate intracerebral tumours, have been identified in human brain cancers. Here we report that bone morphogenetic proteins (BMPs), amongst which BMP4 elicits the strongest effect, trigger a significant reduction in the stem-like, tumour-initiating precursors of human glioblastomas (GBMs). Transient in vitro exposure to BMP4 abolishes the capacity of transplanted GBM cells to establish intracerebral GBMs. Most importantly, in vivo delivery of BMP4 effectively blocks the tumour growth and associated mortality that occur in 100% of mice after intracerebral grafting of human GBM cells. We demonstrate that BMPs activate their cognate receptors (BMPRs) and trigger the Smad signalling cascade in cells isolated from human glioblastomas (GBMs). This is followed by a reduction in proliferation, and increased expression of markers of neural differentiation, with no effect on cell viability. The concomitant reduction in clonogenic ability, in the size of the CD133+ population and in the growth kinetics of GBM cells indicates that BMP4 reduces the tumour-initiating cell pool of GBMs. These findings show that the BMP–BMPR signalling system—which controls the activity of normal brain stem cells—may also act as a key inhibitory regulator of tumour-initiating, stem-like cells from GBMs and the results also identify BMP4 as a novel, non-cytotoxic therapeutic effector, which may be used to prevent growth and recurrence of GBMs in humans. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
190. Injection of adult neurospheres induces recovery in a chronic model of multiple sclerosis.
- Author
-
Pluchino, Stefano, Quattrini, Angelo, Brambilla, Elena, Gritti, Angela, Salani, Giuliana, Dina, Giorgia, Galli, Rossella, Del Carro, Ubaldo, Amadio, Stefano, Bergami, Alessandra, Furlan, Roberto, Comi, Giancarlo, Vescovi, Angelo L., and Martino, Gianvito
- Subjects
- *
MULTIPLE sclerosis , *CENTRAL nervous system , *DEMYELINATION , *NEUROSCIENCES - Abstract
Widespread demyelination and axonal loss are the pathological hallmarks of multiple sclerosis. The multifocal nature of this chronic inflammatory disease of the central nervous system complicates cellular therapy and puts emphasis on both the donor cell origin and the route of cell transplantation. We established syngenic adult neural stem cell cultures and injected them into an animal model of multiple sclerosis-experimental autoimmune encephalomyelitis (EAE) in the mouse-either intravenously or intracerebroventricularly. In both cases, significant numbers of donor cells entered into demyelinating areas of the central nervous system and differentiated into mature brain cells. Within these areas, oligodendrocyte progenitors markedly increased, with many of them being of donor origin and actively remyelinating axons. Furthermore, a significant reduction of astrogliosis and a marked decrease in the extent of demyelination and axonal loss were observed in transplanted animals. The functional impairment caused by EAE was almost abolished in transplanted mice, both clinically and neurophysiologically. Thus, adult neural precursor cells promote multifocal remyelination and functional recovery after intravenous or intrathecal injection in a chronic model of multiple sclerosis. [ABSTRACT FROM AUTHOR]
- Published
- 2003
- Full Text
- View/download PDF
191. A novel, immortal, and multipotent human neural stem cell line generating functional neurons and Oligodendrocytes
- Author
-
Lidia De Filippis, Antonio Malgaroli, Giuseppe Lamorte, Evan Y. Snyder, Angelo L. Vescovi, DE FILIPPIS, L, Lamorte, G, Snyder, E, Malgaroli, A, Vescovi, A, SNYDER E., Y, Malgaroli, Antonio, and Vescovi, A. L.
- Subjects
neural differentiation ,stem cell culture ,Cellular differentiation ,proliferation ,Genes, myc ,Biology ,Cell therapy ,neural stem cell ,Transformation, Genetic ,Neurosphere ,Humans ,Neural cell ,Cell Line, Transformed ,Cell Proliferation ,Neurons ,Neurotransmitter Agents ,Multipotent Stem Cells ,Cell Differentiation ,Cell Biology ,Anatomy ,Neural stem cell ,Cell biology ,Oligodendroglia ,Phenotype ,Cell culture ,Multipotent Stem Cell ,Molecular Medicine ,Developmental Biology ,Multipotentiality - Abstract
The discovery and study of neural stem cells have revolutionized our understanding of the neurogenetic process, and their inherent ability to adopt expansive growth behavior in vitro is of paramount importance for the development of novel therapeutics based on neural cell replacement. Recent advances in high-throughput assays for drug development and gene discovery dictate the need for rapid, reproducible, long-term expansion of human neural stem cells (hNSCs). In this view, the complement of wild-type cell lines currently available is insufficient. Here we report the establishment of a stable human neural stem cell line (immortalized human NSCs [IhNSCs]) by v-myc-mediated immortalization of previously derived wild-type hNSCs. These cells demonstrate three- to fourfold faster proliferation than wild-type cells in response to growth factors but retain rather similar properties, including multipotentiality. By molecular biology, biochemistry, immunocytochemistry, fluorescence microscopy, and electrophysiology, we show that upon growth factor removal, IhNSCs completely downregulate v-myc expression, cease proliferation, and differentiate terminally into three major neural lineages: astrocytes, oligodendrocytes, and neurons. The latter are functional, mature cells displaying clear-cut morphological and physiological features of terminally differentiated neurons, encompassing mostly the GABAergic, glutamatergic, and cholinergic phenotypes. Finally, IhNSCs produce bona fide oligodendrocytes in fractions up to 20% of total cell number. This is in contrast to the negligible propensity of hNSCs to generate oligodendroglia reported so far. Thus, we describe an immortalized hNSC line endowed with the properties of normal hNSCs and suitable for developing the novel, reliable assays and reproducible high-throughput gene and drug screening that are essential in both diagnostics and cell therapy studies. Disclosure of potential conflicts of interest is found at the end of this article.
- Published
- 2007
192. Robust in vivo gene transfer into adult mammalian neural stem cells by lentiviral vectors
- Author
-
Antonia Follenzi, Claudio Bordignon, Angela Gritti, Fred H. Gage, Antonella Consiglio, Luigi Naldini, Angelo L. Vescovi, D. Dolcetta, Consiglio, A., Gritti, A., Dolcetta, D., Follenzi, A., Bordignon, Claudio, Gage, F. H., Vescovi, A. L., and Naldini, Luigi
- Subjects
Population ,Genetic Vectors ,Cell Culture Techniques ,Subventricular zone ,Fluorescent Antibody Technique ,Biology ,Transfection ,Transduction (genetics) ,Mice ,Genes, Reporter ,medicine ,Animals ,education ,reproductive and urinary physiology ,Neurons ,education.field_of_study ,Multidisciplinary ,Stem Cells ,Lentivirus ,Gene Transfer Techniques ,Brain ,Biological Sciences ,Immunohistochemistry ,Neural stem cell ,Recombinant Proteins ,Olfactory bulb ,Cell biology ,medicine.anatomical_structure ,nervous system ,Immunology ,Stem cell ,biological phenomena, cell phenomena, and immunity ,Adult stem cell - Abstract
Stable genetic modification of adult stem cells is fundamental for both developmental studies and therapeutic purposes. Using in vivo marking studies, we showed that injection of lentiviral vectors (LVs) into the subventricular zone of the adult mouse brain enables efficient gene transfer into long-term self-renewing neural precursors and steady, robust vector expression in their neuronal progeny throughout the subventricular zone and its rostral extension, up to the olfactory bulb. By clonal and population analysis in culture, we proved that in vivo -marked neural precursors display self-renewal and multipotency, two essential characteristics of neural stem cells (NSCs). Thus, LVs efficiently target long-term repopulating adult NSCs, and the effect of the initial transduction is amplified by the continuous generation of NSC-derived, transduced progeny. LVs may thus allow novel studies on NSCs' physiology in vivo , and introduction of therapeutic genes into NSCs may allow the development of novel approaches for untreatable CNS diseases.
- Published
- 2004
193. Human neural stem cells derived from fetal human brain communicate with each other and rescue ischemic neuronal cells through tunneling nanotubes.
- Author
-
Capobianco DL, De Zio R, Profico DC, Gelati M, Simone L, D'Erchia AM, Di Palma F, Mormone E, Bernardi P, Sbarbati A, Gerbino A, Pesole G, Vescovi AL, Svelto M, and Pisani F
- Subjects
- Humans, Brain metabolism, Brain embryology, Cell Differentiation, Nanotubes chemistry, Fetus, Brain Ischemia metabolism, Brain Ischemia pathology, Cell Membrane Structures, Neural Stem Cells metabolism, Neural Stem Cells cytology, Neurons metabolism, Mitochondria metabolism, Coculture Techniques, Cell Communication
- Abstract
Pre-clinical trials have demonstrated the neuroprotective effects of transplanted human neural stem cells (hNSCs) during the post-ischemic phase. However, the exact neuroprotective mechanism remains unclear. Tunneling nanotubes (TNTs) are long plasma membrane bridges that physically connect distant cells, enabling the intercellular transfer of mitochondria and contributing to post-ischemic repair processes. Whether hNSCs communicate through TNTs and their role in post-ischemic neuroprotection remains unknown. In this study, non-immortalized hNSC lines derived from fetal human brain tissues were examined to explore these possibilities and assess the post-ischemic neuroprotection potential of these hNSCs. Using Tau-STED super-resolution confocal microscopy, live cell time-lapse fluorescence microscopy, electron microscopy, and direct or non-contact homotypic co-cultures, we demonstrated that hNSCs generate nestin-positive TNTs in both 3D neurospheres and 2D cultures, through which they transfer functional mitochondria. Co-culturing hNSCs with differentiated SH-SY5Y (dSH-SY5Y) revealed heterotypic TNTs allowing mitochondrial transfer from hNSCs to dSH-SY5Y. To investigate the role of heterotypic TNTs in post-ischemic neuroprotection, dSH-SY5Y were subjected to oxygen-glucose deprivation (OGD) followed by reoxygenation (OGD/R) with or without hNSCs in direct or non-contact co-cultures. Compared to normoxia, OGD/R dSH-SY5Y became apoptotic with impaired electrical activity. When OGD/R dSH-SY5Y were co-cultured in direct contact with hNSCs, heterotypic TNTs enabled the transfer of functional mitochondria from hNSCs to OGD/R dSH-SY5Y, rescuing them from apoptosis and restoring the bioelectrical profile toward normoxic dSH-SY5Y. This complete neuroprotection did not occur in the non-contact co-culture. In summary, our data reveal the presence of a functional TNTs network containing nestin within hNSCs, demonstrate the involvement of TNTs in post-ischemic neuroprotection mediated by hNSCs, and highlight the strong efficacy of our hNSC lines in post-ischemic neuroprotection. Human neural stem cells (hNSCs) communicate with each other and rescue ischemic neurons through nestin-positive tunneling nanotubes (TNTs). A Functional mitochondria are exchanged via TNTs between hNSCs. B hNSCs transfer functional mitochondria to ischemic neurons through TNTs, rescuing neurons from ischemia/reperfusion ROS-dependent apoptosis., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
194. Generation of induced pluripotent stem cells (CSSi017-A)(12862) from an ALS patient carrying a repeat expansion in the C9orf72 gene.
- Author
-
Ruotolo G, D'Anzi A, Casamassa A, Mazzoni M, Ferrari D, Lombardi I, Carletti RM, D'Asdia C, Torrente I, Frezza K, Lattante S, Sabatelli M, Pennuto M, Vescovi AL, and Rosati J
- Subjects
- Humans, Cell Differentiation, Fibroblasts metabolism, Cell Line, Male, Induced Pluripotent Stem Cells metabolism, C9orf72 Protein genetics, C9orf72 Protein metabolism, Amyotrophic Lateral Sclerosis genetics, Amyotrophic Lateral Sclerosis pathology, DNA Repeat Expansion
- Abstract
Genetic expansions of the hexanucleotide repeats (GGGGCC) in the C9orf72 gene appear in approximately 40% of patients with familial ALS and 7% of patients with sporadic ALS in the European population, making this mutation one of the most prevalent genetic mutations in ALS. Here, we generated a human induced pluripotent stem cell (hiPSC) line from the dermal fibroblasts of a patient carrying a 56-repeat expansion in an ALS disease-causing allele of C9orf72. These iPSCs showed stable amplification in vitro with normal karyotype and high expression of pluripotent markers and differentiated spontaneously in vivo into three germ layers., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
195. GFAP serves as a structural element of tunneling nanotubes between glioblastoma cells and could play a role in the intercellular transfer of mitochondria.
- Author
-
Simone L, Capobianco DL, Di Palma F, Binda E, Legnani FG, Vescovi AL, Svelto M, and Pisani F
- Abstract
Tunneling nanotubes (TNTs) are long F-actin-positive plasma membrane bridges connecting distant cells, allowing the intercellular transfer of cellular cargoes, and are found to be involved in glioblastoma (GBM) intercellular crosstalk. Glial fibrillary acid protein (GFAP) is a key intermediate filament protein of glial cells involved in cytoskeleton remodeling and linked to GBM progression. Whether GFAP plays a role in TNT structure and function in GBM is unknown. Here, analyzing F-actin and GFAP localization by laser-scan confocal microscopy followed by 3D reconstruction (3D-LSCM) and mitochondria dynamic by live-cell time-lapse fluorescence microscopy, we show the presence of GFAP in TNTs containing functional mitochondria connecting distant human GBM cells. Taking advantage of super-resolution 3D-LSCM, we show the presence of GFAP-positive TNT-like structures in resected human GBM as well. Using H
2 O2 or the pro-apoptotic toxin staurosporine (STS), we show that GFAP-positive TNTs strongly increase during oxidative stress and apoptosis in the GBM cell line. Culturing GBM cells with STS-treated GBM cells, we show that STS triggers the formation of GFAP-positive TNTs between them. Finally, we provide evidence that mitochondria co-localize with GFAP at the tip of close-ended GFAP-positive TNTs and inside receiving STS-GBM cells. Summarizing, here we found that GFAP is a structural component of TNTs generated by GBM cells, that GFAP-positive TNTs are upregulated in response to oxidative stress and pro-apoptotic stress, and that GFAP interacts with mitochondria during the intercellular transfer. These findings contribute to elucidate the molecular structure of TNTs generated by GBM cells, highlighting the structural role of GFAP in TNTs and suggesting a functional role of this intermediate filament component in the intercellular mitochondria transfer between GBM cells in response to pro-apoptotic stimuli., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The reviewer ME declared a shared affiliation with the authors DC, MS, and FP to the handling editor at the time of review., (Copyright © 2023 Simone, Capobianco, Di Palma, Binda, Legnani, Vescovi, Svelto and Pisani.)- Published
- 2023
- Full Text
- View/download PDF
196. Bone morphogenetic proteins regulate tumorigenicity in human glioblastoma stem cells.
- Author
-
Piccirillo SG and Vescovi AL
- Subjects
- Animals, Flow Cytometry, Humans, Immunohistochemistry, Polymerase Chain Reaction, Bone Morphogenetic Proteins physiology, Brain Neoplasms pathology, Cell Transformation, Neoplastic, Glioblastoma pathology, Stem Cells cytology
- Abstract
Human glioblastomas appear to be established and expanded by cancer stem cells, which are endowed with tumour-initiating and perpetuating ability. We report that bone morphogenetic proteins (BMPs), amongst which BMP4 elicits the strongest effect, activate their cognate receptors (BMPRs) and trigger the Smad but not the MAP38 kinase signalling cascade in cells isolated from human glioblastomas (GBMs). This is followed by a reduction in proliferation and increased expression of differentiated neural markers, without affecting cell viability. The concomitant reduction in the clonogenic ability, both in the size of the CD133+ side population and in the growth kinetics of GBM cells, indicates that BMP4 triggers a reduction in the in vitro cancer stem cell (CSC) pool. Accordingly, transient ex vivo exposure to BMP4 abolishes the capacity of transplanted GBM cells to establish intracerebral GBMs. Most important, in vivo delivery of BMP4 effectively blocks the tumour growth and associated mortality which occur in 100% of control mice in less than 12 weeks, following intracerebral grafting of human GBM cells. These findings show that the BMP-BMPR signalling system, which controls the activity of normal brain stem cells, may also act as a key inhibitory regulator of cancer-initiating, GBM stem-like cells and identifies BMP4 as a novel, non-cytotoxic therapeutic effector, which may be used to prevent growth and recurrence of GBMs in humans.
- Published
- 2006
- Full Text
- View/download PDF
197. Expression and modulation of matrix metalloproteinase-2 and tissue inhibitors of metalloproteinases in human embryonic CNS stem cells.
- Author
-
Frölichsthal-Schoeller P, Vescovi AL, Krekoski CA, Murphy G, Edwards DR, and Forsyth P
- Subjects
- Cells, Cultured, Collagenases genetics, Collagenases metabolism, Embryo, Mammalian cytology, Embryo, Mammalian metabolism, Gelatinases genetics, Humans, Matrix Metalloproteinase 2, Matrix Metalloproteinase 9, Metalloendopeptidases genetics, RNA, Messenger metabolism, Tissue Inhibitor of Metalloproteinases genetics, Diencephalon embryology, Gelatinases metabolism, Metalloendopeptidases metabolism, Stem Cells metabolism, Tissue Inhibitor of Metalloproteinases metabolism
- Abstract
The expression and regulation of matrix metalloproteinases (MMPs) and tissue inhibitors of MMPs (TIMPs) in neuroectodermal precursor cells is undocumented. We report the presence of MMP-2, but no MMP-9, and of all the four known TIMPs in neuroepithelial stem cells isolated from the human CNS. The expression of TIMP-1, TIMP-2 and TIMP-3 was unchanged following stem cells differentiation into neurons and glia. In contrast, while MMP-2 and TIMP-4 were localized to both stem and mature CNS cells, their levels of expression were substantially reduced in the latter. TIMP-4 showed a 23-fold reduction in media conditioned by differentiated cells compared with stem cell-conditioned media, reflecting a 6-fold decrease in mRNA expression. Interestingly, TIMP-4 also differed from the other TIMPs in that it was cell-associated in the stem cells, where this fraction remained unchanged upon differentiation. Hence, regulation of selective MMPs and TIMPs occurs during differentiation of human neural precursors suggesting that MMP-2 and TIMP-4 in particular may perform regulatory roles in the developing CNS.
- Published
- 1999
- Full Text
- View/download PDF
198. Steroid metabolizing enzymes in pluripotential progenitor central nervous system cells: effect of differentiation and maturation.
- Author
-
Melcangi RC, Froelichsthal P, Martini L, and Vescovi AL
- Subjects
- 20-alpha-Dihydroprogesterone pharmacology, 3-Hydroxysteroid Dehydrogenases metabolism, 3-Oxo-5-alpha-Steroid 4-Dehydrogenase metabolism, Androgens pharmacology, Animals, Cell Differentiation drug effects, Cell Differentiation physiology, Cells, Cultured, Central Nervous System drug effects, Female, Fluorescent Antibody Technique, Direct, Kinetics, Mice, Neostriatum cytology, Neostriatum drug effects, Neostriatum growth & development, Neuroglia drug effects, Neuroglia enzymology, Neuroglia ultrastructure, Neurons drug effects, Neurons enzymology, Neurons ultrastructure, Pregnancy, Progestins pharmacology, Stem Cells drug effects, Central Nervous System cytology, Central Nervous System enzymology, Stem Cells enzymology, Steroids metabolism
- Abstract
A novel in vitro system which allows extensive culturing of multipotential stem cells from mouse brain has made it possible to test whether enzymes that metabolize androgens and progestagens are present in undifferentiated central nervous system progenitor cells. Embryonic day 14 striatal cells were grown in the presence of either 20 ng/ml of epidermal growth factor (which prevents cell differentiation), or 2% fetal bovine serum (facilitating differentiation). Differentiation was complete by 35 days in vitro when the cell population comprised 86 +/- 2.0% astrocytes, 6 +/- 0.7% neurons 1.6 +/- 0.5% oligodendrocytes and 6.4 +/- 0.5% undifferentiated cells. No changes in the proportions of cell type were observed thereafter (38 and 45 days in vitro). 5 alpha-Reductive conversion (by 5 alpha-reductase) of testosterone and progesterone into dihydrotestosterone and dihydroprogesterone, and subsequent 3-alpha hydroxylation (by 3 alpha-hydroxysteroid dehydrogenase) to 3 alpha-diol and tetrahydroprogesterone were assayed in the cultures at 35, 38 and 45 days in vitro. Undifferentiated epidermal growth factor-treated cells (controls) formed about 10 times more dihydroprogesterone than dihydrotestosterone. Conversions of dihydrotestosterone and dihydroprogesterone, respectively, into 3 alpha-diol and tetrahydroprogesterone were very similar. In the fetal bovine serum-treated differentiating cells, 5 alpha-reductase converting progesterone increased at 38 days in vitro, and remained similarly elevated at 42 days in vitro (4 times). However, the conversion of testosterone into dihydrotestosterone remained at control levels up to 42 days in vitro when an increase was observed. 3 alpha-Hydroxysteroid dehydrogenase activity converting dihydroprogesterone but not dihydrotestosterone was increased at 38 and 42 days in vitro. These results show that undifferentiated central nervous system cells possess androgen and progestagen metabolizing enzymes which are strongly influenced by the cellular differentiation/maturation process.
- Published
- 1996
- Full Text
- View/download PDF
199. Multipotential stem cells from the adult mouse brain proliferate and self-renew in response to basic fibroblast growth factor.
- Author
-
Gritti A, Parati EA, Cova L, Frolichsthal P, Galli R, Wanke E, Faravelli L, Morassutti DJ, Roisen F, Nickel DD, and Vescovi AL
- Subjects
- Action Potentials, Animals, Biomarkers, Cell Differentiation drug effects, Cell Division drug effects, Cell Lineage, Cells, Cultured, Clone Cells, Mice, Neuroglia cytology, Neurons cytology, Neurotransmitter Agents metabolism, Phenotype, Stem Cells cytology, Corpus Striatum cytology, Fibroblast Growth Factor 2 pharmacology, Stem Cells drug effects
- Abstract
It has been established that the adult mouse forebrain contains multipotential (neuronal/glial) progenitor cells that can be induced to proliferate in vitro when epidermal growth factor is provided. These cells are found within the subventricular zone of the lateral ventricles, together with other progenitor cell populations, whose requirements for proliferation remain undefined. Using basic fibroblast growth factor (bFGF), we have isolated multipotential progenitors from adult mouse striatum. These progenitors proliferate and can differentiate into cells displaying the antigenic properties of astrocytes, oligodendrocytes, and neurons. The neuron-like cells possess neuronal features, exhibit neuronal electrophysiological properties, and are immunoreactive for GABA, substance P, choline acetyl-transferase, and glutamate. Clonal analysis confirmed the multipotency of these bFGF-dependent cells. Most significantly, subcloning experiments demonstrated that they were capable of self-renewal, which led to a progressive increase in population size over serial passaging. These results demonstrate that bFGF is mitogenic for multipotential cells from adult mammalian forebrain that possess stem cell properties.
- Published
- 1996
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.