Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 115(37): E8765-E8774, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30150378

ABSTRACT

Huntington disease (HD) is an autosomal dominant neurodegenerative disorder caused by expanded CAG repeats in the huntingtin gene (HTT). Although mutant HTT is expressed during embryonic development and throughout life, clinical HD usually manifests later in adulthood. A number of studies document neurodevelopmental changes associated with mutant HTT, but whether these are reversible under therapy remains unclear. Here, we identify very early behavioral, molecular, and cellular changes in preweaning transgenic HD rats and mice. Reduced ultrasonic vocalization, loss of prepulse inhibition, and increased risk taking are accompanied by disturbances of dopaminergic regulation in vivo, reduced neuronal differentiation capacity in subventricular zone stem/progenitor cells, and impaired neuronal and oligodendrocyte differentiation of mouse embryo-derived neural stem cells in vitro. Interventional treatment of this early phenotype with the histone deacetylase inhibitor (HDACi) LBH589 led to significant improvement in behavioral changes and markers of dopaminergic neurotransmission and complete reversal of aberrant neuronal differentiation in vitro and in vivo. Our data support the notion that neurodevelopmental changes contribute to the prodromal phase of HD and that early, presymptomatic intervention using HDACi may represent a promising novel treatment approach for HD.


Subject(s)
Cell Differentiation/drug effects , Huntington Disease/physiopathology , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Neurons/drug effects , Animals , Animals, Genetically Modified , Cell Differentiation/genetics , Cell Differentiation/physiology , Disease Models, Animal , Female , Histone Deacetylase Inhibitors/pharmacology , Humans , Huntingtin Protein/genetics , Huntington Disease/genetics , Lateral Ventricles/pathology , Male , Mice, Transgenic , Mutation , Neurons/metabolism , Neurons/physiology , Panobinostat , Rats
2.
J Neurosci ; 33(39): 15603-17, 2013 Sep 25.
Article in English | MEDLINE | ID: mdl-24068827

ABSTRACT

Glioblastoma (GBM) remains the most pervasive and lethal of all brain malignancies. One factor that contributes to this poor prognosis is the highly invasive character of the tumor. GBM is characterized by microscopic infiltration of tumor cells throughout the brain, whereas non-neural metastases, as well as select lower grade gliomas, develop as self-contained and clearly delineated lesions. Illustrated by rodent xenograft tumor models as well as pathological human patient specimens, we present evidence that one fundamental switch between these two distinct pathologies--invasion and noninvasion--is mediated through the tumor extracellular matrix. Specifically, noninvasive lesions are associated with a rich matrix containing substantial amounts of glycosylated chondroitin sulfate proteoglycans (CSPGs), whereas glycosylated CSPGs are essentially absent from diffusely infiltrating tumors. CSPGs, acting as central organizers of the tumor microenvironment, dramatically influence resident reactive astrocytes, inducing their exodus from the tumor mass and the resultant encapsulation of noninvasive lesions. Additionally, CSPGs induce activation of tumor-associated microglia. We demonstrate that the astrogliotic capsule can directly inhibit tumor invasion, and its absence from GBM presents an environment favorable to diffuse infiltration. We also identify the leukocyte common antigen-related phosphatase receptor (PTPRF) as a putative intermediary between extracellular glycosylated CSPGs and noninvasive tumor cells. In all, we present CSPGs as critical regulators of brain tumor histopathology and help to clarify the role of the tumor microenvironment in brain tumor invasion.


Subject(s)
Brain Neoplasms/metabolism , Chondroitin Sulfate Proteoglycans/metabolism , Glioma/metabolism , Tumor Microenvironment , Adult , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cells, Cultured , Child , Chondroitin Sulfate Proteoglycans/genetics , Female , Glioma/pathology , Glycosylation , Humans , Male , Mice , Microglia/metabolism , Microglia/pathology , Middle Aged , Neoplasm Invasiveness , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Xenograft Model Antitumor Assays
3.
J Neurooncol ; 117(1): 15-24, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24510433

ABSTRACT

Glioblastoma (GBM) is the most common malignant adult brain tumor and carries a poor prognosis due to primary and acquired resistance. While many cellular features of GBM have been documented, it is unclear if cells within these tumors extend a primary cilium, an organelle whose associated signaling pathways may regulate proliferation, migration, and survival of neural precursor and tumor cells. Using immunohistochemical and electron microscopy (EM) techniques, we screened human GBM tumor biopsies and primary cell lines for cilia. Immunocytochemical staining of five primary GBM cell lines revealed that between 8 and 25 % of the cells in each line possessed gamma tubulin-positive basal bodies from which extended acetylated, alpha-tubulin-positive axonemes. EM analyses confirmed the presence of cilia at the cell surface and revealed that their axonemes contained organized networks of microtubules, a structural feature consistent with our detection of IFT88 and Arl13b, two trafficked cilia proteins, along the lengths of the axonemes. Notably, cilia were detected in each of 23 tumor biopsies (22 primary and 1 recurrent) examined. These cilia were distributed across the tumor landscape including regions proximal to the vasculature and within necrotic areas. Moreover, ciliated cells within these tumors co-stained with Ki67, a marker for actively dividing cells, and ZEB1, a transcription factor that is upregulated in GBM and linked to tumor initiation, invasion, and chemoresistance. Collectively, our data show that subpopulations of cells within human GBM tumors are ciliated. In view of mounting evidence supporting roles of primary cilia in tumor initiation and propagation, it is likely that further study of the effects of cilia on GBM tumor cell function will improve our understanding of GBM pathogenesis and may provide new directions for GBM treatment strategies.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/ultrastructure , Cilia/ultrastructure , Glioblastoma/metabolism , Glioblastoma/ultrastructure , ADP-Ribosylation Factors/metabolism , Aged, 80 and over , Axoneme/metabolism , Axoneme/ultrastructure , Basal Bodies/metabolism , Basal Bodies/ultrastructure , Cell Line, Tumor , Cilia/metabolism , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Ki-67 Antigen/metabolism , Male , Microscopy, Electron , Middle Aged , Transcription Factors/metabolism , Tubulin/metabolism , Tumor Suppressor Proteins/metabolism , Zinc Finger E-box-Binding Homeobox 1
4.
J Control Release ; 372: 433-445, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908756

ABSTRACT

Transdifferentiation (TD), a somatic cell reprogramming process that eliminates pluripotent intermediates, creates cells that are ideal for personalized anti-cancer therapy. Here, we provide the first evidence that extracellular vesicles (EVs) from TD-derived induced neural stem cells (Exo-iNSCs) are an efficacious treatment strategy for brain cancer. We found that genetically engineered iNSCs generated EVs loaded with the tumoricidal gene product TRAIL at nearly twice the rate of their parental fibroblasts, and TRAIL produced by iNSCs was naturally loaded into the lumen of EVs and arrayed across their outer membrane (Exo-iNSC-TRAIL). Uptake studies in ex vivo organotypic brain slice cultures showed that Exo-iNSC-TRAIL selectively accumulates within tumor foci, and co-culture assays demonstrated that Exo-iNSC-TRAIL killed metastatic and primary brain cancer cells more effectively than free TRAIL. In an orthotopic mouse model of brain cancer, Exo-iNSC-TRAIL reduced breast-to-brain tumor xenografts by approximately 3000-fold compared to treatment with free TRAIL, with all Exo-iNSC-TRAIL treated animals surviving through 90 days post-treatment. In additional in vivo testing against aggressive U87 and invasive GBM8 glioblastoma tumors, Exo-iNSC-TRAIL also induced a statistically significant increase in survival. These studies establish a novel, easily generated, stable, tumor-targeted EV to efficaciously treat multiple forms of brain cancer.


Subject(s)
Brain Neoplasms , Exosomes , Neural Stem Cells , TNF-Related Apoptosis-Inducing Ligand , Animals , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/genetics , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Exosomes/metabolism , Humans , Cell Line, Tumor , Female , Mice , Mice, Nude
5.
bioRxiv ; 2024 May 29.
Article in English | MEDLINE | ID: mdl-38854085

ABSTRACT

Transdifferentiation (TD), a somatic cell reprogramming process that eliminates pluripotent intermediates, creates cells that are ideal for personalized anti-cancer therapy. Here, we provide the first evidence that extracellular vesicles (EVs) from TD-derived induced neural stem cells (Exo-iNSCs) are an efficacious treatment strategy for brain cancer. We found that genetically engineered iNSCs generated EVs loaded with the tumoricidal gene product TRAIL at nearly twice the rate as their parental fibroblasts, and the TRAIL produced by iNSCs were naturally loaded into the lumen of EVs and arrayed across their outer membrane (Exo-iNSC-TRAIL). Uptake studies in ex vivo organotypic brain slice cultures showed Exo-iNSC-TRAIL selectively accumulates within tumor foci, and co-culture assays showed that Exo-iNSC-TRAIL killed metastatic and primary brain cancer cells more effectively than free TRAIL. In an orthotopic mouse model of brain cancer, Exo-iNSC-TRAIL reduced breast-to-brain tumor xenografts around 3000-fold greater than treatment with free TRAIL, with all Exo-iNSC-TRAIL treated animals surviving through 90 days post-treatment. In additional in vivo testing against aggressive U87 and invasive GBM8 glioblastoma tumors, Exo-iNSC-TRAIL also induced a statistically significant increase in survival. These studies establish a new easily generated, stable, tumor-targeted EV to efficaciously treat multiple forms of brain cancer.

6.
Nat Med ; 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39085419

ABSTRACT

The ecosystem of brain tumors is considered immunosuppressed, but our current knowledge may be incomplete. Here we analyzed clinical cell and tissue specimens derived from patients presenting with glioblastoma or nonmalignant intracranial disease to report that the cranial bone (CB) marrow, in juxtaposition to treatment-naive glioblastoma tumors, harbors active lymphoid populations at the time of initial diagnosis. Clinical and anatomical imaging, single-cell molecular and immune cell profiling and quantification of tumor reactivity identified CD8+ T cell clonotypes in the CB that were also found in the tumor. These were characterized by acute and durable antitumor response rooted in the entire T cell developmental spectrum. In contrast to distal bone marrow, the CB niche proximal to the tumor showed increased frequencies of tumor-reactive CD8+ effector types expressing the lymphoid egress marker S1PR1. In line with this, cranial enhancement of CXCR4 radiolabel may serve as a surrogate marker indicating focal association with improved progression-free survival. The data of this study advocate preservation and further exploitation of these cranioencephalic units for the clinical care of glioblastoma.

7.
Mod Pathol ; 25(2): 157-62, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22056951

ABSTRACT

The presence of stem and progenitor cells in the adult human brain suggests a putative and persistent role in reparative behaviors following neurological injury and neurological disease. Too few stem/progenitor cells (as in the case of Parkinson's disease) or too many of these cells (as in the case of Huntington's disease and glioma) could contribute to and even signal brain pathology. We address here critical issues faced by the field of stem cell biology and regenerative medicine, arguing from well-documented as well as speculative perspectives for a potential role for stem cells in the pathology of many human neurological diseases. Although stem cell responses may result in regenerative failure, in many cases they may help in the establishment or re-establishment of a functional neural circuitry (eg, after stroke). Therefore, we would argue that stem cells have a crucial-either positive or negative-role in the pathology of many neurological diseases.


Subject(s)
Nervous System Diseases/pathology , Stem Cells/pathology , Animals , Humans
8.
Mol Cell Neurosci ; 47(1): 61-70, 2011 May.
Article in English | MEDLINE | ID: mdl-21420496

ABSTRACT

Cerebellar Purkinje neurons (PNs) possess a well characterized propensity to fuse with bone marrow-derived cells (BMDCs), producing heterokaryons with Purkinje cell identities. This offers the potential to rescue/repair at risk or degenerating PNs in the inherited ataxias, including Spinocerebellar Ataxia 1 (SCA1), by introducing therapeutic factors through BMDCs to potentially halt or reverse disease progression. In this study, we combined gene therapy and a stem cell-based treatment to attempt repair of at-risk PNs through cell-cell fusion in a Sca1(154Q/2Q) knock-in mouse model. BMDCs enriched for the hematopoietic stem cell (HSC) population were genetically modified using adeno-associated viral vector 7 (AAV7) to carry SCA1 modifier genes and transplanted into irradiated Sca1(154Q/2Q) mice. Binucleated Purkinje heterokaryons with sex-mismatched donor Y chromosomes were detected and successfully expressed the modifier genes in vivo. Potential effects of the new genome within Purkinje heterokaryons were evaluated using nuclear inclusions (NIs) as a biological marker to reflect possible modifications of the SCA1 disease process. An overall decrease in number of NIs and an increase in the number of surviving PNs were observed in treated Sca1(154Q/2Q). Furthermore, Bergmann glia were found to have fusogenic potential with the donor population and reveal another potential route of therapeutic entry into at-risk cells of the SCA1 cerebellum. This study presents a first step towards a proof-of-principle that combines somatic cellular fusion events with a neuroprotective gene therapy approach for providing potential neuronal protection/repair in a variety of neurodegenerative disorders.


Subject(s)
Cell Fusion , Gene Transfer Techniques , Purkinje Cells/physiology , Spinocerebellar Ataxias/therapy , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Female , Gene Knock-In Techniques , HEK293 Cells , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Humans , Male , Mice , Mice, Transgenic , Purkinje Cells/cytology , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/pathology
9.
Ann Neurol ; 68(2): 264-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20695020

ABSTRACT

Residual tumor cells remain beyond the margins of every glioblastoma (GBM) resection. Their resistance to postsurgical therapy is considered a major driving force of mortality, but their biology remains largely uncharacterized. In this study, residual tumor cells were derived via experimental biopsy of the resection margin after standard neurosurgery for direct comparison with samples from the routinely resected tumor tissue. In vitro analysis of proliferation, invasion, stem cell qualities, GBM-typical antigens, genotypes, and in vitro drug and irradiation challenge studies revealed these cells as unique entities. Our findings suggest a need for characterization of residual tumor cells to optimize diagnosis and treatment of GBM.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Adult , Aged , Antigens, Neoplasm/metabolism , Brain Neoplasms/drug therapy , Cell Culture Techniques , Cell Proliferation , Cell Separation , Female , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Male , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured
10.
Brain ; 133(11): 3359-72, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20719879

ABSTRACT

The hippocampal dentate gyrus maintains its capacity to generate new neurons throughout life. In animal models, hippocampal neurogenesis is increased by cognitive tasks, and experimental ablation of neurogenesis disrupts specific modalities of learning and memory. In humans, the impact of neurogenesis on cognition remains unclear. Here, we assessed the neurogenic potential in the human hippocampal dentate gyrus by isolating adult human neural stem cells from 23 surgical en bloc hippocampus resections. After proliferation of the progenitor cell pool in vitro we identified two distinct patterns. Adult human neural stem cells with a high proliferation capacity were obtained in 11 patients. Most of the cells in the high proliferation capacity cultures were capable of neuronal differentiation (53 ± 13% of in vitro cell population). A low proliferation capacity was observed in 12 specimens, and only few cells differentiated into neurons (4 ± 2%). This was reflected by reduced numbers of proliferating cells in vivo as well as granule cells immunoreactive for doublecortin, brain-derived neurotrophic factor and cyclin-dependent kinase 5 in the low proliferation capacity group. High and low proliferation capacity groups differed dramatically in declarative memory tasks. Patients with high proliferation capacity stem cells had a normal memory performance prior to epilepsy surgery, while patients with low proliferation capacity stem cells showed severe learning and memory impairment. Histopathological examination revealed a highly significant correlation between granule cell loss in the dentate gyrus and the same patient's regenerative capacity in vitro (r = 0.813; P < 0.001; linear regression: R²(adjusted) = 0.635), as well as the same patient's ability to store and recall new memories (r = 0.966; P = 0.001; linear regression: R²(adjusted) = 0.9). Our results suggest that encoding new memories is related to the regenerative capacity of the hippocampus in the human brain.


Subject(s)
Adult Stem Cells/cytology , Cell Proliferation , Hippocampus/cytology , Memory Disorders/pathology , Adult , Adult Stem Cells/physiology , Age Factors , Cell Differentiation/physiology , Cells, Cultured , Female , Hippocampus/physiology , Humans , Male , Memory Disorders/psychology , Middle Aged , Random Allocation , Young Adult
11.
J Urol ; 183(5): 2045-53, 2010 May.
Article in English | MEDLINE | ID: mdl-20303530

ABSTRACT

PURPOSE: We identified a discrete population of stem cell-like tumor cells expressing 5 essential transcription factors required to reprogram pluripotency in prostate tumor cell lines and primary prostate cancer tissue. MATERIALS AND METHODS: DU145 and PC3 human prostate cancer cell lines (ATCC), tumor tissue from patients with prostate cancer and normal prostate tissue were evaluated for the reprogramming factors OCT3/4 (Cell Signaling Technology), SOX2, Klf4 (Santa Cruz Biotechnology, Santa Cruz, California), Nanog (BioLegend) and c-Myc (Cell Signaling) by semiquantitative reverse transcriptase-polymerase chain reaction, histological and immunohistochemical analysis. Stem cell-like tumor cells were enriched by flow cytometric cell sorting using E-cadherin (R&D Systems) as a surface marker, and soft agar, spheroid and tumorigenicity assays to confirm cancer stem cell-like characteristics. RESULTS: mRNA expression of transcription factors OCT3/4 and SOX2 highly correlated in primary prostate tumor tissue samples. The number of OCT3/4 or SOX2 expressing cells was significantly increased in prostate cancer tissue compared to that in normal prostate or benign prostate hyperplasia tissue (p <0.05). When isolated from the DU145 and PC3 prostate cancer cell lines by flow cytometry, stem cell-like tumor cells expressing high OCT3/4 and SOX2 levels showed high tumorigenicity in immunodeficient mice. In vivo growth of the parental DU145 and PC3 prostate cancer cell lines was inhibited by short hairpin RNA knockdown of OCT3/4 or SOX2. CONCLUSIONS: Data suggest that prostate tumor cells expressing pluripotent stem cell transcription factors are highly tumorigenic. Identifying such cells and their importance in prostate cancer growth could provide opportunities for novel targeting strategies for prostate cancer therapy.


Subject(s)
Pluripotent Stem Cells/metabolism , Prostatic Neoplasms/metabolism , Transcription Factors/metabolism , Adult , Analysis of Variance , Animals , Blotting, Western , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Flow Cytometry , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Mice, SCID , Middle Aged , Nanog Homeobox Protein , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Prostatic Neoplasms/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transcription Factors/genetics , Tumor Cells, Cultured
12.
Stem Cells ; 27(7): 1625-34, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19544457

ABSTRACT

Numerous studies have explored the potential of different stem and progenitor cells to replace at-risk neuronal populations in a variety of neurodegenerative disease models. This study presents data from a side-by-side approach of engrafting two different stem/progenitor cell populations within the postnatal cerebellum of the weaver neurological mutant mouse--cerebellar-derived multipotent astrocytic stem cells and embryonic stem cell-derived neural precursors--for comparative analysis. We show here that both donor populations survive, migrate, and appear to initiate differentiation into neurons within the granuloprival host environment. Neither of these disparate stem/progenitor cell populations adopted significant region-specific identities, despite earlier studies that suggested the potential of these cells to respond to in vivo cues when placed in a permissive/instructive milieu. However, data presented here suggest that molecular and cellular deficits present within weaver homozygous or heterozygous brains may promote a slightly more positive donor cell response toward acquisition of a neuronal phenotype. Hence, it is likely that a fine balance exists between a compromised host environment that is amenable to cell replacement and that of a degenerating cellular milieu where it is perhaps too deleterious to support extensive neuronal differentiation and functional cellular integration. These findings join a growing list of studies that show successful cell replacement depends largely on the interplay between the potentiality of the donor cells and the specific pathological conditions of the recipient environment, and that emergent therapies for neurological disorders involving the use of neural stem cells still require refinement.


Subject(s)
Cerebellum/cytology , Embryonic Stem Cells/cytology , Neurons/cytology , Stem Cell Transplantation/methods , Stem Cells/cytology , Animals , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Cell Movement/physiology , Cells, Cultured , Cerebellum/metabolism , Embryonic Stem Cells/metabolism , Mice , Mice, Neurologic Mutants , Mice, Transgenic , Neurons/metabolism , Stem Cells/metabolism
13.
Stem Cells ; 27(2): 280-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18988710

ABSTRACT

An increasing body of evidence suggests that astrocytic gliomas of the central nervous system may be derived from gliotypic neural stem cells. To date, the study of these tumors, particularly the identification of originating cellular population(s), has been frustrated by technical difficulties in accessing the native niche of stem cells. To identify any hallmark signs of cancer in neural stem cells or their progeny, we cultured subventricular zone-derived tissue in a unique in vitro model that temporally and phenotypically recapitulates adult neurogenesis. Contrary to some reports, we found undifferentiated neural stem cells possess few characteristics, suggesting prototumorigenic potential. However, when induced to differentiate, neural stem cells give rise to intermediate progenitors that transiently exhibit multiple glioma characteristics, including aneuploidy, loss of growth-contact inhibition, alterations in cell cycle, and growth factor insensitivity. Further examination of progenitor populations revealed a subset of cells defined by the aberrant expression of (the pathological glioma marker) class III beta-tubulin that exhibit intrinsic parental properties of gliomas, including multilineage differentiation and continued proliferation in the absence of a complex cellular regulatory environment. As tumorigenic characteristics in progenitor cells normally disappear with the generation of mature progeny, this suggests that developmentally intermediate progenitor cells, rather than neural stem cells, may be the origin of so-called "stem cell-derived" tumors.


Subject(s)
Cell Differentiation/physiology , Glioma/pathology , Neurons/cytology , Stem Cells/pathology , Animals , Astrocytes/cytology , Astrocytes/metabolism , Biomarkers, Tumor/metabolism , Blotting, Western , Cells, Cultured , Flow Cytometry , Glioma/metabolism , Immunohistochemistry , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Polymerase Chain Reaction , Stem Cells/metabolism , Stem Cells/ultrastructure , Tubulin/metabolism
14.
J Comp Neurol ; 528(7): 1203-1215, 2020 05.
Article in English | MEDLINE | ID: mdl-31743443

ABSTRACT

Extracellular vesicles, including exosomes/microvesicles (EMVs), have been described as sensitive biomarkers that represent disease states and response to therapies. In light of recent reports of disease-mirroring EMV molecular signatures, the present study profiled two EMVs from different Parkinson's disease (PD) tissue sources: (a) neural progenitor cells derived from an endogenous adult stem/progenitor cell, called adult human neural progenitor (AHNP) cells, that we found to be pathological when isolated from postmortem PD patients' substantia nigra; and (b) leucine-rich repeat kinase 2 (LRRK2) gene identified patient induced pluripotent stem cells (iPSCs), which were used to isolate EMVs and begin to characterize their cargoes. Initial characterization of EMVs derived from idiopathic patients (AHNPs) and mutant LRRK2 patients showed differences between both phenotypes and when compared with a sibling control in EMV size and release based on Nanosight analysis. Furthermore, molecular profiling disclosed that neurodegenerative-related gene pathways altered in PD can be reversed using gene-editing approaches. In fact, the EMV cargo genes exhibited normal expression patterns after gene editing. This study shows that EMVs have the potential to serve as sensitive biomarkers of disease state in both idiopathic and gene-identified PD patients and that following gene-editing, EMVs reflect a corrected state. This is relevant for both prodromal and symptomatic patient populations where potential responses to therapies can be monitored via non-invasive liquid biopsies and EMV characterizations.


Subject(s)
Biomarkers , Exosomes/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Parkinson Disease , Exosomes/pathology , Humans , Induced Pluripotent Stem Cells , Mutation , Neural Stem Cells , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Transcriptome
15.
Stem Cells ; 26(12): 3218-27, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18802036

ABSTRACT

Bromodeoxyuridine (BrdU) is a halogenated pyrimidine that incorporates into newly synthesized DNA during the S phase. BrdU is used ubiquitously in cell birthdating studies and as a means of measuring the proliferative index of various cell populations. In the absence of secondary stressors, BrdU is thought to incorporate relatively benignly into replicating DNA chains. However, we report here that a single, low-dose pulse of BrdU exerts a profound and sustained antiproliferative effect in cultured murine stem and progenitor cells. This is accompanied by altered terminal differentiation, cell morphology, and protein expression consistent with the induction of senescence. There is no evidence of a significant increase in spontaneous cell death; however, cells are rendered resistant to chemically induced apoptosis. Finally, we show that a brief in vivo BrdU regimen reduces the proliferative potential of subsequently isolated subependymal zone neurosphere-forming cells. We conclude, therefore, that BrdU treatment induces a senescence pathway that causes a progressive decline in the replication of rapidly dividing stem/progenitor cells, suggesting a novel and uncharacterized effect of BrdU. This finding is significant in that BrdU-incorporating neural stem/progenitor cells and their progeny should not be expected to behave normally with respect to proliferative potential and downstream functional parameters. This effect highlights the need for caution when results based on long-term BrdU tracking over multiple rounds of replication are interpreted. Conversely, the reliable induction of senescence in stem/progenitor cells in vitro and in vivo may yield a novel platform for molecular studies designed to address multiple aspects of aging and neurogenesis.


Subject(s)
Bromodeoxyuridine/pharmacology , Neurons/cytology , Stem Cells/cytology , Animals , Apoptosis , Astrocytes/metabolism , Cell Proliferation , Cells, Cultured , Cellular Senescence , Mice , Mice, Inbred C57BL , Neurons/metabolism , Time Factors , beta-Galactosidase/metabolism
16.
Int J Dev Neurosci ; 78: 49-64, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31421150

ABSTRACT

Adult human neural progenitor and stem cells have been implicated as a potential source of brain cancer causing cells, but specific events that might cause cells to progress towards a transformed phenotype remain unclear. The L1CAM (L1) cell adhesion/recognition molecule is expressed abnormally by human glioma cancer cells and is released as a large extracellular ectodomain fragment, which stimulates cell motility and proliferation. This study investigates the effects of ectopic overexpression of the L1 long ectodomain (L1LE; ˜180 kDa) on the motility, proliferation, and differentiation of human neural progenitor cells (HNPs). L1LE was ectopically expressed in HNPs using a lentiviral vector. Surprisingly, overexpression of L1LE resulted in reduced HNP motility in vitro, in stark contrast to the effects on glioma and other cancer cell types. L1LE overexpression resulted in a variable degree of maintenance of HNP proliferation in media without added growth factors but did not increase proliferation. In monolayer culture, HNPs expressed a variety of differentiation markers. L1LE overexpression resulted in loss of glutamine synthetase (GS) and ß3-tubulin expression in normal HNP media, and reduced vimentin and increased GS expression in the absence of added growth factors. When co-cultured with chick embryonic brain cell aggregates, HNPs show increased differentiation potential. Some HNPs expressed p-neurofilaments and oligodendrocytic O4, indicating differentiation beyond that in monolayer culture. Most HNP-L1LE cells lost their vimentin and GFAP (glial fibrillary acidic protein) staining, and many cells were positive for astrocytic GS. However, these cells rarely were positive for neuronal markers ß3-tubulin or p-neurofilaments, and few HNP oligodendrocyte progenitors were found. These results suggest that unlike for glioma cells, L1LE does not increase HNP cell motility, but rather decreases motility and influences the differentiation of normal brain progenitor cells. Therefore, the effect of L1LE on increasing motility and proliferation appears to be limited to already transformed cells.


Subject(s)
Cell Differentiation/physiology , Cell Movement/physiology , Cell Proliferation/physiology , Neural Cell Adhesion Molecule L1/metabolism , Neural Stem Cells/metabolism , Cell Line , Child, Preschool , Ectopic Gene Expression , Humans , Male , Neural Cell Adhesion Molecule L1/genetics , Neural Stem Cells/cytology
17.
Nat Commun ; 10(1): 4529, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31586101

ABSTRACT

Dynamic alterations in the unique brain extracellular matrix (ECM) are involved in malignant brain tumors. Yet studies of brain ECM roles in tumor cell behavior have been difficult due to lack of access to the human brain. We present a tunable 3D bioengineered brain tissue platform by integrating microenvironmental cues of native brain-derived ECMs and live imaging to systematically evaluate patient-derived brain tumor responses. Using pediatric ependymoma and adult glioblastoma as examples, the 3D brain ECM-containing microenvironment with a balance of cell-cell and cell-matrix interactions supports distinctive phenotypes associated with tumor type-specific and ECM-dependent patterns in the tumor cells' transcriptomic and release profiles. Label-free metabolic imaging of the composite model structure identifies metabolically distinct sub-populations within a tumor type and captures extracellular lipid-containing droplets with potential implications in drug response. The versatile bioengineered 3D tumor tissue system sets the stage for mechanistic studies deciphering microenvironmental role in brain tumor progression.


Subject(s)
Brain Neoplasms/pathology , Ependymoma/pathology , Extracellular Matrix/pathology , Glioblastoma/pathology , Tissue Engineering/methods , Brain/cytology , Brain/pathology , Brain/surgery , Brain Neoplasms/surgery , Cell Communication , Child, Preschool , Coculture Techniques , Ependymoma/surgery , Female , Humans , Male , Middle Aged , Neural Stem Cells , Neurons , Primary Cell Culture/methods , Spheroids, Cellular , Tumor Cells, Cultured , Tumor Microenvironment
19.
Glia ; 56(16): 1799-808, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18661554

ABSTRACT

Microglia, the resident immune cells of the brain, have recently been hypothesized to play a role both in neuronal diseases and age-related neurogenic decline, and are theorized to be modulators of adult neurogenesis. Current methods for the isolation of microglia from cultured primary brain tissue result in relatively poor yield, requiring a large tissue sample or multiple specimens to obtain a sufficient number of microglia for cell and molecular analysis. We report here a method for the repetitive isolation of microglia from established glial monolayer cultures from which it is possible to expand the initial population of microglia roughly 10,000-fold. The expanded population expresses appropriate microglial morphology and phenotype markers, and demonstrates functionally normal phagocytosis, thus providing a high-yield assay for the investigation and analysis of microglia from a single initial dissection of primary tissue. Furthermore, this massive expansion is limited to microglia derived from the subventricular zone as the fold expansion of isolatable microglia was found to be up to 20 times greater than cultures from other brain regions, indicating unique properties for this persistently neurogenic region.


Subject(s)
Lateral Ventricles/cytology , Microglia/cytology , Prosencephalon/cytology , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cell Culture Techniques/methods , Cell Proliferation , Cell Separation/methods , Cells, Cultured , Dissection/methods , Mice , Mice, Inbred C57BL , Microglia/physiology , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Neurogenesis/physiology , Phagocytosis/physiology , Phenotype , Prosencephalon/physiology , Stem Cells/cytology , Stem Cells/physiology
20.
Methods Mol Biol ; 438: 135-50, 2008.
Article in English | MEDLINE | ID: mdl-18369755

ABSTRACT

The relatively recent discovery of persistent adult neurogenesis has led to the experimental isolation and characterization of central nervous system neural stem cell populations. Protocols for in vitro analysis and expansion of neural stem cells are crucial for understanding their properties and defining characteristics. The methods described here allow for cell and molecular analysis of individual clones of cells--neurospheres--derived from neural stem/progenitor cells. Neurospheres can be cultivated from a variety of normal, genetically altered, or pathological tissue specimens, even with protracted postmortem intervals, for studies of mechanisms underlying neurogenesis, cell fate decisions, and cell differentiation. Neurosphere-forming cells hold great promise for the development of cell and molecular therapeutics for a variety of neurological diseases.


Subject(s)
Cell Separation/methods , Central Nervous System/cytology , Stem Cells/cytology , Animals , Animals, Newborn , Cell Adhesion , Central Nervous System/ultrastructure , Clone Cells , Gene Expression Regulation , Humans , Mice , Stem Cell Transplantation , Stem Cells/metabolism , Stem Cells/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL