Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 72
Filtrer
1.
bioRxiv ; 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38854085

RÉSUMÉ

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.

2.
J Control Release ; 372: 433-445, 2024 Jun 26.
Article de Anglais | MEDLINE | ID: mdl-38908756

RÉSUMÉ

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.

3.
J Comp Neurol ; 528(7): 1203-1215, 2020 05.
Article de Anglais | MEDLINE | ID: mdl-31743443

RÉSUMÉ

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.


Sujet(s)
Marqueurs biologiques , Exosomes/métabolisme , Leucine-rich repeat serine-threonine protein kinase-2/génétique , Maladie de Parkinson , Exosomes/anatomopathologie , Humains , Cellules souches pluripotentes induites , Mutation , Cellules souches neurales , Maladie de Parkinson/génétique , Maladie de Parkinson/métabolisme , Maladie de Parkinson/anatomopathologie , Transcriptome
4.
Nat Commun ; 10(1): 4529, 2019 10 04.
Article de Anglais | MEDLINE | ID: mdl-31586101

RÉSUMÉ

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.


Sujet(s)
Tumeurs du cerveau/anatomopathologie , Épendymome/anatomopathologie , Matrice extracellulaire/anatomopathologie , Glioblastome/anatomopathologie , Ingénierie tissulaire/méthodes , Encéphale/cytologie , Encéphale/anatomopathologie , Encéphale/chirurgie , Tumeurs du cerveau/chirurgie , Communication cellulaire , Enfant d'âge préscolaire , Techniques de coculture , Épendymome/chirurgie , Femelle , Humains , Mâle , Adulte d'âge moyen , Cellules souches neurales , Neurones , Culture de cellules primaires/méthodes , Sphéroïdes de cellules , Cellules cancéreuses en culture , Microenvironnement tumoral
5.
Int J Dev Neurosci ; 78: 49-64, 2019 Nov.
Article de Anglais | MEDLINE | ID: mdl-31421150

RÉSUMÉ

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.


Sujet(s)
Différenciation cellulaire/physiologie , Mouvement cellulaire/physiologie , Prolifération cellulaire/physiologie , Molécule d'adhérence cellulaire neurale L-1/métabolisme , Cellules souches neurales/métabolisme , Lignée cellulaire , Enfant d'âge préscolaire , Expression génique ectopique , Humains , Mâle , Molécule d'adhérence cellulaire neurale L-1/génétique , Cellules souches neurales/cytologie
7.
Proc Natl Acad Sci U S A ; 115(37): E8765-E8774, 2018 09 11.
Article de Anglais | MEDLINE | ID: mdl-30150378

RÉSUMÉ

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.


Sujet(s)
Différenciation cellulaire/effets des médicaments et des substances chimiques , Maladie de Huntington/physiopathologie , Acides hydroxamiques/pharmacologie , Indoles/pharmacologie , Neurones/effets des médicaments et des substances chimiques , Animaux , Animal génétiquement modifié , Différenciation cellulaire/génétique , Différenciation cellulaire/physiologie , Modèles animaux de maladie humaine , Femelle , Inhibiteurs de désacétylase d'histone/pharmacologie , Humains , Protéine huntingtine/génétique , Maladie de Huntington/génétique , Ventricules latéraux/anatomopathologie , Mâle , Souris transgéniques , Mutation , Neurones/métabolisme , Neurones/physiologie , Panobinostat , Rats
8.
Sci Rep ; 7(1): 11496, 2017 09 13.
Article de Anglais | MEDLINE | ID: mdl-28904337

RÉSUMÉ

One third of humans are infected lifelong with the brain-dwelling, protozoan parasite, Toxoplasma gondii. Approximately fifteen million of these have congenital toxoplasmosis. Although neurobehavioral disease is associated with seropositivity, causality is unproven. To better understand what this parasite does to human brains, we performed a comprehensive systems analysis of the infected brain: We identified susceptibility genes for congenital toxoplasmosis in our cohort of infected humans and found these genes are expressed in human brain. Transcriptomic and quantitative proteomic analyses of infected human, primary, neuronal stem and monocytic cells revealed effects on neurodevelopment and plasticity in neural, immune, and endocrine networks. These findings were supported by identification of protein and miRNA biomarkers in sera of ill children reflecting brain damage and T. gondii infection. These data were deconvoluted using three systems biology approaches: "Orbital-deconvolution" elucidated upstream, regulatory pathways interconnecting human susceptibility genes, biomarkers, proteomes, and transcriptomes. "Cluster-deconvolution" revealed visual protein-protein interaction clusters involved in processes affecting brain functions and circuitry, including lipid metabolism, leukocyte migration and olfaction. Finally, "disease-deconvolution" identified associations between the parasite-brain interactions and epilepsy, movement disorders, Alzheimer's disease, and cancer. This "reconstruction-deconvolution" logic provides templates of progenitor cells' potentiating effects, and components affecting human brain parasitism and diseases.

9.
Cancer Res ; 77(19): 5360-5373, 2017 10 01.
Article de Anglais | MEDLINE | ID: mdl-28807938

RÉSUMÉ

Glioma stem cells (GSC) and epithelial-mesenchymal transition (EMT) are strongly associated with therapy resistance and tumor recurrence, but the underlying mechanisms are incompletely understood. Here, we show that S100A4 is a novel biomarker of GSCs. S100A4+ cells in gliomas are enriched with cancer cells that have tumor-initiating and sphere-forming abilities, with the majority located in perivascular niches where GSCs are found. Selective ablation of S100A4-expressing cells was sufficient to block tumor growth in vitro and in vivo We also identified S100A4 as a critical regulator of GSC self-renewal in mouse and patient-derived glioma tumorspheres. In contrast with previous reports of S100A4 as a reporter of EMT, we discovered that S100A4 is an upstream regulator of the master EMT regulators SNAIL2 and ZEB along with other mesenchymal transition regulators in glioblastoma. Overall, our results establish S100A4 as a central node in a molecular network that controls stemness and EMT in glioblastoma, suggesting S100A4 as a candidate therapeutic target. Cancer Res; 77(19); 5360-73. ©2017 AACR.


Sujet(s)
Marqueurs biologiques/métabolisme , Tumeurs du cerveau/anatomopathologie , Transition épithélio-mésenchymateuse , Glioblastome/anatomopathologie , Cellules souches tumorales/anatomopathologie , Protéine S100A4 liant le calcium/métabolisme , Animaux , Apoptose , Tumeurs du cerveau/génétique , Tumeurs du cerveau/métabolisme , Prolifération cellulaire , Femelle , Régulation de l'expression des gènes tumoraux , Glioblastome/génétique , Glioblastome/métabolisme , Humains , Souris , Cellules souches tumorales/métabolisme , Protéine S100A4 liant le calcium/génétique , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
10.
Adv Nutr ; 8(4): 546-557, 2017 Jul.
Article de Anglais | MEDLINE | ID: mdl-28710142

RÉSUMÉ

Good health while aging depends upon optimal cellular and organ functioning that contribute to the regenerative ability of the body during the lifespan, especially when injuries and diseases occur. Although diet may help in the maintenance of cellular fitness during periods of stability or modest decline in the regenerative function of an organ, this approach is inadequate in an aged system, in which the ability to maintain homeostasis is further challenged by aging and the ensuing suboptimal functioning of the regenerative unit, tissue-specific stem cells. Focused nutritional approaches can be used as an intervention to reduce decline in the body's regenerative capacity. This article brings together nutrition-associated therapeutic approaches with the fields of aging, immunology, neurodegenerative disease, and cancer to propose ways in which diet and nutrition can work with standard-of-care and integrated medicine to help improve the brain's function as it ages. The field of regenerative medicine has exploded during the past 2 decades as a result of the discovery of stem cells in nearly every organ system of the body, including the brain, where neural stem cells persist in discrete areas throughout life. This fact, and the uncovering of the genetic basis of plasticity in somatic cells and cancer stem cells, open a door to a world where maintenance and regeneration of organ systems maintain health and extend life expectancy beyond its present limits. An area that has received little attention in regenerative medicine is the influence on regulatory mechanisms and therapeutic potential of nutrition. We propose that a strong relation exists between brain regenerative medicine and nutrition and that nutritional intervention at key times of life could be used to not only maintain optimal functioning of regenerative units as humans age but also play a primary role in therapeutic treatments to combat injury and diseases (in particular, those that occur in the latter one-third of the lifespan).


Sujet(s)
Vieillissement , Encéphale/physiologie , Régénération nerveuse , État nutritionnel , Animaux , Horloges biologiques/physiologie , Cognition , Dysfonctionnement cognitif/diétothérapie , Régime alimentaire , Microbiome gastro-intestinal , Homéostasie , Humains , Micronutriments/administration et posologie , Modèles animaux , Tumeurs/diétothérapie , Maladies neurodégénératives/diétothérapie , Médecine régénérative , Cellules souches/métabolisme
11.
Clin Cancer Res ; 23(2): 562-574, 2017 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-27521447

RÉSUMÉ

PURPOSE: Investigation of clonal heterogeneity may be key to understanding mechanisms of therapeutic failure in human cancer. However, little is known on the consequences of therapeutic intervention on the clonal composition of solid tumors. EXPERIMENTAL DESIGN: Here, we used 33 single cell-derived subclones generated from five clinical glioblastoma specimens for exploring intra- and interindividual spectra of drug resistance profiles in vitro In a personalized setting, we explored whether differences in pharmacologic sensitivity among subclones could be employed to predict drug-dependent changes to the clonal composition of tumors. RESULTS: Subclones from individual tumors exhibited a remarkable heterogeneity of drug resistance to a library of potential antiglioblastoma compounds. A more comprehensive intratumoral analysis revealed that stable genetic and phenotypic characteristics of coexisting subclones could be correlated with distinct drug sensitivity profiles. The data obtained from differential drug response analysis could be employed to predict clonal population shifts within the naïve parental tumor in vitro and in orthotopic xenografts. Furthermore, the value of pharmacologic profiles could be shown for establishing rational strategies for individualized secondary lines of treatment. CONCLUSIONS: Our data provide a previously unrecognized strategy for revealing functional consequences of intratumor heterogeneity by enabling predictive modeling of treatment-related subclone dynamics in human glioblastoma. Clin Cancer Res; 23(2); 562-74. ©2016 AACR.


Sujet(s)
Association médicamenteuse , Résistance aux médicaments antinéoplasiques/génétique , Hétérogénéité génétique , Glioblastome/traitement médicamenteux , Animaux , Évolution clonale/génétique , Glioblastome/génétique , Glioblastome/anatomopathologie , Humains , Souris , Tests d'activité antitumorale sur modèle de xénogreffe
12.
Biotechniques ; 59(3): 137-48, 2015 Sep.
Article de Anglais | MEDLINE | ID: mdl-26345506

RÉSUMÉ

Diverse cell types have unique transcriptional signatures that are best interrogated at single-cell resolution. Here we describe a novel RNA amplification approach that allows for high fidelity gene profiling of individual cells. This technique significantly diminishes the problem of 3' bias, enabling detection of all regions of transcripts, including the recognition of mRNA with short or completely absent poly(A) tails, identification of noncoding RNAs, and discovery of the full array of splice isoforms from any given gene product. We assess this technique using statistical and bioinformatics analyses of microarray data to establish the limitations of the method. To demonstrate applicability, we profiled individual cells isolated from the mouse subventricular zone (SVZ)-a well-characterized, discrete yet highly heterogeneous neural structure involved in persistent neurogenesis. Importantly, this method revealed multiple splice variants of key germinal zone gene products within individual cells, as well as an unexpected coexpression of several mRNAs considered markers of distinct and separate SVZ cell types. These findings were independently confirmed using RNA-fluorescence in situ hybridization (RNA-FISH), contributing to the utility of this new technology that offers genomic and transcriptomic analysis of small numbers of dynamic and clinically relevant cells.


Sujet(s)
Analyse de profil d'expression de gènes/méthodes , Techniques d'amplification d'acides nucléiques/méthodes , ARN/génétique , Analyse sur cellule unique/méthodes , Antigène AC133 , Animaux , Antigènes CD/génétique , Lignée cellulaire tumorale , Protéines de liaison à l'ADN , Récepteurs ErbB/génétique , Protéines de l'oeil/génétique , Protéine gliofibrillaire acide/génétique , Glycoprotéines/génétique , Protéines à fluorescence verte/génétique , Protéines à homéodomaine/génétique , Humains , Protéine d'inhibition de la différenciation de type 1/génétique , Ventricules latéraux/cytologie , Protéines membranaires/génétique , Souris transgéniques , Protéines de tissu nerveux/génétique , Protéines nucléaires/génétique , Facteur de transcription PAX6 , Facteurs de transcription PAX/génétique , Peptides/génétique , Réaction de polymérisation en chaine en temps réel/méthodes , Protéines de répression/génétique
13.
Front Cell Neurosci ; 8: 180, 2014.
Article de Anglais | MEDLINE | ID: mdl-25076873

RÉSUMÉ

Microglia isolated from the neurogenic subependymal zone (SEZ) and hippocampus (HC) are capable of massive in vitro population expansion that is not possible with microglia isolated from non-neurogenic regions. We asked if this regional heterogeneity in microglial proliferative capacity is cell intrinsic, or is conferred by interaction with respective neurogenic or non-neurogenic niches. By combining SEZ and cerebral cortex (CTX) primary tissue dissociates to generate heterospatial cultures, we find that exposure to the SEZ environment does not enhance CTX microglia expansion; however, the CTX environment exerts a suppressive effect on SEZ microglia expansion. Furthermore, addition of purified donor SEZ microglia to either CTX- or SEZ-derived cultures suppresses the expansion of host microglia, while the addition of donor CTX microglia enhances the over-all microglia yield. These data suggest that SEZ and CTX microglia possess intrinsic, spatially restricted characteristics that are independent of their in vitro environment, and that they represent unique and functionally distinct populations. Finally, we determined that the repeated supplementation of neurogenic SEZ cultures with expanded SEZ microglia allows for sustained levels of inducible neurogenesis, provided that the ratio of microglia to total cells remains within a fairly narrow range.

14.
Trends Mol Med ; 20(7): 368-74, 2014 Jul.
Article de Anglais | MEDLINE | ID: mdl-24835084

RÉSUMÉ

Extracellular vesicles (EVs) are released from many cell types, including normal and pathological cells, and range from 30 to 1000 nm in size. Once thought to be a mechanism for discarding unwanted cellular material, EVs are now thought to play a role in intercellular communication. Evidence is accruing that EVs are capable of carrying mRNAs, miRNAs, noncoding RNAs, and proteins, including those associated with neurodegenerative diseases and cancer, which may be exchanged between cells. For this reason, neurodegenerative diseases and cancers may share a common mechanism of disease spread via EVs. Understanding the role EVs play in disease initiation and progression will aid in the discovery of new clinically relevant biomarkers and the development of better targeted molecular and biological therapies.


Sujet(s)
Communication cellulaire/physiologie , Tumeurs/diagnostic , Maladies neurodégénératives/diagnostic , Vésicules de transport/physiologie , Animaux , Évolution de la maladie , Humains , Tumeurs/métabolisme , Maladies neurodégénératives/métabolisme
15.
PLoS One ; 9(3): e88770, 2014.
Article de Anglais | MEDLINE | ID: mdl-24594681

RÉSUMÉ

OBJECTIVE: Deep brain stimulation (DBS) has been used for more than a decade to treat Parkinson's disease (PD); however, its mechanism of action remains unknown. Given the close proximity of the electrode trajectory to areas of the brain known as the "germinal niches," we sought to explore the possibility that DBS influences neural stem cell proliferation locally, as well as more distantly. METHODS: We studied the brains of a total of 12 idiopathic Parkinson's disease patients that were treated with DBS (the electrode placement occurred 0.5-6 years before death), and who subsequently died of unrelated illnesses. These were compared to the brains of 10 control individuals without CNS disease, and those of 5 PD patients with no DBS. RESULTS: Immunohistochemical analyses of the subventricular zone (SVZ) of the lateral ventricles, the third ventricle lining, and the tissue surrounding the DBS lead revealed significantly greater numbers of proliferating cells expressing markers of the cell cycle, plasticity, and neural precursor cells in PD-DBS tissue compared with both normal brain tissue and tissue from PD patients not treated with DBS. The level of cell proliferation in the SVZ in PD-DBS brains was 2-6 fold greater than that in normal and untreated PD brains. CONCLUSIONS: Our data suggest that DBS is capable of increasing cellular plasticity in the brain, and we hypothesize that it may have more widespread effects beyond the electrode location. It is unclear whether these effects of DBS have any symptomatic or other beneficial influences on PD.


Sujet(s)
Prolifération cellulaire , Stimulation cérébrale profonde , Maladie de Parkinson/thérapie , Humains , Immunohistochimie , Maladie de Parkinson/anatomopathologie
16.
Stem Cells Transl Med ; 3(4): 470-80, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24604282

RÉSUMÉ

Neural progenitor cells (NPCs) in the subventricular zone (SVZ) hold promise for future therapy for neurodegenerative disorders, because the stimulation of adult neurogenesis could potentially restore the function of degenerating neurons and glia. To obtain more knowledge on these NPCs, we developed a method to specifically isolate NPCs from postmortem adult human brains based on the expression of the specific human adult neural stem/progenitor cell marker glial fibrillary acidic protein δ (GFAPδ). An extensive immunophenotyping analysis for cell surface markers resulted in the observation that CD271 was limited to the SVZ-derived GFAPδ-positive cells. CD271(+) cells developed into neurospheres and could be differentiated into astrocytes, neurons, and oligodendrocytes. We are the first to show that a pure population of NPCs can be isolated from the adult human SVZ, which is highly instrumental for developing future therapies based on stimulating endogenous SVZ neurogenesis.


Sujet(s)
Antigènes de différenciation/biosynthèse , Encéphale , Séparation cellulaire , Expression des gènes , Protéines de tissu nerveux/biosynthèse , Cellules souches neurales , Récepteurs facteur croissance nerf/biosynthèse , Adulte , Astrocytes/cytologie , Astrocytes/métabolisme , Encéphale/cytologie , Encéphale/métabolisme , Différenciation cellulaire , Femelle , Protéine gliofibrillaire acide/biosynthèse , Humains , Mâle , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Neurogenèse/physiologie , Neurones/cytologie , Neurones/métabolisme , Oligodendroglie/cytologie , Oligodendroglie/métabolisme
17.
J Neurooncol ; 117(1): 15-24, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24510433

RÉSUMÉ

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.


Sujet(s)
Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/ultrastructure , Cils vibratiles/ultrastructure , Glioblastome/métabolisme , Glioblastome/ultrastructure , Facteurs d'ADP-ribosylation/métabolisme , Sujet âgé de 80 ans ou plus , Axonème/métabolisme , Axonème/ultrastructure , Corpuscules basaux/métabolisme , Corpuscules basaux/ultrastructure , Lignée cellulaire tumorale , Cils vibratiles/métabolisme , Protéines à homéodomaine/métabolisme , Humains , Immunohistochimie , Antigène KI-67/métabolisme , Mâle , Microscopie électronique , Adulte d'âge moyen , Facteurs de transcription/métabolisme , Tubuline/métabolisme , Protéines suppresseurs de tumeurs/métabolisme , Facteur de transcription Zeb1
18.
Transl Res ; 163(4): 432-8, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24286919

RÉSUMÉ

Identifying novel, effective therapeutics for Alzheimer's disease (AD) is one of the major unmet medical needs for the coming decade. Because the current paradigm for developing and testing disease-modifying AD therapies is protracted and likely to be even longer, with the shift toward earlier intervention in preclinical AD, it is an open issue whether we can develop, test, and widely deploy a novel therapy in time to help the current at-risk generation if we continue to follow the standard paradigms of discovery and drug development. There is an imperative need to find safe and effective preventive measures that can be distributed rapidly to stem the coming wave of AD that will potentially engulf the next generation. We can define regenerative medicine broadly as approaches that use stem cell-based therapies or approaches that seek to modulate inherent neurogenesis. Neurogenesis, although most active during prenatal development, has been shown to continue in several small parts of the brain, including the hippocampus and the subventricular zone, suggesting its potential to reverse cognitive deficits. If AD pathology affects neurogenesis, then it follows that conditions that stimulate endogenous neurogenesis (eg, environmental stimuli, physical activity, trophic factors, cytokines, and drugs) may help to promote the regenerative and recovery process. Herein, we review the complex logistics of potentially implementing neurogenesis-based therapeutic strategies for the treatment of AD.


Sujet(s)
Maladie d'Alzheimer/thérapie , Thérapie cellulaire et tissulaire/méthodes , Médecine régénérative/méthodes , Médecine régénérative/tendances , Humains , Neurogenèse/physiologie
19.
J Neurosci ; 33(39): 15603-17, 2013 Sep 25.
Article de Anglais | MEDLINE | ID: mdl-24068827

RÉSUMÉ

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.


Sujet(s)
Tumeurs du cerveau/métabolisme , Protéoglycanes à chondroïtine sulfate/métabolisme , Gliome/métabolisme , Microenvironnement tumoral , Adulte , Animaux , Astrocytes/métabolisme , Astrocytes/anatomopathologie , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire , Cellules cultivées , Enfant , Protéoglycanes à chondroïtine sulfate/génétique , Femelle , Gliome/anatomopathologie , Glycosylation , Humains , Mâle , Souris , Microglie/métabolisme , Microglie/anatomopathologie , Adulte d'âge moyen , Invasion tumorale , Receptor-Like Protein Tyrosine Phosphatases, Class 2/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
20.
Clin Cancer Res ; 19(15): 4124-36, 2013 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-23908450

RÉSUMÉ

PURPOSE: Glioblastoma is a highly malignant, invariably fatal brain tumor for which effective pharmacotherapy remains an unmet medical need. EXPERIMENTAL DESIGN: Screening of a compound library of 160 synthetic and natural toxic substances identified the antihelmintic niclosamide as a previously unrecognized candidate for clinical development. Considering the cellular and interindividual heterogeneity of glioblastoma, a portfolio of short-term expanded primary human glioblastoma cells (pGBM; n = 21), common glioma lines (n = 5), and noncancer human control cells (n = 3) was applied as a discovery platform and for preclinical validation. Pharmacodynamic analysis, study of cell-cycle progression, apoptosis, cell migration, proliferation, and on the frequency of multipotent/self-renewing pGBM cells were conducted in vitro, and orthotopic xenotransplantation was used to confirm anticancer effects in vivo. RESULTS: Niclosamide led to cytostatic, cytotoxic, and antimigratory effects, strongly reduced the frequencies of multipotent/self-renewing cells in vitro, and after exposure significantly diminished the pGBMs' malignant potential in vivo. Mechanism of action analysis revealed that niclosamide simultaneously inhibited intracellular WNT/CTNNB1-, NOTCH-, mTOR-, and NF-κB signaling cascades. Furthermore, combinatorial drug testing established that a heterozygous deletion of the NFKBIA locus in glioblastoma samples could serve as a genomic biomarker for predicting a synergistic activity of niclosamide with temozolomide, the current standard in glioblastoma therapy. CONCLUSIONS: Together, our data advocate the use of pGBMs for exploration of compound libraries to reveal unexpected leads, for example, niclosamide that might be suited for further development toward personalized clinical application.


Sujet(s)
Tumeurs du cerveau/traitement médicamenteux , Glioblastome/traitement médicamenteux , Glioblastome/génétique , Niclosamide/administration et posologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Glioblastome/anatomopathologie , Humains , Souris , Facteur de transcription NF-kappa B/métabolisme , Récepteurs Notch/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...