14 results on '"Steindler DA"'
Search Results
2. Cranioencephalic functional lymphoid units in glioblastoma.
- Author
-
Dobersalske C, Rauschenbach L, Hua Y, Berliner C, Steinbach A, Grüneboom A, Kokkaliaris KD, Heiland DH, Berger P, Langer S, Tan CL, Stenzel M, Landolsi S, Weber F, Darkwah Oppong M, Werner RA, Gull H, Schröder T, Linsenmann T, Buck AK, Gunzer M, Stuschke M, Keyvani K, Forsting M, Glas M, Kipnis J, Steindler DA, Reinhardt HC, Green EW, Platten M, Tasdogan A, Herrmann K, Rambow F, Cima I, Sure U, and Scheffler B
- Subjects
- Humans, Male, Female, Middle Aged, Skull pathology, Bone Marrow pathology, Aged, Adult, Sphingosine-1-Phosphate Receptors metabolism, Glioblastoma pathology, Glioblastoma immunology, Brain Neoplasms pathology, Brain Neoplasms immunology, Receptors, CXCR4 metabolism, Receptors, CXCR4 genetics, CD8-Positive T-Lymphocytes immunology
- 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., (© 2024. The Author(s).)- Published
- 2024
- Full Text
- View/download PDF
3. Auto-loaded TRAIL-exosomes derived from induced neural stem cells for brain cancer therapy.
- Author
-
Zhang X, Taylor H, Valdivia A, Dasari R, Buckley A, Bonacquisti E, Nguyen J, Kanchi K, Corcoran DL, Herring LE, Steindler DA, Baldwin A, Hingtgen S, and Satterlee AB
- Subjects
- Animals, Humans, Cell Line, Tumor, Female, Mice, Mice, Nude, TNF-Related Apoptosis-Inducing Ligand administration & dosage, TNF-Related Apoptosis-Inducing Ligand genetics, Neural Stem Cells, Brain Neoplasms therapy, Brain Neoplasms pathology, Exosomes metabolism
- 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., Competing Interests: Declaration of competing interest None., (Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
4. Sugar substitutes and taste enhancers need more science, sensitivity- and allergy-guided labeling.
- Author
-
Steindler DA
- Published
- 2023
- Full Text
- View/download PDF
5. Exosome/microvesicle content is altered in leucine-rich repeat kinase 2 mutant induced pluripotent stem cell-derived neural cells.
- Author
-
Candelario KM, Balaj L, Zheng T, Skog J, Scheffler B, Breakefield X, Schüle B, and Steindler DA
- Subjects
- Exosomes pathology, Humans, Induced Pluripotent Stem Cells, Mutation, Neural Stem Cells, Transcriptome, Biomarkers, Exosomes metabolism, Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 genetics, Parkinson Disease genetics, Parkinson Disease metabolism, Parkinson Disease pathology
- 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., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
6. Ectopic expression of L1CAM ectodomain alters differentiation and motility, but not proliferation, of human neural progenitor cells.
- Author
-
Pusey MA, Pace K, Fascelli M, Linser PJ, Steindler DA, and Galileo DS
- Subjects
- Cell Line, Child, Preschool, Ectopic Gene Expression, Humans, Male, Neural Cell Adhesion Molecule L1 genetics, Neural Stem Cells cytology, Cell Differentiation physiology, Cell Movement physiology, Cell Proliferation physiology, Neural Cell Adhesion Molecule L1 metabolism, Neural Stem Cells metabolism
- 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 β3-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 β3-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., (Copyright © 2019 ISDN. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
7. 3D extracellular matrix microenvironment in bioengineered tissue models of primary pediatric and adult brain tumors.
- Author
-
Sood D, Tang-Schomer M, Pouli D, Mizzoni C, Raia N, Tai A, Arkun K, Wu J, Black LD 3rd, Scheffler B, Georgakoudi I, Steindler DA, and Kaplan DL
- Subjects
- 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, Brain Neoplasms pathology, Ependymoma pathology, Extracellular Matrix pathology, Glioblastoma pathology, Tissue Engineering methods
- 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.
- Published
- 2019
- Full Text
- View/download PDF
8. Publisher Correction: Toxoplasma Modulates Signature Pathways of Human Epilepsy, Neurodegeneration & Cancer.
- Author
-
Ngô HM, Zhou Y, Lorenzi H, Wang K, Kim TK, Zhou Y, El Bissati K, Mui E, Fraczek L, Rajagopala SV, Roberts CW, Henriquez FL, Montpetit A, Blackwell JM, Jamieson SE, Wheeler K, Begeman IJ, Naranjo-Galvis C, Alliey-Rodriguez N, Davis RG, Soroceanu L, Cobbs C, Steindler DA, Boyer K, Noble AG, Swisher CN, Heydemann PT, Rabiah P, Withers S, Soteropoulos P, Hood L, and McLeod R
- Abstract
A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.
- Published
- 2019
- Full Text
- View/download PDF
9. Early postnatal behavioral, cellular, and molecular changes in models of Huntington disease are reversible by HDAC inhibition.
- Author
-
Siebzehnrübl FA, Raber KA, Urbach YK, Schulze-Krebs A, Canneva F, Moceri S, Habermeyer J, Achoui D, Gupta B, Steindler DA, Stephan M, Nguyen HP, Bonin M, Riess O, Bauer A, Aigner L, Couillard-Despres S, Paucar MA, Svenningsson P, Osmand A, Andreew A, Zabel C, Weiss A, Kuhn R, Moussaoui S, Blockx I, Van der Linden A, Cheong RY, Roybon L, Petersén Å, and von Hörsten S
- Subjects
- 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, Cell Differentiation drug effects, Huntington Disease physiopathology, Hydroxamic Acids pharmacology, Indoles pharmacology, Neurons drug effects
- 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., Competing Interests: The authors declare no conflict of interest., (Copyright © 2018 the Author(s). Published by PNAS.)
- Published
- 2018
- Full Text
- View/download PDF
10. S100A4 Is a Biomarker and Regulator of Glioma Stem Cells That Is Critical for Mesenchymal Transition in Glioblastoma.
- Author
-
Chow KH, Park HJ, George J, Yamamoto K, Gallup AD, Graber JH, Chen Y, Jiang W, Steindler DA, Neilson EG, Kim BYS, and Yun K
- Subjects
- Animals, Apoptosis, Brain Neoplasms genetics, Brain Neoplasms metabolism, Cell Proliferation, Female, Gene Expression Regulation, Neoplastic, Glioblastoma genetics, Glioblastoma metabolism, Humans, Mice, Neoplastic Stem Cells metabolism, S100 Calcium-Binding Protein A4 genetics, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Biomarkers metabolism, Brain Neoplasms pathology, Epithelial-Mesenchymal Transition, Glioblastoma pathology, Neoplastic Stem Cells pathology, S100 Calcium-Binding Protein A4 metabolism
- Abstract
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 ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
11. Toxoplasma Modulates Signature Pathways of Human Epilepsy, Neurodegeneration & Cancer.
- Author
-
Ngô HM, Zhou Y, Lorenzi H, Wang K, Kim TK, Zhou Y, El Bissati K, Mui E, Fraczek L, Rajagopala SV, Roberts CW, Henriquez FL, Montpetit A, Blackwell JM, Jamieson SE, Wheeler K, Begeman IJ, Naranjo-Galvis C, Alliey-Rodriguez N, Davis RG, Soroceanu L, Cobbs C, Steindler DA, Boyer K, Noble AG, Swisher CN, Heydemann PT, Rabiah P, Withers S, Soteropoulos P, Hood L, and McLeod R
- Abstract
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.
- Published
- 2017
- Full Text
- View/download PDF
12. Perspective: Neuroregenerative Nutrition.
- Author
-
Steindler DA and Reynolds BA
- Subjects
- Animals, Biological Clocks physiology, Cognition, Cognitive Dysfunction diet therapy, Diet, Gastrointestinal Microbiome, Homeostasis, Humans, Micronutrients administration & dosage, Models, Animal, Neoplasms diet therapy, Neurodegenerative Diseases diet therapy, Regenerative Medicine, Stem Cells metabolism, Aging, Brain physiology, Nerve Regeneration, Nutritional Status
- Abstract
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)., Competing Interests: Author disclosures: The authors are affiliated with a new start-up company, Prana Therapeutics, Inc. They have received no financial compensation in relation to this article., (© 2017 American Society for Nutrition.)
- Published
- 2017
- Full Text
- View/download PDF
13. Functional Subclone Profiling for Prediction of Treatment-Induced Intratumor Population Shifts and Discovery of Rational Drug Combinations in Human Glioblastoma.
- Author
-
Reinartz R, Wang S, Kebir S, Silver DJ, Wieland A, Zheng T, Küpper M, Rauschenbach L, Fimmers R, Shepherd TM, Trageser D, Till A, Schäfer N, Glas M, Hillmer AM, Cichon S, Smith AA, Pietsch T, Liu Y, Reynolds BA, Yachnis A, Pincus DW, Simon M, Brüstle O, Steindler DA, and Scheffler B
- Subjects
- Animals, Clonal Evolution genetics, Glioblastoma genetics, Glioblastoma pathology, Humans, Mice, Xenograft Model Antitumor Assays, Drug Combinations, Drug Resistance, Neoplasm genetics, Genetic Heterogeneity, Glioblastoma drug therapy
- Abstract
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., Competing Interests: The authors declare no conflict of interests, (©2016 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
14. Application of an RNA amplification method for reliable single-cell transcriptome analysis.
- Author
-
Suslov O, Silver DJ, Siebzehnrubl FA, Orjalo A, Ptitsyn A, and Steindler DA
- Subjects
- AC133 Antigen, Animals, Antigens, CD genetics, Cell Line, Tumor, DNA-Binding Proteins, ErbB Receptors genetics, Eye Proteins genetics, Glial Fibrillary Acidic Protein genetics, Glycoproteins genetics, Green Fluorescent Proteins genetics, Homeodomain Proteins genetics, Humans, Inhibitor of Differentiation Protein 1 genetics, Lateral Ventricles cytology, Membrane Proteins genetics, Mice, Transgenic, Nerve Tissue Proteins genetics, Nuclear Proteins genetics, PAX6 Transcription Factor, Paired Box Transcription Factors genetics, Peptides genetics, Real-Time Polymerase Chain Reaction methods, Repressor Proteins genetics, Gene Expression Profiling methods, Nucleic Acid Amplification Techniques methods, RNA genetics, Single-Cell Analysis methods
- Abstract
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.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.