Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
medRxiv ; 2023 Oct 31.
Article in English | MEDLINE | ID: mdl-37961425

ABSTRACT

INTRODUCTION: The APOE gene is the strongest genetic risk factor for late-onset Alzheimer's Disease (LOAD). However, the gene regulatory mechanisms at this locus have not been fully characterized. METHODS: To identify novel AD-linked functional elements within the APOE locus, we integrated SNP variants with RNA-seq, DNA methylation, and ChIP-seq data from human postmortem brains. RESULTS: We identified an AD-linked APOE transcript (jxn1.2.2) observed in the dorsolateral prefrontal cortex (DLPFC). The APOE jxn1.2.2 transcript is associated with brain neuropathological features in DLPFC. We prioritized an independent functional SNP, rs157580, significantly associated with jxn1.2.2 transcript abundance and DNA methylation levels. rs157580 is located within active chromatin regions and predicted to affect brain-related transcriptional factors binding affinity. rs157580 shared the effects on the jxn1.2.2 transcript between European and African ethnic groups. DISCUSSION: The novel APOE functional elements provide potential therapeutic targets with mechanistic insight into the disease's etiology.

2.
Stem Cells ; 41(6): 570-577, 2023 06 15.
Article in English | MEDLINE | ID: mdl-37186298

ABSTRACT

After ischemia, cells in the brain parenchyma upregulate stromal derived factor 1 (SDF1), driving chemokine receptor CXCR4-mediated migration of adult neural stem cells to the ischemic injury. We discovered a novel regulator of CXCR4 in neural stem cells, low-density lipoprotein receptor related protein 1 (LRP1). We used Nestin-driven knockout of LRP1 and induction of td-tomato in neural stem cells of adult mice. We observed reduced localization of td-tomato positive cells to the lesion, and find disrupted CXCR4-mediated neural stem cell migration in vitro, which is likely driven by LRP1-mediated loss of CXCR4 expression in vivo. Our results suggest that LRP1 is a novel regulator of CXCR4 in neural stem cells. This heretofore unknown interaction between LRP1 and CXCR4 could have significant consequences for multiple aspects of neural stem cell physiology.


Subject(s)
Chemokine CXCL12 , Neural Stem Cells , Mice , Animals , Chemokine CXCL12/metabolism , Neural Stem Cells/metabolism , Cell Movement/physiology , Brain/metabolism , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Ischemia/metabolism
3.
J Neurochem ; 162(5): 430-443, 2022 09.
Article in English | MEDLINE | ID: mdl-35560167

ABSTRACT

Microglia have been implicated in multiple sclerosis (MS) pathogenesis. The fractalkine receptor CX3CR1 limits the activation of pathogenic microglia and the human polymorphic CX3CR1I249/M280 (hCX3CR1I249/M280 ) variant increases disease progression in models of MS. However, the role of hCX3CR1I249/M280 variant on microglial activation and central nervous system repair mechanisms remains unknown. Therefore, using transgenic mice expressing the hCX3CR1I249/M280 variant, we aimed to determine the contribution of defective CX3CR1 signaling to neuroinflammation and remyelination in the cuprizone model of focal demyelination. Here, we report that mice expressing hCX3CR1I249/M280 exhibit marked demyelination and microgliosis following acute cuprizone treatment. Nanostring gene expression analysis in demyelinated lesions showed that hCX3CR1I249/M280 but not CX3CR1-deficient mice up-regulated the cuprizone-induced gene profile linked to inflammatory, oxidative stress, and phagocytic pathways. Although CX3CR1-deficient (CX3CR1-KO) and fractalkine-deficient (FKN-KO) mice displayed a comparable demyelination and microglial activation phenotype to hCX3CR1I249/M280 mice, only CX3CR1-deficient and CX3CR1-WT mice showed significant myelin recovery 1 week from cuprizone withdrawal. Confocal microscopy showed that hCX3CR1I249/M280 variant inhibits the generation of cells involved in myelin repair. Our results show that defective fractalkine signaling contributes to regional differences in demyelination, and suggest that the CX3CR1 pathway activity may be a key mechanism for limiting toxic gene responses in neuroinflammation. Cover Image for this issue: https://doi.org/10.1111/jnc.15416.


Subject(s)
Demyelinating Diseases , Remyelination , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Chemokine CX3CL1/genetics , Chemokine CX3CL1/metabolism , Cuprizone/metabolism , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Disease Models, Animal , Humans , Mice , Mice, Inbred C57BL , Microglia/metabolism , Myelin Sheath , Neuroinflammatory Diseases
4.
Cancers (Basel) ; 13(2)2021 Jan 14.
Article in English | MEDLINE | ID: mdl-33466745

ABSTRACT

Tumor suppressor microRNAs (miRNAs) have been explored as agents to target cancer stem cells. Most strategies use a single miRNA mimic and present many disadvantages, such as the amount of reagent required and the diluted effect on target genes. miRNAs work in a cooperative fashion to regulate distinct biological processes and pathways. Therefore, we propose that miRNA combinations could provide more efficient ways to target cancer stem cells. We have previously shown that miR-124, miR-128, and miR-137 function synergistically to regulate neurogenesis. We used a combination of these three miRNAs to treat glioma stem cells and showed that this treatment was much more effective than single miRNAs in disrupting cell proliferation and survival and promoting differentiation and response to radiation. Transcriptomic analyses indicated that transcription regulation, angiogenesis, metabolism, and neuronal differentiation are among the main biological processes affected by transfection of this miRNA combination. In conclusion, we demonstrated the value of using combinations of neurogenic miRNAs to disrupt cancer phenotypes and glioma stem cell growth. The synergistic effect of these three miRNA amplified the repression of oncogenic factors and the effect on cancer relevant pathways. Future therapeutic approaches would benefit from utilizing miRNA combinations, especially when targeting cancer-initiating cell populations.

5.
Genome Biol ; 21(1): 195, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32762776

ABSTRACT

BACKGROUND: RNA-binding proteins (RBPs) function as master regulators of gene expression. Alterations in RBP expression and function are often observed in cancer and influence critical pathways implicated in tumor initiation and growth. Identification and characterization of oncogenic RBPs and their regulatory networks provide new opportunities for targeted therapy. RESULTS: We identify the RNA-binding protein SERBP1 as a novel regulator of glioblastoma (GBM) development. High SERBP1 expression is prevalent in GBMs and correlates with poor patient survival and poor response to chemo- and radiotherapy. SERBP1 knockdown causes delay in tumor growth and impacts cancer-relevant phenotypes in GBM and glioma stem cell lines. RNAcompete identifies a GC-rich region as SERBP1-binding motif; subsequent genomic and functional analyses establish SERBP1 regulation role in metabolic routes preferentially used by cancer cells. An important consequence of these functions is SERBP1 impact on methionine production. SERBP1 knockdown decreases methionine levels causing a subsequent reduction in histone methylation as shown for H3K27me3 and upregulation of genes associated with neurogenesis, neuronal differentiation, and function. Further analysis demonstrates that several of these genes are downregulated in GBM, potentially through epigenetic silencing as indicated by the presence of H3K27me3 sites. CONCLUSIONS: SERBP1 is the first example of an RNA-binding protein functioning as a central regulator of cancer metabolism and indirect modulator of epigenetic regulation in GBM. By bridging these two processes, SERBP1 enhances glioma stem cell phenotypes and contributes to GBM poorly differentiated state.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , RNA-Binding Proteins/metabolism , Animals , Brain Neoplasms/etiology , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Epigenesis, Genetic , Female , Glioblastoma/etiology , Glioblastoma/mortality , Glioblastoma/therapy , Humans , Male , Mice , Neurogenesis , Phenotype , Prognosis , United States/epidemiology
6.
Aging Cell ; 19(1): e13038, 2020 01.
Article in English | MEDLINE | ID: mdl-31637864

ABSTRACT

Aging is a negative regulator of general homeostasis, tissue function, and regeneration. Changes in organismal energy levels and physiology, through systemic manipulations such as calorie restriction and young blood infusion, can regenerate tissue activity and increase lifespan in aged mice. However, whether these two systemic manipulations could be linked has never been investigated. Here, we report that systemic GDF11 triggers a calorie restriction-like phenotype without affecting appetite or GDF15 levels in the blood, restores the insulin/IGF-1 signaling pathway, and stimulates adiponectin secretion from white adipose tissue by direct action on adipocytes, while repairing neurogenesis in the aged brain. These findings suggest that GDF11 has a pleiotropic effect on an organismal level and that it could be a linking mechanism of rejuvenation between heterochronic parabiosis and calorie restriction. As such, GDF11 could be considered as an important therapeutic candidate for age-related neurodegenerative and metabolic disorders.


Subject(s)
Adiponectin/metabolism , Bone Morphogenetic Proteins/therapeutic use , Caloric Restriction/methods , Growth Differentiation Factors/therapeutic use , Aging , Animals , Bone Morphogenetic Proteins/pharmacology , Growth Differentiation Factors/pharmacology , Mice , Phenotype
7.
Mol Cancer Res ; 18(1): 68-78, 2020 01.
Article in English | MEDLINE | ID: mdl-31624087

ABSTRACT

13-Cis-retinoic acid (RA) is typically used in postremission maintenance therapy in patients with neuroblastoma. However, side effects and recurrence are often observed. We investigated the use of miRNAs as a strategy to replace RA as promoters of differentiation. miR-124 was identified as the top candidate in a functional screen. Genomic target analysis indicated that repression of a network of transcription factors (TF) could be mediating most of miR-124's effect in driving differentiation. To advance miR-124 mimic use in therapy and better define its mechanism of action, a high-throughput siRNA morphologic screen focusing on its TF targets was conducted and ELF4 was identified as a leading candidate for miR-124 repression. By altering its expression levels, we showed that ELF4 maintains neuroblastoma in an undifferentiated state and promotes proliferation. Moreover, ELF4 transgenic expression was able to counteract the neurogenic effect of miR-124 in neuroblastoma cells. With RNA sequencing, we established the main role of ELF4 to be regulation of cell-cycle progression, specifically through the DREAM complex. Interestingly, several cell-cycle genes activated by ELF4 are repressed by miR-124, suggesting that they might form a TF-miRNA regulatory loop. Finally, we showed that high ELF4 expression is often observed in neuroblastomas and is associated with poor survival. IMPLICATIONS: miR-124 induces neuroblastoma differentiation partially through the downregulation of TF ELF4, which drives neuroblastoma proliferation and its undifferentiated phenotype.


Subject(s)
DNA-Binding Proteins/metabolism , MicroRNAs/metabolism , Neuroblastoma/metabolism , Transcription Factors/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Cell Proliferation/physiology , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , MicroRNAs/genetics , Neuroblastoma/genetics , Neuroblastoma/pathology , Survival Rate , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transfection
8.
Curr Opin Pharmacol ; 50: 1-8, 2020 02.
Article in English | MEDLINE | ID: mdl-31756641

ABSTRACT

Neural stem cells exist in specialized regions of the brain and have the capacity to give rise to neurons and glia over the lifespan. The process of giving rise to new neurons, also known as neurogenesis, is thought to be important in cognition and certain types of brain repair. However, during aging, neural stem cell number and function is reduced resulting in fewer new neurons and declines in learning, memory and repair. Recently, research has approached this problem through the lens of rejuvenation that now has produced several strategies, from dietary to pharmacological interventions, to restore functional neurogenesis that resembles the youthful brain. Here, we outline aging in the subventricular zone neurogenic niche, review the multiple modalities of rejuvenation strategies, and propose next steps for future studies to approach translational outcomes.


Subject(s)
Aging/physiology , Brain/physiology , Neurogenesis , Animals , Humans
9.
RNA ; 25(7): 768-782, 2019 07.
Article in English | MEDLINE | ID: mdl-31004009

ABSTRACT

RNA-binding proteins (RBPs) and miRNAs are critical gene expression regulators that interact with one another in cooperative and antagonistic fashions. We identified Musashi1 (Msi1) and miR-137 as regulators of a molecular switch between self-renewal and differentiation. Msi1 and miR-137 have opposite expression patterns and functions, and Msi1 is repressed by miR-137. Msi1 is a stem-cell protein implicated in self-renewal while miR-137 functions as a proneuronal differentiation miRNA. In gliomas, miR-137 functions as a tumor suppressor while Msi1 is a prooncogenic factor. We suggest that the balance between Msi1 and miR-137 is a key determinant in cell fate decisions and disruption of this balance could contribute to neurodegenerative diseases and glioma development. Genomic analyses revealed that Msi1 and miR-137 share 141 target genes associated with differentiation, development, and morphogenesis. Initial results pointed out that these two regulators have an opposite impact on the expression of their target genes. Therefore, we propose an antagonistic model in which this network of shared targets could be either repressed by miR-137 or activated by Msi1, leading to different outcomes (self-renewal, proliferation, tumorigenesis).


Subject(s)
Cell Differentiation , Cell Transformation, Neoplastic/pathology , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , MicroRNAs/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis , RNA-Binding Proteins/metabolism , Animals , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Nerve Tissue Proteins/genetics , RNA-Binding Proteins/genetics , Signal Transduction , Tumor Cells, Cultured
10.
Aging (Albany NY) ; 11(1): 115-126, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30622221

ABSTRACT

The brain can generate new neurons from neural stem cells throughout life. However, the capacity for neurogenesis declines with age, reducing the potential for learning and repair. We explored the effects of calorie restriction, an established anti-aging intervention, on neural stem cells in the subventricular zone of young and aged mice. Calorie restriction transiently enhanced proliferation of neural progenitor cells in young, but not aged mice. However, calorie restriction prevented the age-related loss of neurogenesis in the aged brain. Calorie-restricted mice showed enhanced olfactory memory compared with ad libitum-fed controls, suggesting that calorie restriction can produce functional improvements in the aged brain. Calorie restriction also mitigated the age-related activation of microglia and subsequent increase in pro-inflammatory cytokines. Likewise, calorie restriction prevented increases in senescent cells normally observed in the subventricular zone in aged mice, further protecting this neurogenic niche from pro-inflammatory signals. Together, these data suggest that calorie restriction protects the subventricular zone microenvironment from age-related inflammation, thereby preserving neurogenesis into old age.


Subject(s)
Aging , Caloric Restriction , Cellular Senescence/physiology , Lateral Ventricles/cytology , Neural Stem Cells/physiology , Animals , Female , Male , Mice
11.
Stem Cell Reports ; 12(1): 6-13, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30595545

ABSTRACT

The vascular compartment of the adult brain ventricular-subventricular zone (V-SVZ) is a critical regulator of neural stem cell and progenitor function. Blood enters the V-SVZ via arteries and arterioles to capillaries that then connect with venules and veins to return blood to the heart. We found that stromal cell-derived factor 1 (SDF1) is expressed by a subpopulation of V-SVZ vessels, the capillaries, and that actively proliferating neural stem cells (NSCs) and progenitors are preferentially associated with these SDF1-positive vessels. In contrast, slowly dividing or quiescent NSCs are most prevalent near SDF1-negative vessels. By conditional knockout, we found that loss of SDF1 signaling in NSCs stimulates lineage progression and NSC displacement from the vessel niche. With aging, SDF1/CXCR4 signaling is dysregulated, coincident with reduced proliferation and increased displacement of dividing cells from the vasculature. Our findings demonstrate SDF1-based vascular heterogeneity in the niche and suggest that reduced SDF1 signaling contributes to age-related declines in adult neurogenesis.


Subject(s)
Capillaries/metabolism , Chemokine CXCL12/genetics , Lateral Ventricles/cytology , Neural Stem Cells/metabolism , Neurogenesis , Stem Cell Niche , Animals , Capillaries/cytology , Cell Proliferation , Chemokine CXCL12/metabolism , Lateral Ventricles/blood supply , Lateral Ventricles/growth & development , Mice , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Receptors, CXCR4/metabolism , Signal Transduction
12.
Am J Physiol Heart Circ Physiol ; 313(5): H896-H902, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28801522

ABSTRACT

Neural stem cells (NSCs) persist throughout life in the dentate gyrus and the ventricular-subventricular zone, where they continuously provide new neurons and some glia. These cells are found in specialized niches that regulate quiescence, activation, differentiation, and cell fate choice. A key aspect of the regulatory niche is the vascular plexus, which modulates NSC behavior during tissue homeostasis and regeneration. During aging, NSCs become depleted and dysfunctional, resulting in reduced neurogenesis and poor brain repair. In this review, we discuss the emerging evidence that changes in the vascular niche both structurally and functionally contribute to reduced neurogenesis during aging and how this might contribute to reduced plasticity and repair in the aged brain.


Subject(s)
Aging/physiology , Blood Vessels/innervation , Neural Stem Cells/physiology , Stem Cell Niche/physiology , Animals , Blood Vessels/physiology , Brain/growth & development , Brain/physiology , Humans
13.
Biochem Pharmacol ; 141: 77-85, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28625813

ABSTRACT

Adult neurogenesis is the process of producing new neurons from neural stem cells (NSCs) for integration into the brain circuitry. Neurogenesis occurs throughout life in the ventricular-subventricular zone (V-SVZ) of the lateral ventricle and the subgranular zone (SGZ) of the hippocampal dentate gyrus. However, during aging, NSCs and their progenitors exhibit reduced proliferation and neuron production, which is thought to contribute to age-related cognitive impairment and reduced plasticity that is necessary for some types of brain repair. In this review, we describe NSCs and their niches during tissue homeostasis and how they undergo age-associated remodeling and dysfunction. We also discuss some of the functional ramifications in the brain from NSC aging. Finally, we discuss some recent insights from interventions in NSC aging that could eventually translate into therapies for healthy brain aging.


Subject(s)
Aging/metabolism , Brain/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Aging/pathology , Animals , Brain/pathology , Humans , Neural Stem Cells/pathology , Stem Cell Transplantation/methods
14.
Brain Res ; 1655: 270-276, 2017 01 15.
Article in English | MEDLINE | ID: mdl-26801829

ABSTRACT

Neurogenesis in mammals occurs throughout life in two brain regions: the ventricular-subventricular zone (V-SVZ) and the subgranular zone (SGZ) of the hippocampal dentate gyrus. Development and regulation of the V-SVZ and SGZ is unique to each brain region, but with several similar characteristics. Alterations to the production of new neurons in neurogenic regions have been linked to psychiatric and neurodegenerative disorders. Decline in neurogenesis in the SGZ correlates with affective and psychiatric disorders, and can be reversed by antidepressant and antipsychotic drugs. Likewise, neurogenesis in the V-SVZ can also be enhanced by antidepressant drugs. The regulation of neurogenesis by neurotransmitters, particularly monoamines, in both regions suggests that aberrant neurotransmitter signaling observed in psychiatric disease may play a role in the pathology of these mental health disorders. Similarly, the cognitive deficits that accompany neurodegenerative disease may also be exacerbated by decreased neurogenesis. This review explores the regulation and function of neural stem cells in rodents and humans, and the involvement of factors that contribute to psychiatric and cognitive deficits. This article is part of a Special Issue entitled SI:StemsCellsinPsychiatry.


Subject(s)
Adult Stem Cells/physiology , Mental Disorders/physiopathology , Neurogenesis/physiology , Animals , Brain/physiopathology , Humans , Neurotransmitter Agents/metabolism
16.
Stem Cells Dev ; 25(7): 542-55, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26857912

ABSTRACT

Neural stem cells (NSCs) exist throughout life in the ventricular-subventricular zone (V-SVZ) of the mammalian forebrain. During aging NSC function is diminished through an unclear mechanism. In this study, we establish microglia, the immune cells of the brain, as integral niche cells within the V-SVZ that undergo age-associated repositioning in the V-SVZ. Microglia become activated early before NSC deficits during aging resulting in an antineurogenic microenvironment due to increased inflammatory cytokine secretion. These age-associated changes were not observed in non-neurogenic brain regions, suggesting V-SVZ microglia are specialized. Using a sustained inflammatory model in young adult mice, we induced microglia activation and inflammation that was accompanied by reduced NSC proliferation in the V-SVZ. Furthermore, in vitro studies revealed secreted factors from activated microglia reduced proliferation and neuron production compared to secreted factors from resting microglia. Our results suggest that age-associated chronic inflammation contributes to declines in NSC function within the aging neurogenic niche.


Subject(s)
Brain/growth & development , Microglia/cytology , Neurogenesis , Stem Cell Niche , Animals , Brain/cytology , Cells, Cultured , Mice , Mice, Inbred C57BL , Microglia/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism
17.
Stem Cells ; 34(1): 220-32, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26369286

ABSTRACT

The ventricular-subventricular zone harbors neural stem cells (NSCs) that can differentiate into neurons, astrocytes, and oligodendrocytes. This process requires loss of stem cell properties and gain of characteristics associated with differentiated cells. miRNAs function as important drivers of this transition; miR-124, -128, and -137 are among the most relevant ones and have been shown to share commonalities and act as proneurogenic regulators. We conducted biological and genomic analyses to dissect their target repertoire during neurogenesis and tested the hypothesis that they act cooperatively to promote differentiation. To map their target genes, we transfected NSCs with antagomiRs and analyzed differences in their mRNA profile throughout differentiation with respect to controls. This strategy led to the identification of 910 targets for miR-124, 216 for miR-128, and 652 for miR-137. The target sets show extensive overlap. Inspection by gene ontology and network analysis indicated that transcription factors are a major component of these miRNAs target sets. Moreover, several of these transcription factors form a highly interconnected network. Sp1 was determined to be the main node of this network and was further investigated. Our data suggest that miR-124, -128, and -137 act synergistically to regulate Sp1 expression. Sp1 levels are dramatically reduced as cells differentiate and silencing of its expression reduced neuronal production and affected NSC viability and proliferation. In summary, our results show that miRNAs can act cooperatively and synergistically to regulate complex biological processes like neurogenesis and that transcription factors are heavily targeted to branch out their regulatory effect.


Subject(s)
Cell Differentiation/genetics , Gene Regulatory Networks , MicroRNAs/genetics , MicroRNAs/metabolism , Neurons/cytology , Neurons/metabolism , Sp1 Transcription Factor/metabolism , Animals , Cell Proliferation , Cell Self Renewal , Gene Expression Regulation , Genome , Humans , Mice , Neural Stem Cells/cytology , Oligonucleotides, Antisense/metabolism , Sequence Analysis, RNA , Transfection
18.
BMC Bioinformatics ; 15: 328, 2014 Oct 03.
Article in English | MEDLINE | ID: mdl-25281197

ABSTRACT

BACKGROUND: Neural stem cells are motile and proliferative cells that undergo mitosis, dividing to produce daughter cells and ultimately generating differentiated neurons and glia. Understanding the mechanisms controlling neural stem cell proliferation and differentiation will play a key role in the emerging fields of regenerative medicine and cancer therapeutics. Stem cell studies in vitro from 2-D image data are well established. Visualizing and analyzing large three dimensional images of intact tissue is a challenging task. It becomes more difficult as the dimensionality of the image data increases to include time and additional fluorescence channels. There is a pressing need for 5-D image analysis and visualization tools to study cellular dynamics in the intact niche and to quantify the role that environmental factors play in determining cell fate. RESULTS: We present an application that integrates visualization and quantitative analysis of 5-D (x,y,z,t,channel) and large montage confocal fluorescence microscopy images. The image sequences show stem cells together with blood vessels, enabling quantification of the dynamic behaviors of stem cells in relation to their vascular niche, with applications in developmental and cancer biology. Our application automatically segments, tracks, and lineages the image sequence data and then allows the user to view and edit the results of automated algorithms in a stereoscopic 3-D window while simultaneously viewing the stem cell lineage tree in a 2-D window. Using the GPU to store and render the image sequence data enables a hybrid computational approach. An inference-based approach utilizing user-provided edits to automatically correct related mistakes executes interactively on the system CPU while the GPU handles 3-D visualization tasks. CONCLUSIONS: By exploiting commodity computer gaming hardware, we have developed an application that can be run in the laboratory to facilitate rapid iteration through biological experiments. We combine unsupervised image analysis algorithms with an interactive visualization of the results. Our validation interface allows for each data set to be corrected to 100% accuracy, ensuring that downstream data analysis is accurate and verifiable. Our tool is the first to combine all of these aspects, leveraging the synergies obtained by utilizing validation information from stereo visualization to improve the low level image processing tasks.


Subject(s)
Algorithms , Cell Lineage , Computer Graphics , Image Processing, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Neural Stem Cells/cytology , Automation , Microscopy, Confocal , Microscopy, Fluorescence , Software
19.
Article in English | MEDLINE | ID: mdl-25570174

ABSTRACT

Biological imaging of live cell and tissue using 3D microscopy is able to capture time-lapse image sequences showing multiple molecular markers labeling different biological structures simultaneously. In order to analyze this complex multi-dimensional image sequence data, there is a need for automated quantitative algorithms, and for methods to visualize and interact with both the data and the analytical results. Traditional computational human input devices such as the keyboard and mouse are no longer adequate for complex tasks such as manipulating and navigating 3+ dimensional volumes. In this paper, we have developed a new interaction system for interfacing with big data sets using the human visual system together with touch, force and audio feedback. This system includes real-time dynamic 3D visualization, haptic interaction via exoskeletal glove, and tonal auditory components that seamlessly create an immersive environment for efficient qualitative analysis.


Subject(s)
Image Processing, Computer-Assisted/methods , Stem Cells/cytology , User-Computer Interface , Algorithms , Biomechanical Phenomena , Feedback , Humans , Imaging, Three-Dimensional , Motion , Software , Tendons/physiology , Touch
20.
Cell Stem Cell ; 11(2): 220-30, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22862947

ABSTRACT

Neurons arise in the adult forebrain subventricular zone (SVZ) from Type B neural stem cells (NSCs), raising considerable interest in the molecules that maintain this life-long neurogenic niche. Type B cells are anchored by specialized apical endfeet in the center of a pinwheel of ependymal cells. Here we show that the apical endfeet express high levels of the adhesion and signaling molecule vascular cell adhesion molecule-1 (VCAM1). Disruption of VCAM1 in vivo causes loss of the pinwheels, disrupted SVZ cytoarchitecture, proliferation and depletion of the normally quiescent apical Type B cells, and increased neurogenesis in the olfactory bulb, demonstrating a key role in niche structure and function. We show that VCAM1 signals via NOX2 production of reactive oxygen species (ROS) to maintain NSCs. VCAM1 on Type B cells is increased by IL-1ß, demonstrating that it can act as an environmental sensor, responding to chemokines involved in tissue repair.


Subject(s)
Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Prosencephalon/cytology , Stem Cell Niche/physiology , Vascular Cell Adhesion Molecule-1/metabolism , Animals , Cell Adhesion , Cells, Cultured , Interleukin-1beta/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , NADPH Oxidase 2 , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...