Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Biol Psychiatry ; 95(2): 187-198, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-37454787

ABSTRACT

BACKGROUND: Converging evidence from large-scale genetic and postmortem studies highlights the role of aberrant neurotransmission and genetic regulation in brain-related disorders. However, identifying neuronal activity-regulated transcriptional programs in the human brain and understanding how changes contribute to disease remain challenging. METHODS: To better understand how the activity-dependent regulome contributes to risk for brain-related disorders, we profiled the transcriptomic and epigenomic changes following neuronal depolarization in human induced pluripotent stem cell-derived glutamatergic neurons (NGN2) from 6 patients with schizophrenia and 5 control participants. RESULTS: Multiomic data integration associated global patterns of chromatin accessibility with gene expression and identified enhancer-promoter interactions in glutamatergic neurons. Within 1 hour of potassium chloride-induced depolarization, independent of diagnosis, glutamatergic neurons displayed substantial activity-dependent changes in the expression of genes regulating synaptic function. Depolarization-induced changes in the regulome revealed significant heritability enrichment for schizophrenia and Parkinson's disease, adding to mounting evidence that sequence variation within activation-dependent regulatory elements contributes to the genetic risk for brain-related disorders. Gene coexpression network analysis elucidated interactions among activity-dependent and disease-associated genes and pointed to a key driver (NAV3) that interacted with multiple genes involved in axon guidance. CONCLUSIONS: Overall, we demonstrated that deciphering the activity-dependent regulome in glutamatergic neurons reveals novel targets for advanced diagnosis and therapy.


Subject(s)
Induced Pluripotent Stem Cells , Schizophrenia , Humans , Induced Pluripotent Stem Cells/metabolism , Gene Expression Regulation , Neurons/metabolism , Brain
2.
Sci Transl Med ; 15(693): eadf1147, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37099632

ABSTRACT

Beta-adrenergic blockade has been associated with improved cancer survival in patients with triple-negative breast cancer (TNBC), but the mechanisms of these effects remain unclear. In clinical epidemiological analyses, we identified a relationship between beta-blocker use and anthracycline chemotherapy in protecting against TNBC progression, disease recurrence, and mortality. We recapitulated the effect of beta-blockade on anthracycline efficacy in xenograft mouse models of TNBC. In metastatic 4T1.2 and MDA-MB-231 mouse models of TNBC, beta-blockade improved the efficacy of the anthracycline doxorubicin by reducing metastatic development. We found that anthracycline chemotherapy alone, in the absence of beta-blockade, increased sympathetic nerve fiber activity and norepinephrine concentration in mammary tumors through the induction of nerve growth factor (NGF) by tumor cells. Moreover, using preclinical models and clinical samples, we found that anthracycline chemotherapy up-regulated ß2-adrenoceptor expression and amplified receptor signaling in tumor cells. Neurotoxin inhibition of sympathetic neural signaling in mammary tumors using 6-hydroxydopamine or genetic deletion of NGF or ß2-adrenoceptor in tumor cells enhanced the therapeutic effect of anthracycline chemotherapy by reducing metastasis in xenograft mouse models. These findings reveal a neuromodulatory effect of anthracycline chemotherapy that undermines its potential therapeutic impact, which can be overcome by inhibiting ß2-adrenergic signaling in the tumor microenvironment. Supplementing anthracycline chemotherapy with adjunctive ß2-adrenergic antagonists represents a potential therapeutic strategy for enhancing the clinical management of TNBC.


Subject(s)
Anthracyclines , Triple Negative Breast Neoplasms , Humans , Animals , Mice , Anthracyclines/pharmacology , Anthracyclines/therapeutic use , Triple Negative Breast Neoplasms/genetics , Nerve Growth Factor/therapeutic use , Cell Line, Tumor , Neoplasm Recurrence, Local/drug therapy , Receptors, Adrenergic/therapeutic use , Tumor Microenvironment
3.
Cell Rep ; 38(3): 110282, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35045295

ABSTRACT

Although circular RNAs (circRNAs) are enriched in the brain, their relevance for brain function and psychiatric disorders is poorly understood. Here, we show that circHomer1 is inversely associated with relative HOMER1B mRNA isoform levels in both the orbitofrontal cortex (OFC) and stem-cell-derived neuronal cultures of subjects with psychiatric disorders. We further demonstrate that in vivo circHomer1 knockdown (KD) within the OFC can inhibit the synaptic expression of Homer1b mRNA. Furthermore, we show that circHomer1 directly binds to Homer1b mRNA and that Homer1b-specific KD increases synaptic circHomer1 levels and improves OFC-mediated behavioral flexibility. Importantly, double circHomer1 and Homer1b in vivo co-KD results in a complete rescue in circHomer1-associated alterations in both chance reversal learning and synaptic gene expression. Lastly, we uncover an RNA-binding protein that can directly bind to circHomer1 and promote its biogenesis. Taken together, our data provide mechanistic insights into the importance of circRNAs in brain function and disease.


Subject(s)
Gene Expression Regulation/physiology , Homer Scaffolding Proteins/metabolism , Prefrontal Cortex/metabolism , RNA, Circular/metabolism , Reversal Learning/physiology , Animals , Bipolar Disorder/metabolism , Gene Knockdown Techniques , Humans , Male , Mice , Mice, Inbred C57BL
4.
Neuron ; 106(6): 912-926.e5, 2020 06 17.
Article in English | MEDLINE | ID: mdl-32304628

ABSTRACT

Depression is a common disorder that affects women at twice the rate of men. Here, we report that long non-coding RNAs (lncRNAs), a recently discovered class of regulatory transcripts, represent about one-third of the differentially expressed genes in the brains of depressed humans and display complex region- and sex-specific patterns of regulation. We identified the primate-specific, neuronal-enriched gene LINC00473 as downregulated in prefrontal cortex (PFC) of depressed females but not males. Using viral-mediated gene transfer to express LINC00473 in adult mouse PFC neurons, we mirrored the human sex-specific phenotype by inducing stress resilience solely in female mice. This sex-specific phenotype was accompanied by changes in synaptic function and gene expression selectively in female mice and, along with studies of human neuron-like cells in culture, implicates LINC00473 as a CREB effector. Together, our studies identify LINC00473 as a female-specific driver of stress resilience that is aberrant in female depression.


Subject(s)
Depressive Disorder, Major/genetics , Prefrontal Cortex/metabolism , RNA, Long Noncoding/genetics , Resilience, Psychological , Stress, Psychological/genetics , Adult , Aged , Aged, 80 and over , Animals , Behavior, Animal , Depression/genetics , Depression/metabolism , Depressive Disorder, Major/metabolism , Down-Regulation , Female , Humans , Male , Mice , Mice, Transgenic , Middle Aged , Neurons/metabolism , RNA, Long Noncoding/metabolism , RNA-Seq , Sex Factors , Stress, Psychological/metabolism , Young Adult
5.
Mol Psychiatry ; 25(11): 2712-2727, 2020 11.
Article in English | MEDLINE | ID: mdl-31988434

ABSTRACT

Although circular RNAs (circRNAs) are enriched in the mammalian brain, very little is known about their potential involvement in brain function and psychiatric disease. Here, we show that circHomer1a, a neuronal-enriched circRNA abundantly expressed in the frontal cortex, derived from Homer protein homolog 1 (HOMER1), is significantly reduced in both the prefrontal cortex (PFC) and induced pluripotent stem cell-derived neuronal cultures from patients with schizophrenia (SCZ) and bipolar disorder (BD). Moreover, alterations in circHomer1a were positively associated with the age of onset of SCZ in both the dorsolateral prefrontal cortex (DLPFC) and orbitofrontal cortex (OFC). No correlations between the age of onset of SCZ and linear HOMER1 mRNA were observed, whose expression was mostly unaltered in BD and SCZ postmortem brain. Using in vivo circRNA-specific knockdown of circHomer1a in mouse PFC, we show that it modulates the expression of numerous alternative mRNA transcripts from genes involved in synaptic plasticity and psychiatric disease. Intriguingly, in vivo circHomer1a knockdown in mouse OFC resulted in specific deficits in OFC-mediated cognitive flexibility. Lastly, we demonstrate that the neuronal RNA-binding protein HuD binds to circHomer1a and can influence its synaptic expression in the frontal cortex. Collectively, our data uncover a novel psychiatric disease-associated circRNA that regulates synaptic gene expression and cognitive flexibility.


Subject(s)
Bipolar Disorder/genetics , Cognition , Gene Expression Regulation , RNA, Circular/genetics , Schizophrenia/genetics , Synapses/metabolism , Adult , Animals , Female , Homer Scaffolding Proteins/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Prefrontal Cortex/metabolism
6.
Oncogene ; 38(26): 5265-5280, 2019 06.
Article in English | MEDLINE | ID: mdl-30914801

ABSTRACT

Farnesyl diphosphate synthase (FDPS), a mevalonate pathway enzyme, is highly expressed in several cancers, including prostate cancer (PCa). To date, the mechanistic, functional, and clinical significance of FDPS in cancer remains unexplored. We evaluated the FDPS expression and its cancer-associated phenotypes using in vitro and in vivo methods in PTEN-deficient and sufficient human and mouse PCa cells and tumors. Interestingly, FDPS overexpression synergizes with PTEN deficiency in PTEN conditionally knockout mice (P < 0.05) and expressed significantly higher in human (P < 0.001) PCa tissues, cell lines, and murine tumoroids compared to respective controls. In silico analysis revealed that FDPS is associated with increasing Gleason score, PTEN functionally deficient status, and poor survival of PCa. Ectopic overexpression of FDPS promotes oncogenic phenotypes such as colony formation (P < 0.01) and proliferation (P < 0.01) through activation of AKT and ERK signaling by prenylating Rho A, Rho G, and CDC42 small GTPases. Of interest, knockdown of FDPS in PCa cells exhibits decreased colony growth and proliferation (P < 0.001) by modulating AKT and ERK pathways. Further, genetic and pharmacological inhibition of PI3K but not AKT reduced FDPS expression. Pharmacological targeting of FDPS by zoledronic acid (ZOL), which is already in clinics, exhibit reduced growth and clonogenicity of human and murine PCa cells (P < 0.01) and 3D tumoroids (P < 0.02) by disrupting AKT and ERK signaling through direct interference of small GTPases protein prenylation. Thus, FDPS plays an oncogenic role in PTEN-deficient PCa through GTPase/AKT axis. Identifying mevalonate pathway proteins could serve as a therapeutic target in PTEN dysregulated tumors.


Subject(s)
Adenocarcinoma/pathology , Geranyltranstransferase/physiology , Monomeric GTP-Binding Proteins/metabolism , PTEN Phosphohydrolase/genetics , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Adenocarcinoma/genetics , Animals , Cell Line, Tumor , Disease Progression , Gene Deletion , Geranyltranstransferase/genetics , Humans , Male , Mice , Mice, Knockout , Prostatic Neoplasms/genetics , Signal Transduction/physiology
7.
Nat Commun ; 9(1): 4926, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30451900

ABSTRACT

In the originally published version of this Article, the affiliation details for Eric E. Schadt and Radoslav Savic incorrectly omitted 'Sema4, a Mount Sinai venture, Stamford, Connecticut, USA'. This has been corrected in both the PDF and HTML versions of the Article.

8.
Nat Commun ; 9(1): 4412, 2018 10 24.
Article in English | MEDLINE | ID: mdl-30356048

ABSTRACT

A lack of biologically relevant screening models hinders the discovery of better treatments for schizophrenia (SZ) and other neuropsychiatric disorders. Here we compare the transcriptional responses of 8 commonly used cancer cell lines (CCLs) directly with that of human induced pluripotent stem cell (hiPSC)-derived neural progenitor cells (NPCs) from 12 individuals with SZ and 12 controls across 135 drugs, generating 4320 unique drug-response transcriptional signatures. We identify those drugs that reverse post-mortem SZ-associated transcriptomic signatures, several of which also differentially regulate neuropsychiatric disease-associated genes in a cell type (hiPSC NPC vs. CCL) and/or a diagnosis (SZ vs. control)-dependent manner. Overall, we describe a proof-of-concept application of transcriptomic drug screening to hiPSC-based models, demonstrating that the drug-induced gene expression differences observed with patient-derived hiPSC NPCs are enriched for SZ biology, thereby revealing a major advantage of incorporating cell type and patient-specific platforms in drug discovery.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Schizophrenia/metabolism , Cell Line , Cell Line, Tumor , Dimethyl Sulfoxide/pharmacology , Humans , Induced Pluripotent Stem Cells/drug effects , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Quality Control , Transcriptome
9.
Exp Lung Res ; 44(1): 13-24, 2018 02.
Article in English | MEDLINE | ID: mdl-29324052

ABSTRACT

Claudins are tight junctional proteins implicated in cell polarity and epithelial barrier maintenance. Claudin misregulation adversely impacts developmental aspects of cell differentiation and proliferation. The current research evaluated transcriptional expression for Claudins 1-11 and 18 in the developing murine lung at embryonic days (E) 14.5, 16.5, and 18.5 and at post-natal day (PN) 3 and PN15. Mouse lungs were also assessed by immunohistochemical analysis to qualitatively evaluate Claudin protein expression. Pregnant dams were further exposed to secondhand smoke (SHS) from embryonic day (E)15.5 to 18.5 and Claudin mRNA was immediately screened in pup lungs. Other than Claudin-6, mRNA expression patterns for Claudin family members tended to decrease at E16.5, increase at E18.5, and decrease again at PN3 before reaching a peak of expression at PN15. Claudin-6 mRNA expression decreased through gestation and into post-natal periods. Immunohistochemical profiling implicated a subset of Claudins as plausible orchestrators of proximal vs. distal lung barrier establishment. Assessment of Claudin mRNA expression at E18.5 following SHS exposure revealed a significant reduction in transcription for all Claudins except Claudin-18 (no change). These data support the need for further studies using gene targeted mice that knock-in/out specific Claudins so that precise functions in the normal and diseased lung can be determined.


Subject(s)
Claudins/biosynthesis , Lung/growth & development , Tobacco Smoke Pollution/adverse effects , Transcriptome/drug effects , Animals , Claudins/drug effects , Claudins/genetics , Embryo, Mammalian , Female , Lung/drug effects , Lung/embryology , Mice , Pregnancy , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Tight Junctions
10.
Nat Commun ; 8(1): 2225, 2017 12 20.
Article in English | MEDLINE | ID: mdl-29263384

ABSTRACT

The power of human induced pluripotent stem cell (hiPSC)-based studies to resolve the smaller effects of common variants within the size of cohorts that can be realistically assembled remains uncertain. We identified and accounted for a variety of technical and biological sources of variation in a large case/control schizophrenia (SZ) hiPSC-derived cohort of neural progenitor cells and neurons. Reducing the stochastic effects of the differentiation process by correcting for cell type composition boosted the SZ signal and increased the concordance with post-mortem data sets. We predict a growing convergence between hiPSC and post-mortem studies as both approaches expand to larger cohort sizes. For studies of complex genetic disorders, to maximize the power of hiPSC cohorts currently feasible, in most cases and whenever possible, we recommend expanding the number of individuals even at the expense of the number of replicate hiPSC clones.


Subject(s)
Brain/metabolism , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Neurons/metabolism , RNA, Messenger/metabolism , Schizophrenia/genetics , Adolescent , Adult , Antigens, Surface/genetics , Autopsy , Case-Control Studies , Child , DNA Copy Number Variations , Female , Humans , Linear Models , Male , Nanog Homeobox Protein/genetics , Nestin/genetics , Octamer Transcription Factor-3/genetics , Proteoglycans/genetics , SOXB1 Transcription Factors/genetics , Sequence Analysis, RNA , Stage-Specific Embryonic Antigens/genetics , Synapsins/genetics , Transcriptome , Young Adult
12.
Stem Cell Reports ; 9(2): 615-628, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28757163

ABSTRACT

Modulation of transcription, either synthetic activation or repression, via dCas9-fusion proteins is a relatively new methodology with the potential to facilitate high-throughput up- or downregulation studies of gene function. Genetic studies of neurodevelopmental disorders have identified a growing list of risk variants, including both common single-nucleotide variants and rare copy-number variations, many of which are associated with genes having limited functional annotations. By applying a CRISPR-mediated gene-activation/repression platform to populations of human-induced pluripotent stem cell-derived neural progenitor cells, neurons, and astrocytes, we demonstrate that it is possible to manipulate endogenous expression levels of candidate neuropsychiatric risk genes across these three cell types. Although proof-of-concept studies using catalytically inactive Cas9-fusion proteins to modulate transcription have been reported, here we present a detailed survey of the reproducibility of gRNA positional effects across a variety of neurodevelopmental disorder-relevant risk genes, donors, neural cell types, and dCas9 effectors.


Subject(s)
Astrocytes/cytology , Astrocytes/metabolism , Induced Pluripotent Stem Cells/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Calcium/metabolism , Cell Differentiation , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Induced Pluripotent Stem Cells/metabolism , Molecular Imaging , Transcriptome
13.
Stem Cell Reports ; 9(2): 600-614, 2017 08 08.
Article in English | MEDLINE | ID: mdl-28757165

ABSTRACT

Growing evidence implicates the importance of glia, particularly astrocytes, in neurological and psychiatric diseases. Here, we describe a rapid and robust method for the differentiation of highly pure populations of replicative astrocytes from human induced pluripotent stem cells (hiPSCs), via a neural progenitor cell (NPC) intermediate. We evaluated this protocol across 42 NPC lines (derived from 30 individuals). Transcriptomic analysis demonstrated that hiPSC-astrocytes from four individuals are highly similar to primary human fetal astrocytes and characteristic of a non-reactive state. hiPSC-astrocytes respond to inflammatory stimulants, display phagocytic capacity, and enhance microglial phagocytosis. hiPSC-astrocytes also possess spontaneous calcium transient activity. Our protocol is a reproducible, straightforward (single medium), and rapid (<30 days) method to generate populations of hiPSC-astrocytes that can be used for neuron-astrocyte and microglia-astrocyte co-cultures for the study of neuropsychiatric disorders.


Subject(s)
Astrocytes/cytology , Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Astrocytes/metabolism , Calcium/metabolism , Cell Culture Techniques , Cell Differentiation/genetics , Cells, Cultured , Computational Biology/methods , Cytokines/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Induced Pluripotent Stem Cells/metabolism , Microglia/immunology , Microglia/metabolism , Molecular Sequence Annotation , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Phagocytosis , Transcriptome
14.
Cell Stem Cell ; 21(2): 274-283.e5, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28736217

ABSTRACT

Zika virus (ZIKV) infects fetal and adult human brain and is associated with serious neurological complications. To date, no therapeutic treatment is available to treat ZIKV-infected patients. We performed a high-content chemical screen using human pluripotent stem cell-derived cortical neural progenitor cells (hNPCs) and found that hippeastrine hydrobromide (HH) and amodiaquine dihydrochloride dihydrate (AQ) can inhibit ZIKV infection in hNPCs. Further validation showed that HH also rescues ZIKV-induced growth and differentiation defects in hNPCs and human fetal-like forebrain organoids. Finally, HH and AQ inhibit ZIKV infection in adult mouse brain in vivo. Strikingly, HH suppresses viral propagation when administered to adult mice with active ZIKV infection, highlighting its therapeutic potential. Our approach highlights the power of stem cell-based screens and validation in human forebrain organoids and mouse models in identifying drug candidates for treating ZIKV infection and related neurological complications in fetal and adult patients.


Subject(s)
Antiviral Agents/therapeutic use , Brain/virology , Drug Evaluation, Preclinical/methods , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/metabolism , Organoids/virology , Zika Virus Infection/drug therapy , Zika Virus/physiology , Adolescent , Amaryllidaceae Alkaloids/pharmacology , Amodiaquine/pharmacology , Animals , Antiviral Agents/pharmacology , Cell Line , Child , Female , Fetus/drug effects , Fetus/virology , Humans , Induced Pluripotent Stem Cells/drug effects , Mice, SCID , Neural Stem Cells/drug effects , Organoids/drug effects , Zika Virus/drug effects , Zika Virus Infection/pathology
15.
Nat Genet ; 49(8): 1239-1250, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28671686

ABSTRACT

We report locus-specific disintegration of megabase-scale chromosomal conformations in brain after neuronal ablation of Setdb1 (also known as Kmt1e; encodes a histone H3 lysine 9 methyltransferase), including a large topologically associated 1.2-Mb domain conserved in humans and mice that encompasses >70 genes at the clustered protocadherin locus (hereafter referred to as cPcdh). The cPcdh topologically associated domain (TADcPcdh) in neurons from mutant mice showed abnormal accumulation of the transcriptional regulator and three-dimensional (3D) genome organizer CTCF at cryptic binding sites, in conjunction with DNA cytosine hypomethylation, histone hyperacetylation and upregulated expression. Genes encoding stochastically expressed protocadherins were transcribed by increased numbers of cortical neurons, indicating relaxation of single-cell constraint. SETDB1-dependent loop formations bypassed 0.2-1 Mb of linear genome and radiated from the TADcPcdh fringes toward cis-regulatory sequences within the cPcdh locus, counterbalanced shorter-range facilitative promoter-enhancer contacts and carried loop-bound polymorphisms that were associated with genetic risk for schizophrenia. We show that the SETDB1 repressor complex, which involves multiple KRAB zinc finger proteins, shields neuronal genomes from excess CTCF binding and is critically required for structural maintenance of TADcPcdh.


Subject(s)
Chromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Neurons/metabolism , Animals , CCCTC-Binding Factor , Cadherins/genetics , Cell Line , DNA Methylation , Epigenesis, Genetic , Female , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/genetics , Humans , Male , Mice , Mutation , Nucleic Acid Conformation , Protein Binding , Protein Domains , Repressor Proteins/metabolism
16.
Article in English | MEDLINE | ID: mdl-28149327

ABSTRACT

BACKGROUND: Intrauterine exposure to maternal smoking is linked to impaired executive function and behavioral problems in the offspring. Maternal smoking is associated with reduced fetal brain growth and smaller volume of cortical gray matter in childhood, indicating that prenatal exposure to tobacco may impact cortical development and manifest as behavioral problems. Cellular development is mediated by changes in epigenetic modifications such as DNA methylation, which can be affected by exposure to tobacco. RESULTS: In this study, we sought to ascertain how maternal smoking during pregnancy affects global DNA methylation profiles of the developing dorsolateral prefrontal cortex (DLPFC) during the second trimester of gestation. When DLPFC methylation profiles (assayed via Illumina, HM450) of smoking-exposed and unexposed fetuses were compared, no differentially methylated regions (DMRs) passed the false discovery correction (FDR ≤ 0.05). However, the most significant DMRs were hypomethylated CpG Islands within the promoter regions of GNA15 and SDHAP3 of smoking-exposed fetuses. Interestingly, the developmental up-regulation of SDHAP3 mRNA was delayed in smoking-exposed fetuses. Interaction analysis between gestational age and smoking exposure identified significant DMRs annotated to SYCE3, C21orf56/LSS, SPAG1 and RNU12/POLDIP3 that passed FDR. Furthermore, utilizing established methods to estimate cell proportions by DNA methylation, we found that exposed DLPFC samples contained a lower proportion of neurons in samples from fetuses exposed to maternal smoking. We also show through in vitro experiments that nicotine impedes the differentiation of neurons independent of cell death. CONCLUSIONS: We found evidence that intrauterine smoking exposure alters the developmental patterning of DNA methylation and gene expression and is associated with reduced mature neuronal content, effects that are likely driven by nicotine.


Subject(s)
Brain/metabolism , DNA Methylation , Maternal Exposure , Smoking , Brain/pathology , Female , Fetal Development/genetics , Fetus/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Gestational Age , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells/metabolism , Male , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/metabolism , Pregnancy , Pregnancy Trimester, Second , Promoter Regions, Genetic , Succinate Dehydrogenase/genetics , Tubulin/genetics , Tubulin/metabolism
17.
Neurochem Int ; 106: 85-93, 2017 Jun.
Article in English | MEDLINE | ID: mdl-27744003

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) can theoretically yield limitless supplies of cells fated to any cell type that comprise the human organism, making them a new tool by which to potentially overcome caveats in current biomedical research. In vitro derivation of central nervous system (CNS) cell types has the potential to provide material for drug discovery and validation, safety and toxicity assays, cell replacement therapy and the elucidation of previously unknown disease mechanisms. However, current two-dimensional (2D) CNS differentiation protocols do not faithfully recapitulate the spatial organization of heterogeneous tissue, nor the cell-cell interactions, cell-extracellular matrix interactions, or specific physiological functions generated within complex tissue such as the brain. In an effort to overcome 2D protocol limitations, there have been advancements in deriving highly complicated 3D neural organoid structures. Herein we provide a synopsis of the derivation and application of neural organoids and discuss recent advancements and remaining challenges on the full potential of this novel technological platform.


Subject(s)
Developmental Biology/trends , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/physiology , Organoids/drug effects , Organoids/physiology , Phenotype , Animals , Cell Culture Techniques/methods , Cell Culture Techniques/trends , Developmental Biology/methods , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/trends , Humans , Pharmaceutical Preparations/administration & dosage , Toxicity Tests/methods , Toxicity Tests/trends
18.
Article in English | MEDLINE | ID: mdl-27763528

ABSTRACT

It has long been understood that increased epithelial permeability contributes to inflammation observed in many respiratory diseases. Recently, evidence has revealed that environmental exposure to noxious material such as cigarette smoke reduces tight junction barrier integrity, thus enhancing inflammatory conditions. Claudin-6 (Cldn6) is a tetraspanin transmembrane protein found within the tight junctional complex and is implicated in maintaining lung epithelial barriers. To test the hypothesis that increased Cldn6 ameliorates inflammation at the respiratory barrier, we utilized the Tet-On inducible transgenic system to conditionally over-express Clnd6 in the distal lung. Cldn6 transgenic (TG) and control mice were continuously provided doxycycline from postnatal day (PN) 30 until euthanasia date at PN90. A subset of Cldn6 TG and control mice were also subjected to daily secondhand tobacco smoke (SHS) via a nose only inhalation system from PN30-90 and compared to room air (RA) controls. Animals were euthanized on PN90 and lungs were harvested for histological and molecular characterization. Bronchoalveolar lavage fluid (BALF) was procured for the assessment of inflammatory cells and molecules. Quantitative RT-PCR and immunoblotting revealed increased Cldn6 expression in TG vs. control animals and SHS decreased Cldn6 expression regardless of genetic up-regulation. Histological evaluations revealed no adverse pulmonary remodeling via Hematoxylin and Eosin (H&E) staining or any qualitative alterations in the abundance of type II pneumocytes or proximal non-ciliated epithelial cells via staining for cell specific propeptide of Surfactant Protein-C (proSP-C) or Club Cell Secretory Protein (CCSP), respectively. Immunoblotting and qRT-PCR confirmed the differential expression of Cldn6 and the pro-inflammatory cytokines TNF-α and IL-1ß. As a general theme, inflammation induced by SHS exposure was influenced by the availability of Cldn6. These data reveal captivating information suggesting a role for Cldn6 in lungs exposed to tobacco smoke. Further research is critically necessary in order to fully explain roles for tight junctional components such as Cldn6 and other related molecules in lungs coping with exposure.


Subject(s)
Claudins/metabolism , Inflammation/chemically induced , Lung/drug effects , Tobacco Smoke Pollution/adverse effects , Animals , Bronchoalveolar Lavage Fluid , Cytokines/metabolism , Gene Expression Regulation , Interleukin-1beta , Lung/metabolism , Mice , Smoke , Nicotiana/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
19.
Cell Rep ; 15(5): 1024-1036, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27117414

ABSTRACT

Converging evidence indicates that microRNAs (miRNAs) may contribute to disease risk for schizophrenia (SZ). We show that microRNA-9 (miR-9) is abundantly expressed in control neural progenitor cells (NPCs) but also significantly downregulated in a subset of SZ NPCs. We observed a strong correlation between miR-9 expression and miR-9 regulatory activity in NPCs as well as between miR-9 levels/activity, neural migration, and diagnosis. Overexpression of miR-9 was sufficient to ameliorate a previously reported neural migration deficit in SZ NPCs, whereas knockdown partially phenocopied aberrant migration in control NPCs. Unexpectedly, proteomic- and RNA sequencing (RNA-seq)-based analysis revealed that these effects were mediated primarily by small changes in expression of indirect miR-9 targets rather than large changes in direct miR-9 targets; these indirect targets are enriched for migration-associated genes. Together, these data indicate that aberrant levels and activity of miR-9 may be one of the many factors that contribute to SZ risk, at least in a subset of patients.


Subject(s)
Gene Expression Regulation , MicroRNAs/genetics , Neural Stem Cells/metabolism , Schizophrenia/genetics , Schizophrenia/pathology , Case-Control Studies , Cell Movement/genetics , Gene Expression Profiling , Genetic Association Studies , Humans , Induced Pluripotent Stem Cells/metabolism , MicroRNAs/metabolism , Models, Biological , Molecular Sequence Annotation , Neural Stem Cells/pathology , Neurons/metabolism , Proteome/metabolism , Transcription Factors/metabolism
20.
Methods ; 101: 113-24, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26626326

ABSTRACT

Since the discovery of somatic reprogramming, human induced pluripotent stem cells (hiPSCs) have been exploited to model a variety of neurological and psychiatric disorders. Because hiPSCs represent an almost limitless source of patient-derived neurons that retain the genetic variations thought to contribute to disease etiology, they have been heralded as a patient-specific platform for high throughput drug screening. However, the utility of current protocols for generating neurons from hiPSCs remains limited by protracted differentiation timelines and heterogeneity of the neuronal phenotypes produced. Neuronal induction via the forced expression of exogenous transcription factors rapidly induces defined populations of functional neurons from fibroblasts and hiPSCs. Here, we describe an adapted protocol that accelerates maturation of functional excitatory neurons from hiPSC-derived neural progenitor cells (NPCs) via lentiviral transduction of Neurogenin 2 (using both mNgn2 and hNGN2). This methodology, relying upon a robust and scalable starting population of hiPSC NPCs, should be readily amenable to scaling for hiPSC-based high-throughput drug screening.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Induced Pluripotent Stem Cells/physiology , Nerve Tissue Proteins/physiology , Cell Culture Techniques , Cells, Cultured , Cellular Reprogramming , Gene Expression , Humans , Lentivirus , Neural Stem Cells/physiology , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL