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

Publication year range
1.
J Neurosci ; 33(29): 11839-51, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-23864674

ABSTRACT

Little is known about chromosomal loopings involving proximal promoter and distal enhancer elements regulating GABAergic gene expression, including changes in schizophrenia and other psychiatric conditions linked to altered inhibition. Here, we map in human chromosome 2q31 the 3D configuration of 200 kb of linear sequence encompassing the GAD1 GABA synthesis enzyme gene locus, and we describe a loop formation involving the GAD1 transcription start site and intergenic noncoding DNA elements facilitating reporter gene expression. The GAD1-TSS(-50kbLoop) was enriched with nucleosomes epigenetically decorated with the transcriptional mark, histone H3 trimethylated at lysine 4, and was weak or absent in skin fibroblasts and pluripotent stem cells compared with neuronal cultures differentiated from them. In the prefrontal cortex of subjects with schizophrenia, GAD1-TSS(-50kbLoop) was decreased compared with controls, in conjunction with downregulated GAD1 expression. We generated transgenic mice expressing Gad2 promoter-driven green fluorescent protein-conjugated histone H2B and confirmed that Gad1-TSS(-55kbLoop), the murine homolog to GAD1-TSS(-50kbLoop), is a chromosomal conformation specific for GABAergic neurons. In primary neuronal culture, Gad1-TSS(-55kbLoop) and Gad1 expression became upregulated when neuronal activity was increased. We conclude that 3D genome architectures, including chromosomal loopings for promoter-enhancer interactions involved in the regulation of GABAergic gene expression, are conserved between the rodent and primate brain, and subject to developmental and activity-dependent regulation, and disordered in some cases with schizophrenia. More broadly, the findings presented here draw a connection between noncoding DNA, spatial genome architecture, and neuronal plasticity in development and disease.


Subject(s)
Glutamate Decarboxylase/genetics , Prefrontal Cortex/metabolism , Schizophrenia/genetics , Animals , Antipsychotic Agents/pharmacology , Cells, Cultured , Chromosomes, Human, Pair 2 , Clozapine/pharmacology , DNA Methylation , Down-Regulation , Fibroblasts/metabolism , Gene Expression Regulation , Glutamate Decarboxylase/metabolism , Haloperidol/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Histones/genetics , Histones/metabolism , Humans , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , Schizophrenia/metabolism
2.
Differentiation ; 83(4): 169-78, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22381624

ABSTRACT

While the pathologies associated with in utero smoke exposure are well established, their underlying molecular mechanisms are incompletely understood. We differentiated human embryonic stem cells in the presence of physiological concentrations of tobacco smoke and nicotine. Using post hoc microarray analysis, quantitative PCR, and immunoblot analysis, we demonstrated that tobacco smoke has lineage- and stage-specific effects on human embryonic stem cell differentiation, through both nicotine-dependent and -independent pathways. We show that three major stem cell pluripotency/differentiation pathways, Notch, canonical Wnt, and transforming growth factor-ß, are affected by smoke exposure, and that Nodal signaling through SMAD2 is specifically impacted by effects on Lefty1, Nodal, and FoxH1. These events are associated with upregulation of microRNA-302a, a post-transcriptional silencer of Lefty1. The described studies provide insight into the mechanisms by which tobacco smoke influences fetal development at the cellular level, and identify specific transcriptional, post-transcriptional, and signaling pathways by which this likely occurs.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Nicotiana , Nodal Protein/physiology , Smoke , Transforming Growth Factor beta/physiology , Blotting, Western , Humans , Real-Time Polymerase Chain Reaction
3.
PLoS Pathog ; 5(5): e1000444, 2009 May.
Article in English | MEDLINE | ID: mdl-19461888

ABSTRACT

It is widely believed that innate immune responses to Borrelia burgdorferi (Bb) are primarily triggered by the spirochete's outer membrane lipoproteins signaling through cell surface TLR1/2. We recently challenged this notion by demonstrating that phagocytosis of live Bb by peripheral blood mononuclear cells (PBMCs) elicited greater production of proinflammatory cytokines than did equivalent bacterial lysates. Using whole genome microarrays, we show herein that, compared to lysates, live spirochetes elicited a more intense and much broader transcriptional response involving genes associated with diverse cellular processes; among these were IFN-beta and a number of interferon-stimulated genes (ISGs), which are not known to result from TLR2 signaling. Using isolated monocytes, we demonstrated that cell activation signals elicited by live Bb result from cell surface interactions and uptake and degradation of organisms within phagosomes. As with PBCMs, live Bb induced markedly greater transcription and secretion of TNF-alpha, IL-6, IL-10 and IL-1beta in monocytes than did lysates. Secreted IL-18, which, like IL-1beta, also requires cleavage by activated caspase-1, was generated only in response to live Bb. Pro-inflammatory cytokine production by TLR2-deficient murine macrophages was only moderately diminished in response to live Bb but was drastically impaired against lysates; TLR2 deficiency had no significant effect on uptake and degradation of spirochetes. As with PBMCs, live Bb was a much more potent inducer of IFN-beta and ISGs in isolated monocytes than were lysates or a synthetic TLR2 agonist. Collectively, our results indicate that the enhanced innate immune responses of monocytes following phagocytosis of live Bb have both TLR2-dependent and -independent components and that the latter induce transcription of type I IFNs and ISGs.


Subject(s)
Borrelia Infections/immunology , Borrelia burgdorferi/immunology , Interferon-beta/immunology , Monocytes/immunology , Toll-Like Receptor 2/immunology , Adult , Animals , Bacteriolysis , Cells, Cultured , Cytokines/genetics , Cytokines/immunology , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Interferon-beta/biosynthesis , Interleukin-18/immunology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Monocytes/metabolism , Oligonucleotide Array Sequence Analysis , Signal Transduction , Toll-Like Receptor 5/immunology , Transcription, Genetic
4.
Sci Transl Med ; 13(586)2021 03 24.
Article in English | MEDLINE | ID: mdl-33762438

ABSTRACT

A substantial number of patients with leukemia and lymphoma treated with anti-CD19 or anti-CD22 monoCAR-T cell therapy relapse because of antigen loss or down-regulation. We hypothesized that B cell tumor antigen escape may be overcome by a chimeric antigen receptor (CAR) design that simultaneously targets three B cell leukemia antigens. We engineered trispecific duoCAR-T cells with lentiviral vectors encoding two CAR open reading frames that target CD19, CD20, and CD22. The duoCARs were composed of a CAR with a tandem CD19- and CD20-targeting binder, linked by the P2A self-cleaving peptide to a second CAR targeting CD22. Multiple combinations of intracellular T cell signaling motifs were evaluated. The most potent duoCAR architectures included those with ICOS, OX40, or CD27 signaling domains rather than those from CD28 or 4-1BB. We identified four optimal binder and signaling combinations that potently rejected xenografted leukemia and lymphoma tumors in vivo. Moreover, in mice bearing a mixture of B cell lymphoma lines composed of parental triple-positive cells, CD19-negative, CD20-negative, and CD22-negative variants, only the trispecific duoCAR-T cells rapidly and efficiently rejected the tumors. Each of the monoCAR-T cells failed to prevent tumor progression. Analysis of intracellular signaling profiles demonstrates that the distinct signaling of the intracellular domains used may contribute to these differential effects. Multispecific duoCAR-T cells are a promising strategy to prevent antigen loss-mediated relapse or the down-regulation of target antigen in patients with B cell malignancies.


Subject(s)
Immunotherapy, Adoptive , Lymphoma, B-Cell , Animals , Antigens, CD19 , B-Lymphocytes , Humans , Lymphoma, B-Cell/therapy , Mice , Receptors, Antigen, T-Cell , Sialic Acid Binding Ig-like Lectin 2 , T-Lymphocytes
5.
Sci Rep ; 11(1): 389, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431989

ABSTRACT

Lentiviral vectors (LV) have emerged as a robust technology for therapeutic gene delivery into human cells as advanced medicinal products. As these products are increasingly commercialized, there are concomitant demands for their characterization to ensure safety, efficacy and consistency. Standards are essential for accurately measuring parameters for such product characterization. A critical parameter is the vector copy number (VCN) which measures the genetic dose of a transgene present in gene-modified cells. Here we describe a set of clonal Jurkat cell lines with defined copy numbers of a reference lentiviral vector integrated into their genomes. Genomic DNA was characterized for copy number, genomic integrity and integration coordinates and showed uniform performance across independent quantitative PCR assays. Stability studies during continuous long-term culture demonstrated sustained renewability of the reference standard source material. DNA from the Jurkat VCN standards would be useful for control of quantitative PCR assays for VCN determination in LV gene-modified cellular products and clinical samples.


Subject(s)
Gene Dosage , Lentivirus/genetics , Transduction, Genetic , Calibration/standards , Gene Transfer Techniques/standards , Genetic Vectors/genetics , Humans , Jurkat Cells , Mutagenesis, Insertional/genetics , Reference Standards , Reproducibility of Results , Transduction, Genetic/methods , Transduction, Genetic/standards , Transfection/methods , Transfection/standards , Validation Studies as Topic , Virus Integration/genetics
6.
Cancers (Basel) ; 13(19)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34638227

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has emerged as an attractive strategy for cancer immunotherapy. Despite remarkable success for hematological malignancies, excessive activity and poor control of CAR T cells can result in severe adverse events requiring control strategies to improve safety. This work illustrates the feasibility of a zinc finger-based inducible switch system for transcriptional regulation of an anti-CD20 CAR in primary T cells providing small molecule-inducible control over therapeutic functions. We demonstrate time- and dose-dependent induction of anti-CD20 CAR expression and function with metabolites of the clinically-approved drug tamoxifen, and the absence of background CAR activity in the non-induced state. Inducible CAR T cells executed fine-tuned cytolytic activity against target cells both in vitro and in vivo, whereas CAR-related functions were lost upon drug discontinuation. This zinc finger-based transcriptional control system can be extended to other therapeutically important CARs, thus paving the way for safer cellular therapies.

7.
Stem Cells ; 27(7): 1538-47, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19544450

ABSTRACT

Mouse embryonic stem cells (ESCs) proliferate with rapid cell cycle kinetics but without loss of pluripotency. The histone methyltransferase Dot1L is responsible for methylation of histone H3 at lysine 79 (H3K79me). We investigated whether ESCs require Dot1L for proper stem cell behavior. ESCs deficient in Dot1L tolerate a nearly complete loss of H3K79 methylation without a substantial impact on proliferation or morphology. However, shortly after differentiation is induced, Dot1L-deficient cells cease proliferating and arrest in G2/M-phase of the cell cycle, with increased levels of aneuploidy. In addition, many aberrant mitotic spindles occur in Dot1L-deficient cells. Surprisingly, these mitotic and cell cycle defects fail to trigger apoptosis, indicating that mouse ESCs lack stringent cell cycle checkpoint control during initial stages of differentiation. Transcriptome analysis indicates that Dot1L deficiency causes the misregulation of a select set of genes, including many with known roles in cell cycle control and cellular proliferation as well as markers of endoderm differentiation. The data indicate a requirement for Dot1L function for early stages of ESC differentiation where Dot1L is necessary for faithful execution of mitosis and proper transcription of many genes throughout the genome.


Subject(s)
Apoptosis/physiology , Cell Cycle/physiology , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Methyltransferases/physiology , Animals , Apoptosis/genetics , Cell Cycle/genetics , Cell Differentiation/genetics , Cell Proliferation , Chromatin Immunoprecipitation , Histone-Lysine N-Methyltransferase , Karyotyping , Methyltransferases/genetics , Mice , Polymerase Chain Reaction , RNA Interference
8.
Nat Med ; 26(10): 1569-1575, 2020 10.
Article in English | MEDLINE | ID: mdl-33020647

ABSTRACT

Chimeric antigen receptor (CAR) T cells targeting CD19 are a breakthrough treatment for relapsed, refractory B cell malignancies1-5. Despite impressive outcomes, relapse with CD19- disease remains a challenge. We address this limitation through a first-in-human trial of bispecific anti-CD20, anti-CD19 (LV20.19) CAR T cells for relapsed, refractory B cell malignancies. Adult patients with B cell non-Hodgkin lymphoma or chronic lymphocytic leukemia were treated on a phase 1 dose escalation and expansion trial (NCT03019055) to evaluate the safety of 4-1BB-CD3ζ LV20.19 CAR T cells and the feasibility of on-site manufacturing using the CliniMACS Prodigy system. CAR T cell doses ranged from 2.5 × 105-2.5 × 106 cells per kg. Cell manufacturing was set at 14 d with the goal of infusing non-cryopreserved LV20.19 CAR T cells. The target dose of LV20.19 CAR T cells was met in all CAR-naive patients, and 22 patients received LV20.19 CAR T cells on protocol. In the absence of dose-limiting toxicity, a dose of 2.5 × 106 cells per kg was chosen for expansion. Grade 3-4 cytokine release syndrome occurred in one (5%) patient, and grade 3-4 neurotoxicity occurred in three (14%) patients. Eighteen (82%) patients achieved an overall response at day 28, 14 (64%) had a complete response, and 4 (18%) had a partial response. The overall response rate to the dose of 2.5 × 106 cells per kg with non-cryopreserved infusion (n = 12) was 100% (complete response, 92%; partial response, 8%). Notably, loss of the CD19 antigen was not seen in patients who relapsed or experienced treatment failure. In conclusion, on-site manufacturing and infusion of non-cryopreserved LV20.19 CAR T cells were feasible and therapeutically safe, showing low toxicity and high efficacy. Bispecific CARs may improve clinical responses by mitigating target antigen downregulation as a mechanism of relapse.


Subject(s)
Antigens, CD19/immunology , Antigens, CD20/immunology , Immunotherapy, Adoptive/methods , Leukemia, B-Cell/therapy , Lymphoma, B-Cell/therapy , Adult , Aged , Dose-Response Relationship, Immunologic , Female , Humans , Leukemia, B-Cell/immunology , Leukemia, B-Cell/pathology , Lymphocyte Count , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Male , Middle Aged , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , Recurrence , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation
9.
Front Immunol ; 11: 1941, 2020.
Article in English | MEDLINE | ID: mdl-32849651

ABSTRACT

Chimeric antigen receptor T cells (CAR-T cell) targeting CD19 are effective against several subtypes of CD19-expressing hematologic malignancies. Centralized manufacturing has allowed rapid expansion of this cellular therapy, but it may be associated with treatment delays due to the required logistics. We hypothesized that point of care manufacturing of CAR-T cells on the automated CliniMACS Prodigy® device allows reproducible and fast delivery of cells for the treatment of patients with non-Hodgkin lymphoma. Here we describe cell manufacturing results and characterize the phenotype and effector function of CAR-T cells used in a phase I/II study. We utilized a lentiviral vector delivering a second-generation CD19 CAR construct with 4-1BB costimulatory domain and TNFRSF19 transmembrane domain. Our data highlight the successful generation of CAR-T cells at numbers sufficient for all patients treated, a shortened duration of production from 12 to 8 days followed by fresh infusion into patients, and the detection of CAR-T cells in patient circulation up to 1-year post-infusion.


Subject(s)
Antigens, CD19/immunology , Cell Engineering , Immunotherapy, Adoptive , Lymphoma, Non-Hodgkin/therapy , Point-of-Care Systems , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/transplantation , Animals , Antigens, CD19/genetics , Antigens, CD19/metabolism , Automation , Cell Culture Techniques , Cells, Cultured , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Cytotoxicity, Immunologic , Humans , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/metabolism , Mice, Inbred NOD , Phenotype , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transplantation, Autologous , Treatment Outcome , Workload , Xenograft Model Antitumor Assays
10.
Birth Defects Res C Embryo Today ; 87(4): 297-313, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19960551

ABSTRACT

Following fertilization, the newly formed zygote faces several critical decisions regarding cell fate and lineage commitment. First, the parental genomes must be reprogrammed and reset for the zygotic genome to assume responsibility for gene expression. Second, blastomeres must be committed to form either the inner cell mass or trophectoderm before implantation. A variety of epigenetic mechanisms underlies each of these steps, allowing for proper activation of transcriptional circuits which function to specify a cell's identity and maintain or adjust that state as developmental and environmental conditions dictate. These epigenetic mechanisms encompass DNA methylation, post-translational histone modification, chromatin remodeling, and alterations in nuclear architecture. In recent years, stem cells derived from the inner cell mass have been used to examine the epigenetic pathways that regulate pluripotency, differentiation, and lineage commitment. From a technical standpoint, embryonic stem cells provide an easier system to work with compared to preimplantation embryos; however, it is currently unknown how closely the epigenetic mechanisms of cultured stem cells resemble their counterparts in the intact embryo. Furthermore, it remains unclear how similar the reprogramming pathways in artificially created systems, such as nuclear transfer-derived embryos and induced pluripotent stem cells, are to those in naturally created embryos. In this review, we summarize the current knowledge of epigenetic influences during preimplantation development and shed light on the extent to which these pathways are conserved in cultured pluripotent cells in vitro. In doing so, we demonstrate the critical role that epigenetic mechanisms play in the establishment of cell fate during the earliest stages of mammalian development.


Subject(s)
Embryonic Development/genetics , Epigenesis, Genetic , Animals , Chromatin Assembly and Disassembly , DNA Methylation , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental , Histones/metabolism , Humans , Models, Genetic , Oocytes/metabolism , Pluripotent Stem Cells/metabolism , Pregnancy , RNA, Untranslated/genetics , X Chromosome Inactivation , Zygote/metabolism
11.
Front Immunol ; 10: 2001, 2019.
Article in English | MEDLINE | ID: mdl-31507603

ABSTRACT

Genetic engineering is an important tool for redirecting the function of various types of immune cells and their use for therapeutic purpose. Although NK cells have many beneficial therapeutic features, genetic engineering of immune cells for targeted therapy focuses mostly on T cells. One of the major obstacles for NK cell immunotherapy is the lack of an efficient method for gene transfer. Lentiviral vectors have been proven to be a safe tool for genetic engineering, however lentiviral transduction is inefficient for NK cells. We show in this study that lentiviral vectors pseudotyped with a modified baboon envelope glycoprotein can transduce NK cells 20-fold or higher in comparison to VSV-G pseudotyped lentiviral vector. When we investigated the mechanism of transduction, we found that activated NK cells expressed baboon envelope receptor ASCT-2. Further analysis revealed that only a subset of NK cells could be expanded and transduced with an expression profile of NK56bright, CD16dim, TRAILhigh, and CX3CR1neg. Using CD19-CAR, we could show that CD19 redirected NK cells efficiently and specifically kill cell lines expressing CD19. Taken together, the results from this study will be important for future genetic modification and for redirecting of NK cell function for therapeutic purpose.

13.
Sci Transl Med ; 11(504)2019 08 07.
Article in English | MEDLINE | ID: mdl-31391322

ABSTRACT

Adoptive immunotherapy using chimeric antigen receptor-modified T cells (CAR-T) has made substantial contributions to the treatment of certain B cell malignancies. Such treatment modalities could potentially obviate the need for long-term antiretroviral drug therapy in HIV/AIDS. Here, we report the development of HIV-1-based lentiviral vectors that encode CARs targeting multiple highly conserved sites on the HIV-1 envelope glycoprotein using a two-molecule CAR architecture, termed duoCAR. We show that transduction with lentiviral vectors encoding multispecific anti-HIV duoCARs confer primary T cells with the capacity to potently reduce cellular HIV infection by up to 99% in vitro and >97% in vivo. T cells are the targets of HIV infection, but the transduced T cells are protected from genetically diverse HIV-1 strains. The CAR-T cells also potently eliminated PBMCs infected with broadly neutralizing antibody-resistant HIV strains, including VRC01/3BNC117-resistant HIV-1. Furthermore, multispecific anti-HIV duoCAR-T cells demonstrated long-term control of HIV infection in vivo and prevented the loss of CD4+ T cells during HIV infection using a humanized NSG mouse model of intrasplenic HIV infection. These data suggest that multispecific anti-HIV duoCAR-T cells could be an effective approach for the treatment of patients with HIV-1 infection.


Subject(s)
Antiviral Agents/therapeutic use , HIV Infections/immunology , HIV Infections/therapy , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/immunology , Animals , Antibodies, Neutralizing/immunology , Cells, Cultured , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Disease Models, Animal , HIV-1/immunology , Humans , Lentivirus/metabolism , Lymphocyte Activation/immunology , Lysosomal-Associated Membrane Protein 1/metabolism , Mice , T-Lymphocytes/immunology , Th1 Cells/metabolism , env Gene Products, Human Immunodeficiency Virus/metabolism
14.
J Mol Neurosci ; 32(2): 111-9, 2007.
Article in English | MEDLINE | ID: mdl-17873295

ABSTRACT

Certain neurobehavioral deficiencies associated with Turner Syndrome have been attributed to brain volumetric abnormalities, particularly of the amygdala. Haplo-insufficiency of a non-dosage compensated gene or genes on the X chromosome has been hypothesized to be the cause of the neuroanatomical defect. We examined gene expression levels of 6,628 genes in developing amygdalae of late-stage embryos of a mouse model for Turner Syndrome. In total, 161 genes show significant differences in expression level between TS and normal female amygdala. In silico pathway analysis of both X-linked and autosomal mis-regulated genes suggests that modulation of Wnt signaling is a critical factor in the normal growth and development of the amygdala.


Subject(s)
Amygdala/physiopathology , Turner Syndrome/genetics , Wnt Proteins/genetics , X Chromosome , Amygdala/growth & development , Animals , DNA Primers , Disease Models, Animal , Female , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction , Turner Syndrome/embryology
15.
Gene ; 555(2): 430-7, 2015 Jan 25.
Article in English | MEDLINE | ID: mdl-25447897

ABSTRACT

TNFα-induced protein 3-interacting protein 1 (TNIP1) represses signaling pathways initiated by specific nuclear and transmembrane receptors. This effect results in reduced activity of distinct transcription factors such as retinoic acid receptors (RAR), peroxisome-proliferator-activated receptors (PPAR), and NFκB. TNIP1-null and TNIP1-knockin defective for ubiquitin-binding mice show increased liver apoptosis, and enlarged spleen and lymph nodes, respectively. To complement current knowledge of TNIP1's broad physiologic functions as interpreted from in vivo studies and specific expression consequences from transcription factor repression, we determined effects of excess TNIP1 on global gene regulation. Following experimentally increased expression of TNIP1 in cultured keratinocytes, our gene expression microarray analysis not only confirmed TNIP1's association in previously known pathways and functions but also found a novel TNIP1-regulated pathway - the cell stress response. Under standard culture conditions, expression of several heat shock proteins, including HSPA1A, HSPA6, DNAJA1 and DNAJB1, was reduced. In heat-stressed conditions, differential regulation of HSPA1A and HSPA6 was observed, where only HSPA6 expression was reduced after heat-shock. Using HSPA6 as a model to elucidate the mechanism of the TNIP1-mediated HSP repression, we determined that TNIP1 likely represses HSPs through factors other than RAR, PPAR or NFκB despite the presence of these factors' binding sites in the HSPA6 promoter. These results indicate that regulation of HSPs may be through a yet unknown TNIP1-associated pathway. Additionally, these results suggest that TNIP1's reduction of HSP expression levels could negatively impact HSP chaperone capacity or their participation in the cell stress response.


Subject(s)
DNA-Binding Proteins/metabolism , Gene Expression Regulation , HSP70 Heat-Shock Proteins/metabolism , Promoter Regions, Genetic , Binding Sites , Cell Line , Cell Line, Tumor , Cluster Analysis , Hot Temperature , Humans , Keratinocytes/cytology , Oligonucleotide Array Sequence Analysis , Transcription Factors/metabolism , Transcriptional Activation
16.
J Clin Transl Hepatol ; 2(3): 143-52, 2014 Sep.
Article in English | MEDLINE | ID: mdl-26355943

ABSTRACT

The host-dependent nature of idiosyncratic drug-induced liver injury (iDILI) suggests that rare genetic polymorphisms may contribute to the disease. Indeed, a few mutations in key genes have already been identified using conventional human genetics approaches. Over 50 commonly used drugs can precipitate iDILI, making this a substantial medical problem. Only recently have human induced pluripotent stem cells been used as a research tool to discover novel iDILI genes and to study the mechanisms of iDILI in vitro. Here we review the current state of stem cell use in the investigation of iDILI, with a special focus on genetics. In addition, the concerns and difficulties associated with genetics and animal model research are discussed. We then present the features of patient-specific pluripotent stem cells (which may be derived from iDILI patients themselves), and explain why these cells may be of great utility. A variety of recent approaches to produce hepatocyte-like cells from pluripotent cells and the associated advantages and limitations of such cells are discussed. Future directions for the use of stem cell science to investigate iDILI include novel ways to identify new iDILI genes, a consideration of epigenetic impacts on iDILI, and the development of new and improved strategies for the production of hepatocytes from human pluripotent cells.

17.
Biol Psychiatry ; 75(12): 961-9, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-23958183

ABSTRACT

Less than 1.5% of the human genome encodes protein. However, vast portions of the human genome are subject to transcriptional and epigenetic regulation, and many noncoding regulatory DNA elements are thought to regulate the spatial organization of interphase chromosomes. For example, chromosomal "loopings" are pivotal for the orderly process of gene expression, by enabling distal regulatory enhancer or silencer elements to directly interact with proximal promoter and transcription start sites, potentially bypassing hundreds of kilobases of interspersed sequence on the linear genome. To date, however, epigenetic studies in the human brain are mostly limited to the exploration of DNA methylation and posttranslational modifications of the nucleosome core histones. In contrast, very little is known about the regulation of supranucleosomal structures. Here, we show that chromosome conformation capture, a widely used approach to study higher-order chromatin, is applicable to tissue collected postmortem, thereby informing about genome organization in the human brain. We introduce chromosome conformation capture protocols for brain and compare higher-order chromatin structures at the chromosome 6p22.2-22.1 schizophrenia and bipolar disorder susceptibility locus, and additional neurodevelopmental risk genes, (DPP10, MCPH1) in adult prefrontal cortex and various cell culture systems, including neurons derived from reprogrammed skin cells. We predict that the exploration of three-dimensional genome architectures and function will open up new frontiers in human brain research and psychiatric genetics and provide novel insights into the epigenetic risk architectures of regulatory noncoding DNA.


Subject(s)
Bipolar Disorder/genetics , Brain/metabolism , Chromosome Positioning , Genome, Human/genetics , Schizophrenia/genetics , Bipolar Disorder/pathology , Brain/pathology , Cell Culture Techniques , Chromatin/genetics , Chromatin/metabolism , Chromosomes, Human, Pair 6/genetics , Chromosomes, Human, Pair 6/metabolism , Epigenesis, Genetic , Gene Expression Regulation , Humans , Models, Neurological , Schizophrenia/pathology
18.
Cell Reprogram ; 15(2): 151-8, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23550731

ABSTRACT

Multiple methods exist that can reprogram differentiated cells to a pluripotent state similar to that of embryonic stem cells (ESCs). These include somatic cell nuclear transfer (SCNT), fusion-mediated reprogramming (FMR) of somatic cells with ESCs, and the production of induced pluripotent stem cells (iPSCs). All of these methods yield cells in which the endogenous Oct4 gene is reactivated. We were interested in comparing the activity of the Oct4 promoter in three different classes of pluripotent cells, including normal ESCs, FMR cells (FMRCs), and iPSCs. We prepared cells of all three types that harbor a transgene composed of the mouse Oct4 promoter driving green fluorescent protein (Oct4-GFP). All cell derivations started with a characterized transgenic Oct4-GFP mouse, and from this we derived ESCs, FMRCs, and iPSCs with the Oct4-GFP transgene present in an identical genomic integration site in all three cell types. Using flow cytometry we assessed Oct4 promoter expression, cell cycle behavior, and differentiation kinetics. We found similar levels of GFP expression in all three cell types and no significant alterations in pluripotency or differentiation. Our results suggest that the pluripotent condition is a potent "local attractor" state, because it can be achieved through three vastly different avenues.


Subject(s)
Cell Transdifferentiation , Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/biosynthesis , Pluripotent Stem Cells/metabolism , Promoter Regions, Genetic , Animals , Cell Line , Embryonic Stem Cells/cytology , Mice , Mice, Transgenic , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/cytology , Transgenes
19.
PLoS One ; 8(7): e67296, 2013.
Article in English | MEDLINE | ID: mdl-23874411

ABSTRACT

Hepatocytes play a central and crucial role in cholesterol and lipid homeostasis, and their proper function is of key importance for cardiovascular health. In particular, hepatocytes (especially periportal hepatocytes) endogenously synthesize large amounts of cholesterol and secrete it into circulating blood via apolipoprotein particles. Cholesterol-secreting hepatocytes are also the clinically-relevant cells targeted by statin treatment in vivo. The study of cholesterol homeostasis is largely restricted to the use of animal models and immortalized cell lines that do not recapitulate those key aspects of normal human hepatocyte function that result from genetic variation of individuals within a population. Hepatocyte-like cells (HLCs) derived from human embryonic and induced pluripotent stem cells can provide a cell culture model for the study of cholesterol homeostasis, dyslipidemias, the action of statins and other pharmaceuticals important for cardiovascular health. We have analyzed expression of core components for cholesterol homeostasis in untreated human iPS cells and in response to pravastatin. Here we show the production of differentiated cells resembling periportal hepatocytes from human pluripotent stem cells. These cells express a broad range of apolipoproteins required for secretion and elimination of serum cholesterol, actively secrete cholesterol into the medium, and respond functionally to statin treatment by reduced cholesterol secretion. Our research shows that HLCs derived from human pluripotent cells provide a robust cell culture system for the investigation of the hepatic contribution to human cholesterol homeostasis at both cellular and molecular levels. Importantly, it permits for the first time to also functionally assess the impact of genetic polymorphisms on cholesterol homeostasis. Finally, the system will also be useful for mechanistic studies of heritable dyslipidemias, drug discovery, and investigation of modes of action of cholesterol-modulatory drugs.


Subject(s)
Cardiovascular Physiological Phenomena , Cholesterol/metabolism , Embryonic Stem Cells/physiology , Hepatocytes/physiology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Induced Pluripotent Stem Cells/physiology , Adult , Apolipoproteins/metabolism , Cardiovascular System/metabolism , Cardiovascular System/physiopathology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cells, Cultured , Child, Preschool , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/physiology , Hepatocytes/drug effects , Hepatocytes/metabolism , Homeostasis/drug effects , Homeostasis/physiology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Liver/drug effects , Liver/metabolism , Liver/physiology
20.
Free Radic Biol Med ; 65: 584-594, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23911619

ABSTRACT

Isoniazid (INH) is an antituberculosis drug that has been associated with idiosyncratic liver injury in susceptible patients. The underlying mechanisms are still unclear, but there is growing evidence that INH and/or its major metabolite, hydrazine, may interfere with mitochondrial function. However, hepatic mitochondria have a large reserve capacity, and minor disruption of energy homeostasis does not necessarily induce cell death. We explored whether pharmacologic or genetic impairment of mitochondrial complex I may amplify mitochondrial dysfunction and precipitate INH-induced hepatocellular injury. We found that INH (≤ 3000 µM) did not induce cell injury in cultured mouse hepatocytes, although it decreased hepatocellular respiration and ATP levels in a concentration-dependent fashion. However, coexposure of hepatocytes to INH and nontoxic concentrations of the complex I inhibitors rotenone (3 µM) or piericidin A (30 nM) resulted in massive ATP depletion and cell death. Although both rotenone and piericidin A increased MitoSox-reactive fluorescence, Mito-TEMPO or N-acetylcysteine did not attenuate the extent of cytotoxicity. However, preincubation of cells with the acylamidase inhibitor bis-p-nitrophenol phosphate provided protection from hepatocyte injury induced by rotenone/INH (but not rotenone/hydrazine), suggesting that hydrazine was the cell-damaging species. Indeed, we found that hydrazine directly inhibited the activity of solubilized complex II. Hepatocytes isolated from mutant Ndufs4(+/-) mice, although featuring moderately lower protein expression levels of this complex I subunit in liver mitochondria, exhibited unchanged hepatic complex I activity and were therefore not sensitized to INH. These data indicate that underlying inhibition of complex I, which alone is not acutely toxic, can trigger INH-induced hepatocellular injury.


Subject(s)
Antitubercular Agents/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Electron Transport Complex I/drug effects , Hepatocytes/drug effects , Isoniazid/toxicity , Mitochondria/drug effects , Animals , Antitubercular Agents/metabolism , Blotting, Western , Cell Respiration/drug effects , Hydrazines/metabolism , Isoniazid/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL