Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
NAR Genom Bioinform ; 6(2): lqae070, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38881577

ABSTRACT

Ribosomal DNA (rDNA) repeat units are organized into tandem clusters in eukaryotic cells. In mice, these clusters are located on at least eight chromosomes and show extensive variation in the number of repeats between mouse genomes. To analyze intra- and inter-genomic variation of mouse rDNA repeats, we selectively isolated 25 individual rDNA units using Transformation-Associated Recombination (TAR) cloning. Long-read sequencing and subsequent comparative sequence analysis revealed that each full-length unit comprises an intergenic spacer (IGS) and a ∼13.4 kb long transcribed region encoding the three rRNAs, but with substantial variability in rDNA unit size, ranging from ∼35 to ∼46 kb. Within the transcribed regions of rDNA units, we found 209 variants, 70 of which are in external transcribed spacers (ETSs); but the rDNA size differences are driven primarily by IGS size heterogeneity, due to indels containing repetitive elements and some functional signals such as enhancers. Further evolutionary analysis categorized rDNA units into distinct clusters with characteristic IGS lengths; numbers of enhancers; and presence/absence of two common SNPs in promoter regions, one of which is located within promoter (p)RNA and may influence pRNA folding stability. These characteristic features of IGSs also correlated significantly with 5'ETS variant patterns described previously and associated with differential expression of rDNA units. Our results suggest that variant rDNA units are differentially regulated and open a route to investigate the role of rDNA variation on nucleolar formation and possible associations with pathology.

2.
Oncotarget ; 12(15): 1444-1456, 2021 Jul 20.
Article in English | MEDLINE | ID: mdl-34316326

ABSTRACT

Telomerase/telomere-targeting therapy is a potentially promising approach for cancer treatment because even transient telomere dysfunction can induce chromosomal instability (CIN) and may be a barrier to tumor growth. We recently developed a dual-HAC (Human Artificial Chromosome) assay that enables identification and ranking of compounds that induce CIN as a result of telomere dysfunction. This assay is based on the use of two isogenic HT1080 cell lines, one carrying a linear HAC (containing telomeres) and the other carrying a circular HAC (lacking telomeres). Disruption of telomeres in response to drug treatment results in specific destabilization of the linear HAC. Results: In this study, we used the dual-HAC assay for the analysis of the platinum-derived G4 ligand Pt-tpy and five of its derivatives: Pt-cpym, Pt-vpym, Pt-ttpy, Pt(PA)-tpy, and Pt-BisQ. Our analysis revealed four compounds, Pt-tpy, Pt-ttpy, Pt-vpym and Pt-cpym, that induce a specific loss of a linear but not a circular HAC. Increased CIN after treatment by these compounds correlates with the induction of double-stranded breaks (DSBs) predominantly localized at telomeres and reflecting telomere-associated DNA damage. Analysis of the mitotic phenotypes induced by these drugs revealed an elevated rate of chromatin bridges (CBs) in late mitosis and cytokinesis. These terpyridine platinum-derived G4 ligands are promising compounds for cancer treatment.

3.
Sci Rep ; 11(1): 2997, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33542373

ABSTRACT

The rDNA clusters and flanking sequences on human chromosomes 13, 14, 15, 21 and 22 represent large gaps in the current genomic assembly. The organization and the degree of divergence of the human rDNA units within an individual nucleolar organizer region (NOR) are only partially known. To address this lacuna, we previously applied transformation-associated recombination (TAR) cloning to isolate individual rDNA units from chromosome 21. That approach revealed an unexpectedly high level of heterogeneity in human rDNA, raising the possibility of corresponding variations in ribosome dynamics. We have now applied the same strategy to analyze an entire rDNA array end-to-end from a copy of chromosome 22. Sequencing of TAR isolates provided the entire NOR sequence, including proximal and distal junctions that may be involved in nucleolar function. Comparison of the newly sequenced rDNAs to reference sequence for chromosomes 22 and 21 revealed variants that are shared in human rDNA in individuals from different ethnic groups, many of them at high frequency. Analysis infers comparable intra- and inter-individual divergence of rDNA units on the same and different chromosomes, supporting the concerted evolution of rDNA units. The results provide a route to investigate further the role of rDNA variation in nucleolar formation and in the empirical associations of nucleoli with pathology.


Subject(s)
Chromosomes, Human, Pair 22/genetics , DNA, Ribosomal/genetics , Genome, Human/genetics , Nucleolus Organizer Region/genetics , Cell Nucleolus/genetics , Cloning, Molecular , Genetic Heterogeneity , Genomics , Humans , Molecular Sequence Annotation , Ribosomes/genetics
4.
Cancer Res ; 78(21): 6282-6296, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30166419

ABSTRACT

The targeting of telomerase and telomere maintenance mechanisms represents a promising therapeutic approach for various types of cancer. In this work, we designed a new protocol to screen for and rank the efficacy of compounds specifically targeting telomeres and telomerase. This approach used two isogenic cell lines containing a circular human artificial chromosome (HAC, lacking telomeres) and a linear HAC (containing telomeres) marked with the EGFP transgene; compounds that target telomerase or telomeres should preferentially induce loss of the linear HAC but not the circular HAC. Our assay allowed quantification of chromosome loss by routine flow cytometry. We applied this dual-HAC assay to rank a set of known and newly developed compounds, including G-quadruplex (G4) ligands. Among the latter group, two compounds, Cu-ttpy and Pt-ttpy, induced a high rate of linear HAC loss with no significant effect on the mitotic stability of a circular HAC. Analysis of the mitotic phenotypes induced by these drugs revealed an elevated rate of chromatin bridges in late mitosis and cytokinesis as well as UFB (ultrafine bridges). Chromosome loss after Pt-ttpy or Cu-ttpy treatment correlated with the induction of telomere-associated DNA damage. Overall, this platform enables identification and ranking of compounds that greatly increase chromosome mis-segregation rates as a result of telomere dysfunction and may expedite the development of new therapeutic strategies for cancer treatment.Significance: An assay provides a unique opportunity to screen thousands of chemical compounds for their ability to inactivate replication of telomeric ends in cancer cells and holds potential to lay the foundation for the discovery of new treatments for cancer. Cancer Res; 78(21); 6282-96. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Telomerase/antagonists & inhibitors , Telomere/drug effects , Cell Line , Cell Line, Tumor , Cell Survival , Chromatin , Chromosomes , Chromosomes, Artificial, Human , DNA Damage , Drug Design , HCT116 Cells , Humans , Hydroxamic Acids/pharmacology , Mitosis , Neoplasms/genetics , Transgenes
5.
Nucleic Acids Res ; 46(13): 6712-6725, 2018 07 27.
Article in English | MEDLINE | ID: mdl-29788454

ABSTRACT

Despite the key role of the human ribosome in protein biosynthesis, little is known about the extent of sequence variation in ribosomal DNA (rDNA) or its pre-rRNA and rRNA products. We recovered ribosomal DNA segments from a single human chromosome 21 using transformation-associated recombination (TAR) cloning in yeast. Accurate long-read sequencing of 13 isolates covering ∼0.82 Mb of the chromosome 21 rDNA complement revealed substantial variation among tandem repeat rDNA copies, several palindromic structures and potential errors in the previous reference sequence. These clones revealed 101 variant positions in the 45S transcription unit and 235 in the intergenic spacer sequence. Approximately 60% of the 45S variants were confirmed in independent whole-genome or RNA-seq data, with 47 of these further observed in mature 18S/28S rRNA sequences. TAR cloning and long-read sequencing enabled the accurate reconstruction of multiple rDNA units and a new, high-quality 44 838 bp rDNA reference sequence, which we have annotated with variants detected from chromosome 21 of a single individual. The large number of variants observed reveal heterogeneity in human rDNA, opening up the possibility of corresponding variations in ribosome dynamics.


Subject(s)
Chromosomes, Human, Pair 21 , DNA, Ribosomal/chemistry , Genes, rRNA , Genetic Variation , Animals , Cell Line , Cloning, Molecular , DNA, Ribosomal/isolation & purification , DNA, Ribosomal Spacer/chemistry , Humans , Mice , Nucleic Acid Conformation , Nucleolus Organizer Region/chemistry , RNA, Ribosomal/chemistry , RNA, Ribosomal/metabolism , Sequence Analysis, DNA
6.
ACS Synth Biol ; 7(1): 63-74, 2018 01 19.
Article in English | MEDLINE | ID: mdl-28799737

ABSTRACT

The production of cells capable of carrying multiple transgenes to Mb-size genomic loci has multiple applications in biomedicine and biotechnology. In order to achieve this goal, three key steps are required: (i) cloning of large genomic segments; (ii) insertion of multiple DNA blocks at a precise location and (iii) the capability to eliminate the assembled region from cells. In this study, we designed the iterative integration system (IIS) that utilizes recombinases Cre, ΦC31 and ΦBT1, and combined it with a human artificial chromosome (HAC) possessing a regulated kinetochore (alphoidtetO-HAC). We have demonstrated that the IIS-alphoidtetO-HAC system is a valuable genetic tool by reassembling a functional gene from multiple segments on the HAC. IIS-alphoidtetO-HAC has several notable advantages over other artificial chromosome-based systems. This includes the potential to assemble an unlimited number of genomic DNA segments; a DNA assembly process that leaves only a small insertion (<60 bp) scar between adjacent DNA, allowing genes reassembled from segments to be spliced correctly; a marker exchange system that also changes cell color, and counter-selection markers at each DNA insertion step, simplifying selection of correct clones; and presence of an error proofing mechanism to remove cells with misincorporated DNA segments, which improves the integrity of assembly. In addition, the IIS-alphoidtetO-HAC carrying a locus of interest is removable, offering the unique possibility to revert the cell line to its pretransformed state and compare the phenotypes of human cells with and without a functional copy of a gene(s). Thus, IIS-alphoidtetO-HAC allows investigation of complex biomedical pathways, gene(s) regulation, and has the potential to engineer synthetic chromosomes with a predetermined set of genes.


Subject(s)
Chromosomes, Artificial, Human/genetics , DNA/metabolism , Integrases/genetics , Kinetochores/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , DNA/genetics , Humans , In Situ Hybridization, Fluorescence , Integrases/metabolism , Plasmids/genetics , Plasmids/metabolism , Recombination, Genetic , Von Hippel-Lindau Tumor Suppressor Protein/genetics
7.
Oncotarget ; 7(12): 14841-56, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26943579

ABSTRACT

Accumulating data indicates that chromosome instability (CIN) common to cancer cells can be used as a target for cancer therapy. At present the rate of chromosome mis-segregation is quantified by laborious techniques such as coupling clonal cell analysis with karyotyping or fluorescence in situ hybridization (FISH). Recently, a novel assay was developed based on the loss of a non-essential human artificial chromosome (HAC) carrying a constitutively expressed EGFP transgene ("loss of signal" assay). Using this system, anticancer drugs can be easily ranked on by their effect on HAC loss. However, it is problematic to covert this "loss of signal" assay into a high-throughput screen to identify drugs and mutations that increase CIN levels. To address this point, we re-designed the HAC-based assay. In this new system, the HAC carries a constitutively expressed shRNA against the EGFP transgene integrated into human genome. Thus, cells that inherit the HAC display no green fluorescence, while cells lacking the HAC do. We verified the accuracy of this "gain of signal" assay by measuring the level of CIN induced by known antimitotic drugs and added to the list of previously ranked CIN inducing compounds, two newly characterized inhibitors of the centromere-associated protein CENP-E, PF-2771 and GSK923295 that exhibit the highest effect on chromosome instability measured to date. The "gain of signal" assay was also sensitive enough to detect increase of CIN after siRNA depletion of known genes controlling mitotic progression through distinct mechanisms. Hence this assay can be utilized in future experiments to uncover novel human CIN genes, which will provide novel insight into the pathogenesis of cancer. Also described is the possible conversion of this new assay into a high-throughput screen using a fluorescence microplate reader to characterize chemical libraries and identify new conditions that modulate CIN level.


Subject(s)
Biological Assay/methods , Chromosomal Instability , Chromosomes, Artificial, Human/genetics , Fibrosarcoma/diagnosis , Fibrosarcoma/genetics , Green Fluorescent Proteins/metabolism , Apoptosis , Cell Proliferation , Green Fluorescent Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Tumor Cells, Cultured
8.
Cancer Res ; 76(4): 902-11, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26837770

ABSTRACT

Whole chromosomal instability (CIN), manifested as unequal chromosome distribution during cell division, is a distinguishing feature of most cancer types. CIN is generally considered to drive tumorigenesis, but a threshold level exists whereby further increases in CIN frequency in fact hinder tumor growth. While this attribute is appealing for therapeutic exploitation, drugs that increase CIN beyond this therapeutic threshold are currently limited. In our previous work, we developed a quantitative assay for measuring CIN based on the use of a nonessential human artificial chromosome (HAC) carrying a constitutively expressed EGFP transgene. Here, we used this assay to rank 62 different anticancer drugs with respect to their effects on chromosome transmission fidelity. Drugs with various mechanisms of action, such as antimicrotubule activity, histone deacetylase inhibition, mitotic checkpoint inhibition, and targeting of DNA replication and damage responses, were included in the analysis. Ranking of the drugs based on their ability to induce HAC loss revealed that paclitaxel, gemcitabine, dactylolide, LMP400, talazoparib, olaparib, peloruside A, GW843682, VX-680, and cisplatin were the top 10 drugs demonstrating HAC loss at a high frequency. Therefore, identification of currently used compounds that greatly increase chromosome mis-segregation rates should expedite the development of new therapeutic strategies to target and leverage the CIN phenotype in cancer cells.


Subject(s)
Cell Transformation, Neoplastic/genetics , Chromosomal Instability/genetics , Cell Line, Tumor , Genes, Tumor Suppressor , Humans , Transgenes
9.
Cell Mol Life Sci ; 70(19): 3723-37, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23677492

ABSTRACT

Human artificial chromosomes (HACs) are vectors that offer advantages of capacity and stability for gene delivery and expression. Several studies have even demonstrated their use for gene complementation in gene-deficient recipient cell lines and animal transgenesis. Recently, we constructed an advance HAC-based vector, alphoid(tetO)-HAC, with a conditional centromere. In this HAC, a gene-loading site was inserted into a centrochromatin domain critical for kinetochore assembly and maintenance. While by definition this domain is permissive for transcription, there have been no long-term studies on transgene expression within centrochromatin. In this study, we compared the effects of three chromatin insulators, cHS4, gamma-satellite DNA, and tDNA, on the expression of an EGFP transgene inserted into the alphoid(tetO)-HAC vector. Insulator function was essential for stable expression of the transgene in centrochromatin. In two analyzed host cell lines, a tDNA insulator composed of two functional copies of tRNA genes showed the highest barrier activity. We infer that proximity to centrochromatin does not protect genes lacking chromatin insulators from epigenetic silencing. Barrier elements that prevent gene silencing in centrochromatin would thus help to optimize transgenesis using HAC vectors.


Subject(s)
Chromatin/genetics , Chromosomes, Artificial, Human , Genetic Vectors/genetics , Transgenes , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , DNA, Satellite/genetics , Gene Expression , Gene Silencing , Green Fluorescent Proteins/genetics , HeLa Cells , Humans , RNA, Transfer/genetics
10.
BMC Cancer ; 13: 252, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23694679

ABSTRACT

BACKGROUND: Aneuploidy is a feature of most cancer cells that is often accompanied by an elevated rate of chromosome mis-segregation termed chromosome instability (CIN). While CIN can act as a driver of cancer genome evolution and tumor progression, recent findings point to the existence of a threshold level beyond which CIN becomes a barrier to tumor growth and therefore can be exploited therapeutically. Drugs known to increase CIN beyond the therapeutic threshold are currently few in number, and the clinical promise of targeting the CIN phenotype warrants new screening efforts. However, none of the existing methods, including the in vitro micronuclei (MNi) assay, developed to quantify CIN, is entirely satisfactory. METHODS: We have developed a new assay for measuring CIN. This quantitative assay for chromosome mis-segregation is based on the use of a non-essential human artificial chromosome (HAC) carrying a constitutively expressed EGFP transgene. Thus, cells that inherit the HAC display green fluorescence, while cells lacking the HAC do not. This allows the measurement of HAC loss rate by routine flow cytometry. RESULTS: Using the HAC-based chromosome loss assay, we have analyzed several well-known anti-mitotic, spindle-targeting compounds, all of which have been reported to induce micronuclei formation and chromosome loss. For each drug, the rate of HAC loss was accurately measured by flow cytometry as a proportion of non-fluorescent cells in the cell population which was verified by FISH analysis. Based on our estimates, despite their similar cytotoxicity, the analyzed drugs affect the rates of HAC mis-segregation during mitotic divisions differently. The highest rate of HAC mis-segregation was observed for the microtubule-stabilizing drugs, taxol and peloruside A. CONCLUSION: Thus, this new and simple assay allows for a quick and efficient screen of hundreds of drugs to identify those affecting chromosome mis-segregation. It also allows ranking of compounds with the same or similar mechanism of action based on their effect on the rate of chromosome loss. The identification of new compounds that increase chromosome mis-segregation rates should expedite the development of new therapeutic strategies to target the CIN phenotype in cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Chromosomal Instability/drug effects , Chromosomes, Artificial, Human/genetics , Genetic Techniques , Green Fluorescent Proteins/genetics , Cell Line, Tumor , Flow Cytometry , Humans , In Situ Hybridization, Fluorescence , Transgenes
11.
J Biol Chem ; 287(44): 37171-84, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-22923615

ABSTRACT

Olfm1, a secreted highly conserved glycoprotein, is detected in peripheral and central nervous tissues and participates in neural progenitor maintenance, cell death in brain, and optic nerve arborization. In this study, we identified Olfm1 as a molecule promoting axon growth through interaction with the Nogo A receptor (NgR1) complex. Olfm1 is coexpressed with NgR1 in dorsal root ganglia and retinal ganglion cells in embryonic and postnatal mice. Olfm1 specifically binds to NgR1, as judged by alkaline phosphatase assay and coimmunoprecipitation. The addition of Olfm1 inhibited the growth cone collapse of dorsal root ganglia neurons induced by myelin-associated inhibitors, indicating that Olfm1 attenuates the NgR1 receptor functions. Olfm1 caused the inhibition of NgR1 signaling by interfering with interaction between NgR1 and its coreceptors p75NTR or LINGO-1. In zebrafish, inhibition of optic nerve extension by olfm1 morpholino oligonucleotides was partially rescued by dominant negative ngr1 or lingo-1. These data introduce Olfm1 as a novel NgR1 ligand that may modulate the functions of the NgR1 complex in axonal growth.


Subject(s)
Axons/physiology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Extracellular Matrix Proteins/physiology , Glycoproteins/physiology , Nerve Tissue Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , COS Cells , Chlorocebus aethiops , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Green Fluorescent Proteins/biosynthesis , Growth Cones/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin Proteins/physiology , Nogo Proteins , Optic Nerve/cytology , Optic Nerve/embryology , Organ Specificity , PC12 Cells , Protein Binding , Rats , Receptor, Nerve Growth Factor/metabolism , Zebrafish , rhoA GTP-Binding Protein/metabolism
12.
Genes Chromosomes Cancer ; 51(10): 933-48, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22733720

ABSTRACT

Several linkage studies provided evidence for the presence of the hereditary prostate cancer locus, HPCX1, at Xq27-q28. The strongest linkage peak of prostate cancer overlies a variable region of ~750 kb at Xq27 enriched by segmental duplications (SDs), suggesting that the predisposition to prostate cancer may be a genomic disorder caused by recombinational interaction between SDs. The large size of SDs and their sequence similarity make it difficult to examine this region for possible rearrangements using standard methods. To overcome this problem, direct isolation of a set of genomic segments by in vivo recombination in yeast (a TAR cloning technique) was used to perform a mutational analysis of the 750 kb region in X-linked families. We did not detect disease-specific rearrangements within this region. In addition, transcriptome and computational analyses were performed to search for nonannotated genes within the Xq27 region, which may be associated with genetic predisposition to prostate cancer. Two candidate genes were identified, one of which is a novel gene termed SPANXL that represents a highly diverged member of the SPANX gene family, and the previously described CDR1 gene that is expressed at a high level in both normal and malignant prostate cells, and mapped 210 kb of upstream the SPANX gene cluster. No disease-specific alterations were identified in these genes. Our results exclude the 750-kb genetically unstable region at Xq27 as a candidate locus for prostate malignancy. Adjacent regions appear to be the most likely candidates to identify the elusive HPCX1 locus.


Subject(s)
Chromosomes, Human, X/genetics , DNA, Neoplasm/genetics , Genetic Loci , Prostatic Neoplasms/genetics , Autoantigens/genetics , Chromosome Mapping , Chromosomes, Human, X/chemistry , DNA Mutational Analysis , Family , Female , Genetic Linkage , Genetic Predisposition to Disease , Humans , Male , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/diagnosis , Recombination, Genetic , Saccharomyces cerevisiae/genetics , Segmental Duplications, Genomic
13.
ACS Synth Biol ; 1(12): 590-601, 2012 Dec 21.
Article in English | MEDLINE | ID: mdl-23411994

ABSTRACT

Human artificial chromosomes (HACs) represent a novel promising episomal system for functional genomics, gene therapy, and synthetic biology. HACs are engineered from natural and synthetic alphoid DNA arrays upon transfection into human cells. The use of HACs for gene expression studies requires the knowledge of their structural organization. However, none of the de novo HACs constructed so far has been physically mapped in detail. Recently we constructed a synthetic alphoid(tetO)-HAC that was successfully used for expression of full-length genes to correct genetic deficiencies in human cells. The HAC can be easily eliminated from cell populations by inactivation of its conditional kinetochore. This unique feature provides a control for phenotypic changes attributed to expression of HAC-encoded genes. This work describes organization of a megabase-size synthetic alphoid DNA array in the alphoid(tetO)-HAC that has been formed from a ~50 kb synthetic alphoid(tetO)-construct. Our analysis showed that this array represents a 1.1 Mb continuous sequence assembled from multiple copies of input DNA, a significant part of which was rearranged before assembling. The tandem and inverted alphoid DNA repeats in the HAC range in size from 25 to 150 kb. In addition, we demonstrated that the structure and functional domains of the HAC remains unchanged after several rounds of its transfer into different host cells. The knowledge of the alphoid(tetO)-HAC structure provides a tool to control HAC integrity during different manipulations. Our results also shed light on a mechanism for de novo HAC formation in human cells.


Subject(s)
Centromere/genetics , Chromosomes, Artificial, Human , DNA/genetics , Animals , CHO Cells , Cell Line, Tumor , Cricetulus , Gene Expression , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Kinetochores/metabolism , Oligonucleotide Array Sequence Analysis/methods , Tandem Repeat Sequences
14.
DNA Res ; 17(5): 293-301, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20798231

ABSTRACT

Human artificial chromosomes (HACs), which carry a fully functional centromere and are maintained as a single-copy episome, are not associated with random mutagenesis and offer greater control over expression of ectopic genes on the HAC. Recently, we generated a HAC with a conditional centromere, which includes the tetracycline operator (tet-O) sequence embedded in the alphoid DNA array. This conditional centromere can be inactivated, loss of the alphoid(tet-O) (tet-O HAC) by expression of tet-repressor fusion proteins. In this report, we describe adaptation of the tet-O HAC vector for gene delivery and gene expression in human cells. A loxP cassette was inserted into the tet-O HAC by homologous recombination in chicken DT40 cells following a microcell-mediated chromosome transfer (MMCT). The tet-O HAC with the loxP cassette was then transferred into Chinese hamster ovary cells, and EGFP transgene was efficiently and accurately incorporated into the tet-O HAC vector. The EGFP transgene was stably expressed in human cells after transfer via MMCT. Because the transgenes inserted on the tet-O HAC can be eliminated from cells by HAC loss due to centromere inactivation, this HAC vector system provides important novel features and has potential applications for gene expression studies and gene therapy.


Subject(s)
Centromere/genetics , Chromosomes, Artificial, Human/genetics , Gene Expression , Gene Transfer Techniques , Genetic Vectors , Transgenes , Animals , CHO Cells , Cell Line , Chickens , Cricetinae , Cricetulus , Gene Targeting/methods , Genetic Therapy , Green Fluorescent Proteins/genetics , Humans , In Situ Hybridization, Fluorescence , Operator Regions, Genetic , Plasmids , Polymerase Chain Reaction , Recombination, Genetic , Tetracyclines
15.
Mol Cell Biol ; 29(8): 2139-54, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19188438

ABSTRACT

It is well documented that mutations in the MYOCILIN gene may lead to juvenile- and adult-onset primary open-angle glaucoma. However, the functions of wild-type myocilin are still not well understood. To study the functions of human myocilin and its two proteolytic fragments, these proteins were expressed in HEK293 cells. Conditioned medium from myocilin-expressing cells, as well as purified myocilin, induced the formation of stress fibers in primary cultures of human trabecular meshwork or NIH 3T3 cells. Stress fiber-inducing activity of myocilin was blocked by antibodies against myocilin, as well as secreted inhibitors of Wnt signaling, secreted Frizzled-related protein 1 (sFRP1) or sFRP3, and beta-catenin small interfering RNA. Interaction of myocilin with sFRP1, sFRP3, and several Frizzled receptors was confirmed by immunoprecipitation experiments and by binding of myocilin to the surface of cells expressing cysteine-rich domains of different Frizzled and sFRPs. Treatment of NIH 3T3 cells with myocilin and its fragments induced intracellular redistribution of beta-catenin and its accumulation on the cellular membrane but did not induce nuclear accumulation of beta-catenin. Overexpression of myocilin in the eye angle tissues of transgenic mice stimulated accumulation of beta-catenin in these tissues. Myocilin and Wnt proteins may perform redundant functions in the mammalian eye, since myocilin modulates Wnt signaling by interacting with components of this signaling pathway.


Subject(s)
Cytoskeletal Proteins/physiology , Eye Proteins/physiology , Glycoproteins/physiology , Wnt Proteins/metabolism , Animals , Cell Line , Cells, Cultured , Cytoskeletal Proteins/metabolism , Eye Proteins/metabolism , Frizzled Receptors/metabolism , Glycoproteins/metabolism , Humans , Mice , Mice, Transgenic , beta Catenin/metabolism
16.
J Neurosci ; 28(31): 7900-10, 2008 Jul 30.
Article in English | MEDLINE | ID: mdl-18667622

ABSTRACT

Olfactomedin 1 (Olfm1) is a secreted glycoprotein belonging to a family of olfactomedin domain-containing proteins. It is involved in the regulation of neural crest production in chicken and promotes neuronal differentiation in Xenopus. Here, we investigate the functions of Olfm1 in zebrafish eye development. Overexpression of full-length Olfm1, and especially its BMY form lacking the olfactomedin domain, increased the thickness of the optic nerve and produced a more extended projection field in the optic tectum compared with control embryos. In contrast, injection of olfm1-morpholino oligonucleotide (Olfm1-MO) reduced the eye size, inhibited optic nerve extension, and increased the number of apoptotic cells in the retinal ganglion cell and inner nuclear layers. Overexpression of full-length Olfm1 increased the lateral separation of the expression domains of eye-field markers, rx3 and six3. The Olfm1-MO had the opposite effect. These data suggest that zebrafish Olfm1 may play roles in the early eye determination, differentiation, optic nerve extension, and branching of the retinal ganglion cell axon terminals, with the N-terminal region of Olfm1 being critical for these effects. Injection of RNA encoding WIF-1, a secreted inhibitor of Wnt signaling, caused changes in the expression pattern of rx3 similar to those observed after Olfm1-MO injection. Simultaneous overexpression of WIF-1 and Olfm1 abolished the WIF-1 effect. Physical interaction of WIF-1 and Olfm1 was demonstrated by coimmunoprecipitation experiments. We concluded that Olfm1 serves as a modulator of Wnt signaling.


Subject(s)
Axons/physiology , Extracellular Matrix Proteins/physiology , Glycoproteins/physiology , Retina/growth & development , Signal Transduction/physiology , Wnt Proteins/physiology , Zebrafish Proteins/physiology , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/chemistry , Extracellular Matrix Proteins/genetics , Glycoproteins/biosynthesis , Glycoproteins/chemistry , Glycoproteins/genetics , Humans , Mice , Mice, Knockout , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Retina/embryology , Wnt Proteins/antagonists & inhibitors , Wnt Proteins/metabolism , Zebrafish
17.
Exp Cell Res ; 313(1): 98-108, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17054946

ABSTRACT

Optimedin, also known as olfactomedin 3, belongs to a family of olfactomedin domain-containing proteins. It is expressed in neural tissues and Pax6 is involved in the regulation of its promoter. To study possible effects of optimedin on the differentiation of neural cells, we produced stably transfected PC12 cell lines expressing optimedin under a tetracycline-inducible promoter. Cells expressing high levels of optimedin showed higher growth rates and stronger adhesion to the collagen extracellular matrix as compared with control PC12 cells. After stimulation with nerve growth factor (NGF), optimedin-expressing cells demonstrated elevated levels of N-cadherin, beta-catenin, alpha-catenin and occludin as compared with stimulated, control PC12 cells. Expression of optimedin induced Ca(2+)-dependent aggregation of NGF-stimulated PC12 cells and this aggregation was blocked by the expression of N-cadherin siRNA. Expression of optimedin also changed the organization of the actin cytoskeleton and inhibited neurite outgrowth in NGF-stimulated PC12 cells. We suggest that expression of optimedin stimulates the formation of adherent and tight junctions on the cell surface and this may play an important role in the differentiation of the brain and retina through the modulation of cytoskeleton organization, cell-cell adhesion and migration.


Subject(s)
Cadherins/metabolism , Extracellular Matrix Proteins/metabolism , Glycoproteins/metabolism , Nerve Growth Factor/pharmacology , Neurons/drug effects , Neurons/metabolism , Animals , Base Sequence , Cadherins/antagonists & inhibitors , Cadherins/genetics , Cell Aggregation/drug effects , DNA Primers/genetics , Extracellular Matrix Proteins/genetics , Gene Expression , Glycoproteins/genetics , Neurites/metabolism , Neurons/cytology , PC12 Cells , RNA, Small Interfering/genetics , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transfection , alpha Catenin/metabolism , beta Catenin/metabolism
18.
Invest Ophthalmol Vis Sci ; 47(1): 206-12, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16384964

ABSTRACT

PURPOSE: The present study compared properties of wild-type and mutated mouse and human myocilin (Myoc) proteins as a prerequisite for development of a mouse model of glaucoma. METHODS: cDNA encoding full-length mouse Myoc was cloned into the p3XFLAG-CMV-14 vector. Tyr423His and Ile463Ser mutations were introduced into the mouse Myoc protein by in vitro mutagenesis. Intracellular localization and secretion of wild-type and mutated mouse Myoc proteins were studied in immunostaining and Western blotting experiments, respectively, after transfection into COS-7 cells. RESULTS: Similar to human MYOC, wild-type and mutated mouse Myoc demonstrated vesicular staining in transfected cells. However, while wild-type human and mouse Myoc were preferentially located in both the endoplasmic reticulum and Golgi, mutated human and mouse Myoc were located mainly in the endoplasmic reticulum and were excluded from Golgi. Similar to mutations in human MYOC, mutations in mouse Myoc dramatically reduced its secretion from transfected cells. Secretion of mutated Myoc was partially restored by culturing cells at 30 degrees C instead of 37 degrees C. The presence of mutated human MYOC prevented secretion of wild-type mouse Myoc but did not dramatically affect secretion of alkaline phosphatase, thrombospondin, Timp3 or olfactomedin-1. CONCLUSIONS: Properties of the mouse Myoc protein are similar to those of the human MYOC. The presence of mutated mouse or human Myoc does not block a general secretory pathway. Expression of mutated Myoc in the eye in mice may mimic human glaucoma and lead to development of a genetic mouse model of glaucoma.


Subject(s)
Cytoskeletal Proteins/physiology , Eye Proteins/physiology , Genetic Vectors , Glycoproteins/physiology , Transfection , Amino Acid Sequence , Animals , Blotting, Western , COS Cells/metabolism , Chlorocebus aethiops , Endoplasmic Reticulum/metabolism , Extracellular Matrix Proteins/genetics , Glycoproteins/genetics , Golgi Apparatus/metabolism , Humans , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation/physiology , Plasmids
SELECTION OF CITATIONS
SEARCH DETAIL
...