Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 32
1.
Genome Biol ; 25(1): 135, 2024 May 23.
Article En | MEDLINE | ID: mdl-38783323

BACKGROUND: Dilated cardiomyopathy (DCM) is one of the most common causes of heart failure. Multiple identified mutations in nexilin (NEXN) have been suggested to be linked with severe DCM. However, the exact association between multiple mutations of Nexn and DCM remains unclear. Moreover, it is critical for the development of precise and effective therapeutics in treatments of DCM. RESULTS: In our study, Nexn global knockout mice and mice carrying human equivalent G645del mutation are studied using functional gene rescue assays. AAV-mediated gene delivery is conducted through systemic intravenous injections at the neonatal stage. Heart tissues are analyzed by immunoblots, and functions are assessed by echocardiography. Here, we identify functional components of Nexilin and demonstrate that exogenous introduction could rescue the cardiac function and extend the lifespan of Nexn knockout mouse models. Similar therapeutic effects are also obtained in G645del mice, providing a promising intervention for future clinical therapeutics. CONCLUSIONS: In summary, we demonstrated that a single injection of AAV-Nexn was capable to restore the functions of cardiomyocytes and extended the lifespan of Nexn knockout and G645del mice. Our study represented a long-term gene replacement therapy for DCM that potentially covers all forms of loss-of-function mutations in NEXN.


Cardiomyopathy, Dilated , Genetic Therapy , Mice, Knockout , Animals , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/therapy , Mice , Humans , Dependovirus/genetics , Myocytes, Cardiac/metabolism , Disease Models, Animal , Mutation , Genetic Vectors/administration & dosage , Gene Transfer Techniques
2.
Cell Rep Med ; 5(3): 101449, 2024 Mar 19.
Article En | MEDLINE | ID: mdl-38508141

Tissue regeneration following an injury requires dynamic cell-state transitions that allow for establishing the cell identities required for the restoration of tissue homeostasis and function. Here, we present a biochemical intervention that induces an intermediate cell state mirroring a transition identified during normal differentiation of myoblasts and other multipotent and pluripotent cells to mature cells. When applied in somatic differentiated cells, the intervention, composed of one-carbon metabolites, reduces some dedifferentiation markers without losing the lineage identity, thus inducing limited reprogramming into a more flexible cell state. Moreover, the intervention enabled accelerated repair after muscle injury in young and aged mice. Overall, our study uncovers a conserved biochemical transitional phase that enhances cellular plasticity in vivo and hints at potential and scalable biochemical interventions of use in regenerative medicine and rejuvenation interventions that may be more tractable than genetic ones.


Muscles , Myoblasts , Mice , Animals , Cell Differentiation , Myoblasts/metabolism
3.
Cell Rep ; 39(4): 110730, 2022 04 26.
Article En | MEDLINE | ID: mdl-35476977

Mammals have limited regenerative capacity, whereas some vertebrates, like fish and salamanders, are able to regenerate their organs efficiently. The regeneration in these species depends on cell dedifferentiation followed by proliferation. We generate a mouse model that enables the inducible expression of the four Yamanaka factors (Oct-3/4, Sox2, Klf4, and c-Myc, or 4F) specifically in hepatocytes. Transient in vivo 4F expression induces partial reprogramming of adult hepatocytes to a progenitor state and concomitantly increases cell proliferation. This is indicated by reduced expression of differentiated hepatic-lineage markers, an increase in markers of proliferation and chromatin modifiers, global changes in DNA accessibility, and an acquisition of liver stem and progenitor cell markers. Functionally, short-term expression of 4F enhances liver regenerative capacity through topoisomerase2-mediated partial reprogramming. Our results reveal that liver-specific 4F expression in vivo induces cellular plasticity and counteracts liver failure, suggesting that partial reprogramming may represent an avenue for enhancing tissue regeneration.


Cellular Reprogramming , Liver , Animals , Cell Dedifferentiation , Hepatocytes/metabolism , Liver/metabolism , Liver Regeneration , Mammals , Mice
4.
Nat Commun ; 12(1): 3330, 2021 06 07.
Article En | MEDLINE | ID: mdl-34099664

Human pluripotent stem cell (hPSC)-derived pancreatic ß cells are an attractive cell source for treating diabetes. However, current derivation methods remain inefficient, heterogeneous, and cell line dependent. To address these issues, we first devised a strategy to efficiently cluster hPSC-derived pancreatic progenitors into 3D structures. Through a systematic study, we discovered 10 chemicals that not only retain the pancreatic progenitors in 3D clusters but also enhance their potentiality towards NKX6.1+/INS+ ß cells. We further systematically screened signaling pathway modulators in the three steps from pancreatic progenitors toward ß cells. The implementation of all these strategies and chemical combinations resulted in generating ß cells from different sources of hPSCs with high efficiency. The derived ß cells are functional and can reverse hyperglycemia in mice within two weeks. Our protocol provides a robust platform for studying human ß cells and developing hPSC-derived ß cells for cell replacement therapy.


Homeodomain Proteins/metabolism , Insulin-Secreting Cells/metabolism , Pancreas/metabolism , Pluripotent Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Cell- and Tissue-Based Therapy , Diabetes Mellitus/metabolism , Diabetes Mellitus, Experimental , Homeodomain Proteins/genetics , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Signal Transduction
5.
Nat Commun ; 12(1): 3094, 2021 05 25.
Article En | MEDLINE | ID: mdl-34035273

Short-term, systemic expression of the Yamanaka reprogramming factors (Oct-3/4, Sox2, Klf4 and c-Myc [OSKM]) has been shown to rejuvenate aging cells and promote tissue regeneration in vivo. However, the mechanisms by which OSKM promotes tissue regeneration are unknown. In this work, we focus on a specific tissue and demonstrate that local expression of OSKM, specifically in myofibers, induces the activation of muscle stem cells or satellite cells (SCs), which accelerates muscle regeneration in young mice. In contrast, expressing OSKM directly in SCs does not improve muscle regeneration. Mechanistically, expressing OSKM in myofibers regulates the expression of genes important for the SC microenvironment, including upregulation of p21, which in turn downregulates Wnt4. This is critical because Wnt4 is secreted by myofibers to maintain SC quiescence. Thus, short-term induction of the Yamanaka factors in myofibers may promote tissue regeneration by modifying the stem cell niche.


Cell Differentiation/genetics , Cellular Reprogramming/genetics , Myofibrils/metabolism , Regeneration/genetics , Satellite Cells, Skeletal Muscle/metabolism , Stem Cell Niche , Animals , Cells, Cultured , Female , Gene Expression , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice, Transgenic , Myofibrils/physiology , Octamer Transcription Factor-3/genetics , Proto-Oncogene Proteins c-myc/genetics , SOXB1 Transcription Factors/genetics , Satellite Cells, Skeletal Muscle/cytology , Wnt4 Protein/genetics
6.
Elife ; 92020 08 11.
Article En | MEDLINE | ID: mdl-32779569

One key bottleneck in understanding the human genome is the relative under-characterization of 90% of protein coding regions. We report a collection of 1200 transgenic zebrafish strains made with the gene-break transposon (GBT) protein trap to simultaneously report and reversibly knockdown the tagged genes. Protein trap-associated mRFP expression shows previously undocumented expression of 35% and 90% of cloned genes at 2 and 4 days post-fertilization, respectively. Further, investigated alleles regularly show 99% gene-specific mRNA knockdown. Homozygous GBT animals in ryr1b, fras1, tnnt2a, edar and hmcn1 phenocopied established mutants. 204 cloned lines trapped diverse proteins, including 64 orthologs of human disease-associated genes with 40 as potential new disease models. Severely reduced skeletal muscle Ca2+ transients in GBT ryr1b homozygous animals validated the ability to explore molecular mechanisms of genetic diseases. This GBT system facilitates novel functional genome annotation towards understanding cellular and molecular underpinnings of vertebrate biology and human disease.


The human genome counts over 20,000 genes, which can be turned on and off to create the proteins required for most of life processes. Once produced, proteins need move to specific locations in the cell, where they are able to perform their jobs. Despite striking scientific advances, 90% of human genes are still under-studied; where the proteins they code for go, and what they do remains unknown. Zebrafish share many genes with humans, but they are much easier to manipulate genetically. Here, Ichino et al. used various methods in zebrafish to create a detailed 'catalogue' of previously poorly understood genes, focusing on where the proteins they coded for ended up and the biological processes they were involved with. First, a genetic tool called gene-breaking transposons (GBTs) was used to create over 1,200 strains of genetically altered fish in which a specific protein was both tagged with a luminescent marker and unable to perform its role. Further analysis of 204 of these strains revealed new insight into the role of each protein, with many having unexpected roles and localisations. For example, in one zebrafish strain, the affected gene was similar to a human gene which, when inactivated, causes severe muscle weakness. These fish swam abnormally slowly and also had muscle problems, suggesting that the GBT fish strains could 'model' the human disease. This work sheds new light on the role of many previously poorly understood genes. In the future, similar collections of GBT fish strains could help researchers to study both normal human biology and disease. They could especially be useful in cases where the genes responsible for certain conditions are still difficult to identify.


Gene Knockdown Techniques , Gene Library , Genes, Reporter , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , RNA, Messenger/genetics , Zebrafish/metabolism , Zebrafish Proteins/metabolism
7.
Sci Adv ; 5(12): eaaw1715, 2019 12.
Article En | MEDLINE | ID: mdl-31844658

Follicular helper T (TFH) cells are essential for generating protective humoral immunity. To date, microRNAs (miRNAs) have emerged as important players in regulating TFH cell biology. Here, we show that loss of miR-23~27~24 clusters in T cells resulted in elevated TFH cell frequencies upon different immune challenges, whereas overexpression of this miRNA family led to reduced TFH cell responses. Mechanistically, miR-23~27~24 clusters coordinately control TFH cells through targeting a network of genes that are crucial for TFH cell biology. Among them, thymocyte selection-associated HMG-box protein (TOX) was identified as a central transcription regulator in TFH cell development. TOX is highly up-regulated in both mouse and human TFH cells in a BCL6-dependent manner. In turn, TOX promotes the expression of multiple molecules that play critical roles in TFH cell differentiation and function. Collectively, our results establish a key miRNA regulon that maintains optimal TFH cell responses for resultant humoral immunity.


Cell Differentiation/genetics , Immunity, Humoral/genetics , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes/immunology , Animals , Gene Expression Regulation, Developmental/immunology , High Mobility Group Proteins/genetics , Humans , Immunity, Humoral/immunology , Lymphocyte Activation/immunology , Mice , MicroRNAs/genetics , Proto-Oncogene Proteins c-bcl-6/genetics , Signal Transduction , T-Lymphocytes, Helper-Inducer/metabolism
8.
Cell ; 179(3): 687-702.e18, 2019 Oct 17.
Article En | MEDLINE | ID: mdl-31626770

A single mouse blastomere from an embryo until the 8-cell stage can generate an entire blastocyst. Whether laboratory-cultured cells retain a similar generative capacity remains unknown. Starting from a single stem cell type, extended pluripotent stem (EPS) cells, we established a 3D differentiation system that enabled the generation of blastocyst-like structures (EPS-blastoids) through lineage segregation and self-organization. EPS-blastoids resembled blastocysts in morphology and cell-lineage allocation and recapitulated key morphogenetic events during preimplantation and early postimplantation development in vitro. Upon transfer, some EPS-blastoids underwent implantation, induced decidualization, and generated live, albeit disorganized, tissues in utero. Single-cell and bulk RNA-sequencing analysis revealed that EPS-blastoids contained all three blastocyst cell lineages and shared transcriptional similarity with natural blastocysts. We also provide proof of concept that EPS-blastoids can be generated from adult cells via cellular reprogramming. EPS-blastoids provide a unique platform for studying early embryogenesis and pave the way to creating viable synthetic embryos by using cultured cells.


Blastocyst/cytology , Cell Lineage , Embryo Implantation , Induced Pluripotent Stem Cells/cytology , Mouse Embryonic Stem Cells/cytology , Research Embryo Creation/methods , Animals , Blastocyst/metabolism , Cell Differentiation , Cell Line , Cells, Cultured , Cellular Reprogramming Techniques/methods , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mouse Embryonic Stem Cells/metabolism , Transcriptome
9.
Nat Med ; 25(3): 419-422, 2019 03.
Article En | MEDLINE | ID: mdl-30778240

Hutchinson-Gilford progeria syndrome (HGPS) is a rare lethal genetic disorder characterized by symptoms reminiscent of accelerated aging. The major underlying genetic cause is a substitution mutation in the gene coding for lamin A, causing the production of a toxic isoform called progerin. Here we show that reduction of lamin A/progerin by a single-dose systemic administration of adeno-associated virus-delivered CRISPR-Cas9 components suppresses HGPS in a mouse model.


CRISPR-Cas Systems , Genetic Therapy/methods , Lamin Type A/genetics , Longevity , Progeria/genetics , Animals , Disease Models, Animal , Lamin Type A/metabolism , Mice , Mutation , Protein Isoforms/genetics , Protein Isoforms/metabolism
10.
Cell ; 171(7): 1495-1507.e15, 2017 Dec 14.
Article En | MEDLINE | ID: mdl-29224783

Current genome-editing systems generally rely on inducing DNA double-strand breaks (DSBs). This may limit their utility in clinical therapies, as unwanted mutations caused by DSBs can have deleterious effects. CRISPR/Cas9 system has recently been repurposed to enable target gene activation, allowing regulation of endogenous gene expression without creating DSBs. However, in vivo implementation of this gain-of-function system has proven difficult. Here, we report a robust system for in vivo activation of endogenous target genes through trans-epigenetic remodeling. The system relies on recruitment of Cas9 and transcriptional activation complexes to target loci by modified single guide RNAs. As proof-of-concept, we used this technology to treat mouse models of diabetes, muscular dystrophy, and acute kidney disease. Results demonstrate that CRISPR/Cas9-mediated target gene activation can be achieved in vivo, leading to measurable phenotypes and amelioration of disease symptoms. This establishes new avenues for developing targeted epigenetic therapies against human diseases. VIDEO ABSTRACT.


CRISPR-Cas Systems , Epigenesis, Genetic , Gene Targeting/methods , Genetic Therapy/methods , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Utrophin/genetics , Animals , Base Sequence , Disease Models, Animal , Dystrophin/genetics , Interleukin-10/genetics , Klotho Proteins , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Transcriptional Activation
11.
Science ; 356(6337): 503-508, 2017 05 05.
Article En | MEDLINE | ID: mdl-28473583

CpG islands (CGIs) are primarily promoter-associated genomic regions and are mostly unmethylated within highly methylated mammalian genomes. The mechanisms by which CGIs are protected from de novo methylation remain elusive. Here we show that insertion of CpG-free DNA into targeted CGIs induces de novo methylation of the entire CGI in human pluripotent stem cells (PSCs). The methylation status is stably maintained even after CpG-free DNA removal, extensive passaging, and differentiation. By targeting the DNA mismatch repair gene MLH1 CGI, we could generate a PSC model of a cancer-related epimutation. Furthermore, we successfully corrected aberrant imprinting in induced PSCs derived from an Angelman syndrome patient. Our results provide insights into how CpG-free DNA induces de novo CGI methylation and broaden the application of targeted epigenome editing for a better understanding of human development and disease.


CpG Islands , DNA Methylation , Epigenesis, Genetic , Pluripotent Stem Cells/metabolism , DNA/metabolism , DNA Mismatch Repair/genetics , DNA Repair/genetics , Humans , MutL Protein Homolog 1/genetics , Mutagenesis, Insertional , Neurons/metabolism , Ubiquitin-Protein Ligases/genetics
12.
Cell Stem Cell ; 19(4): 516-529, 2016 10 06.
Article En | MEDLINE | ID: mdl-27570066

Transit-amplifying nephron progenitor cells (NPCs) generate all of the nephrons of the mammalian kidney during development. Their limited numbers, poor in vitro expansion, and difficult accessibility in humans have slowed basic and translational research into renal development and diseases. Here, we show that with appropriate 3D culture conditions, it is possible to support long-term expansion of primary mouse and human fetal NPCs as well as NPCs derived from human induced pluripotent stem cells (iPSCs). Expanded NPCs maintain genomic stability, molecular homogeneity, and nephrogenic potential in vitro, ex vivo, and in vivo. Cultured NPCs are amenable to gene targeting and can form nephron organoids that engraft in vivo, functionally couple to the host's circulatory system, and produce urine-like metabolites via filtration. Together, these findings provide a technological platform for studying human nephrogenesis, modeling and diagnosing renal diseases, and drug discovery.


Cell Culture Techniques/methods , Nephrons/cytology , Organogenesis , Stem Cells/cytology , Acute Kidney Injury/pathology , Acute Kidney Injury/physiopathology , Animals , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Gene Editing , Humans , Kidney Function Tests , Mice , Organoids/cytology , Paracrine Communication , Stem Cells/metabolism , Time Factors
14.
Nat Commun ; 6: 6413, 2015 Mar 10.
Article En | MEDLINE | ID: mdl-25752527

To combat hostile viruses, bacteria and archaea have evolved a unique antiviral defense system composed of clustered regularly interspaced short palindromic repeats (CRISPRs), together with CRISPR-associated genes (Cas). The CRISPR/Cas9 system develops an adaptive immune resistance to foreign plasmids and viruses by creating site-specific DNA double-stranded breaks (DSBs). Here we adapt the CRISPR/Cas9 system to human cells for intracellular defense against foreign DNA and viruses. Using HIV-1 infection as a model, our results demonstrate that the CRISPR/Cas9 system disrupts latently integrated viral genome and provides long-term adaptive defense against new viral infection, expression and replication in human cells. We show that engineered human-induced pluripotent stem cells stably expressing HIV-targeted CRISPR/Cas9 can be efficiently differentiated into HIV reservoir cell types and maintain their resistance to HIV-1 challenge. These results unveil the potential of the CRISPR/Cas9 system as a new therapeutic strategy against viral infections.


CD4-Positive T-Lymphocytes/immunology , CRISPR-Cas Systems/immunology , HIV Infections/prevention & control , Pluripotent Stem Cells/immunology , Base Sequence , Blotting, Western , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Dosage/genetics , HEK293 Cells , Humans , INDEL Mutation/genetics , Microscopy, Fluorescence , Molecular Sequence Data , Plasmids/genetics , Pluripotent Stem Cells/metabolism , Proviruses/genetics , Proviruses/metabolism
15.
Arterioscler Thromb Vasc Biol ; 35(4): 865-76, 2015 Apr.
Article En | MEDLINE | ID: mdl-25722433

OBJECTIVE: The E26 transformation-specific domain transcription factor Etv2/Etsrp/ER71 is a master regulator of vascular endothelial differentiation during vasculogenesis, although its later role in sprouting angiogenesis remains unknown. Here, we investigated in the zebrafish model a role for Etv2 and related E26 transformation-specific factors, Fli1a and Fli1b in developmental angiogenesis. APPROACH AND RESULTS: Zebrafish fli1a and fli1b mutants were obtained using transposon-mediated gene trap approach. Individual fli1a and fli1b homozygous mutant embryos display normal vascular patterning, yet the angiogenic recovery observed in older etv2 mutant embryos does not occur in embryos lacking both etv2 and fli1b. Etv2 and fli1b double-deficient embryos fail to form any angiogenic sprouts and show greatly increased apoptosis throughout the axial vasculature. In contrast, fli1a mutation did not affect the recovery of etv2 mutant phenotype. Overexpression analyses indicate that both etv2 and fli1b, but not fli1a, induce the expression of multiple vascular markers and of each other. Temporal inhibition of Etv2 function using photoactivatable morpholinos indicates that the function of Etv2 and Fli1b during angiogenesis is independent from the early requirement of Etv2 during vasculogenesis. RNA-Seq analysis and chromatin immunoprecipitation suggest that Etv2 and Fli1b share the same transcriptional targets and bind to the same E26 transformation-specific sites. CONCLUSIONS: Our data argue that there are 2 phases of early vascular development with distinct requirements of E26 transformation-specific transcription factors. Etv2 alone is required for early vasculogenesis, whereas Etv2 and Fli1b function redundantly during late vasculogenesis and early embryonic angiogenesis.


Angiogenic Proteins/metabolism , Endothelial Cells/metabolism , Neovascularization, Physiologic , Proto-Oncogene Protein c-fli-1/metabolism , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Angiogenic Proteins/genetics , Animals , Animals, Genetically Modified , Apoptosis , Binding Sites , Embryo, Nonmammalian/blood supply , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Genotype , Morpholinos/metabolism , Mutation , Phenotype , Promoter Regions, Genetic , Proto-Oncogene Protein c-fli-1/genetics , Signal Transduction , Time Factors , Transcription Factors/genetics , Transcription, Genetic , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
16.
Cell Stem Cell ; 15(1): 31-6, 2014 Jul 03.
Article En | MEDLINE | ID: mdl-24996168

The utility of genome editing technologies for disease modeling and developing cellular therapies has been extensively documented, but the impact of these technologies on mutational load at the whole-genome level remains unclear. We performed whole-genome sequencing to evaluate the mutational load at single-base resolution in individual gene-corrected human induced pluripotent stem cell (hiPSC) clones in three different disease models. In single-cell clones, gene correction by helper-dependent adenoviral vector (HDAdV) or Transcription Activator-Like Effector Nuclease (TALEN) exhibited few off-target effects and a low level of sequence variation, comparable to that accumulated in routine hiPSC culture. The sequence variants were randomly distributed and unique to individual clones. We also combined both technologies and developed a TALEN-HDAdV hybrid vector, which significantly increased gene-correction efficiency in hiPSCs. Therefore, with careful monitoring via whole-genome sequencing it is possible to apply genome editing to human pluripotent cells with minimal impact on genomic mutational load.


Adenoviridae/genetics , Endonucleases/metabolism , Genetic Therapy , Genetic Vectors/metabolism , Induced Pluripotent Stem Cells/physiology , CRISPR-Cas Systems/genetics , Clone Cells , DNA Repair/genetics , Endonucleases/genetics , Genetic Vectors/genetics , Genome/genetics , HEK293 Cells , Humans , Mutation/genetics , Regenerative Medicine , Sequence Analysis, DNA
17.
PLoS One ; 7(4): e34627, 2012.
Article En | MEDLINE | ID: mdl-22506037

BACKGROUND: Pen c 13, identified as a 33-kDa alkaline serine protease, is a major allergen secreted by Penicillium citrinum. Detailed knowledge about the epitopes responsible for IgE binding would help inform the diagnosis/prognosis of fungal allergy and facilitate the rational design of hypoallergenic candidate vaccines. The goal of the present study was to characterize the IgE epitopes of Pen c 13. METHODOLOGY/PRINCIPAL FINDINGS: Serum samples were collected from 10 patients with mold allergy and positive Pen c 13 skin test results. IgE-binding epitopes on rPen c 13 were mapped using an enzymatic digestion and chemical cleavage method, followed by dot-blotting and mass spectrometry. A B-cell epitope-predicting server and molecular modeling were used to predict the residues most likely involved in IgE binding. Theoretically predicted IgE-binding regions were further confirmed by site-directed mutagenesis assays. At least twelve different IgE-binding epitopes located throughout Pen c 13 were identified. Of these, peptides S16 (A(148)-E(166)) and S22 (A(243)-K(274)) were recognized by sera from 90% and 100% of the patients tested, and were further confirmed by inhibition assays. Peptide S22 was selected for further analysis of IgE-binding ability. The results of serum screening showed that the majority of IgE-binding ability resided in the C-terminus. One Pen c 13 mutant, G270A (T(261)-K(274)), exhibited clearly enhanced IgE reactivity, whereas another, K274A, exhibited dramatically reduced IgE reactivity. CONCLUSIONS/SIGNIFICANCE: Experimental analyses confirmed in silico-predicted residues involved in an important antigenic region of Pen c 13. The G270A mutant of Pen c 13 has the potential to serve as an additional tool for the diagnosis/prognosis of mold allergy, and the K274A mutant, as a hypoallergenic form of the epitope, may provide a framework for the design and development of a safe and efficient therapeutic strategy for treating human allergic diseases.


Allergens/chemistry , Antigens, Fungal/chemistry , Fungal Proteins/chemistry , Immunodominant Epitopes/chemistry , Immunoglobulin E/chemistry , Penicillium/immunology , Allergens/immunology , Allergens/metabolism , Amino Acids/chemistry , Amino Acids/metabolism , Antigens, Fungal/immunology , Antigens, Fungal/metabolism , Binding Sites, Antibody , Epitope Mapping/methods , Epitopes, B-Lymphocyte/chemistry , Epitopes, B-Lymphocyte/metabolism , Fungal Proteins/metabolism , Humans , Hypersensitivity/immunology , Hypersensitivity/metabolism , Immunodominant Epitopes/immunology , Immunodominant Epitopes/metabolism , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Mutagenesis, Site-Directed/methods , Penicillium/metabolism
18.
Dev Dyn ; 241(2): 415-25, 2012 Feb.
Article En | MEDLINE | ID: mdl-22275008

BACKGROUND: The single spanning transmembrane amyloid precursor protein (APP) and its proteolytic product, amyloid-beta (Ab) peptide, have been intensely studied due to their role in the pathogenesis of Alzheimer's disease. However, the biological role of the secreted ectodomain of APP, which is also generated by proteolytic cleavage, is less well understood. Here, we report Tol2 red fluorescent protein (RFP) transposon gene trap integrations in the zebrafish amyloid precursor protein a (appa) and amyloid precursor-like protein 2 (aplp2) genes. The transposon integrations are predicted to disrupt the appa and aplp2 genes to primarily produce secreted ectodomains of the corresponding proteins that are fused to RFP. RESULTS: Our results indicate the Appa-RFP and Aplp2 fusion proteins are likely secreted from the central nervous system and accumulate in the embryonic veins independent of blood flow. CONCLUSIONS: The zebrafish appa and aplp2 transposon insertion alleles will be useful for investigating the biological role of the secreted form of APP.


Amyloid beta-Protein Precursor/metabolism , Amyloidogenic Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Alleles , Amyloid beta-Protein Precursor/genetics , Amyloidogenic Proteins/genetics , Animals , DNA Transposable Elements/genetics , Fluorescent Dyes/metabolism , Genetic Techniques , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mutagenesis, Insertional , Veins/embryology , Veins/metabolism , Zebrafish Proteins/genetics , Red Fluorescent Protein
19.
PLoS One ; 6(4): e18826, 2011 Apr 21.
Article En | MEDLINE | ID: mdl-21533036

Large-scale sequencing of human cancer genomes and mouse transposon-induced tumors has identified a vast number of genes mutated in different cancers. One of the outstanding challenges in this field is to determine which genes, when mutated, contribute to cellular transformation and tumor progression. To identify new and conserved genes that drive tumorigenesis we have developed a novel cancer model in a distantly related vertebrate species, the zebrafish, Danio rerio. The Sleeping Beauty (SB) T2/Onc transposon system was adapted for somatic mutagenesis in zebrafish. The carp ß-actin promoter was cloned into T2/Onc to create T2/OncZ. Two transgenic zebrafish lines that contain large concatemers of T2/OncZ were isolated by injection of linear DNA into the zebrafish embryo. The T2/OncZ transposons were mobilized throughout the zebrafish genome from the transgene array by injecting SB11 transposase RNA at the 1-cell stage. Alternatively, the T2/OncZ zebrafish were crossed to a transgenic line that constitutively expresses SB11 transposase. T2/OncZ transposon integration sites were cloned by ligation-mediated PCR and sequenced on a Genome Analyzer II. Between 700-6800 unique integration events in individual fish were mapped to the zebrafish genome. The data show that introduction of transposase by transgene expression or RNA injection results in an even distribution of transposon re-integration events across the zebrafish genome. SB11 mRNA injection resulted in neoplasms in 10% of adult fish at ∼10 months of age. T2/OncZ-induced zebrafish tumors contain many mutated genes in common with human and mouse cancer genes. These analyses validate our mutagenesis approach and provide additional support for the involvement of these genes in human cancers. The zebrafish T2/OncZ cancer model will be useful for identifying novel and conserved genetic drivers of human cancers.


DNA Transposable Elements , Mutagenesis , Animals , Animals, Genetically Modified , Base Sequence , DNA/genetics , Molecular Sequence Data , Polymerase Chain Reaction , Zebrafish
20.
Nucleic Acids Res ; 39(10): 4166-79, 2011 May.
Article En | MEDLINE | ID: mdl-21266475

The bacterial recombinase RecA forms a nucleic acid-protein filament on single-stranded (ss) DNA during the repair of double-strand breaks (DSBs) that efficiently undergoes a homology search and engages in pairing with the complementary DNA sequence. We utilized the pairing activity of RecA-DNA filaments to tether biochemical activities to specific chromosomal sites. Different filaments with chimeric RecA proteins were tested for the ability to induce loss of heterozygosity at the golden locus in zebrafish after injection at the one-cell stage. A fusion protein between RecA containing a nuclear localization signal (NLS) and the DNA-binding domain of Gal4 (NLS-RecA-Gal4) displayed the most activity. Our results demonstrate that complementary ssDNA filaments as short as 60 nucleotides coated with NLS-RecA-Gal4 protein are able to cause loss of heterozygosity in ∼3% of the injected embryos. We demonstrate that lesions in ∼9% of the F0 zebrafish are transmitted to subsequent generations as large chromosomal deletions. Co-injection of linear DNA with the NLS-RecA-Gal4 DNA filaments promotes the insertion of the DNA into targeted genomic locations. Our data support a model whereby NLS-RecA-Gal4 DNA filaments bind to complementary target sites on chromatin and stall DNA replication forks, resulting in a DNA DSB.


Loss of Heterozygosity , Rec A Recombinases/metabolism , Zebrafish/genetics , Animals , DNA, Single-Stranded/administration & dosage , DNA-Binding Proteins/genetics , Embryo, Nonmammalian/anatomy & histology , Eye Color , Gene Targeting , Genes, Reporter , Genome , Injections , Mutation , Nuclear Localization Signals , Rec A Recombinases/genetics , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Zebrafish/anatomy & histology , Zebrafish/embryology
...