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










Publication year range
1.
PLoS One ; 13(7): e0200657, 2018.
Article in English | MEDLINE | ID: mdl-30024920

ABSTRACT

Mutations in the gene Adenomatous Polyposis Coli or APC appear in most sporadic cases of colorectal cancer and it is the most frequent mutation causing hereditary Familial Adenomatous Polyposis. The detailed molecular mechanism by which APC mutations predispose to the development of colorectal cancer is not completely understood. This is in part due to the lack of accessibility to appropriate models that recapitulate the early events associated with APC mediated intestinal transformation. We have established a novel platform utilizing human induced Pluripotent Stem cells or iPSC from normal or FAP-specific APC mutant individuals and evaluated the effect of the mutation in the cells before and after differentiation into intestinal organoids. In order to minimize genetic background effects, we also established an isogenic platform using TALEN-mediated gene editing. Comparison of normal and APC mutant iPSC revealed a significant defect in cell identity and polarity due to the presence of APC in heterozygosity as well as chromosomal aberrations including abnormal anaphases and centrosome numbers. Importantly, upon specification into intestinal progeny, APC heterozygosity was responsible for a major change in the transcriptional identity of the cells with dysregulation of key signaling pathways, including metabolic reprogramming, abnormal lipid metabolism and intestinal-specific cadherin expression. In conclusion, we have developed a novel iPSC/intestinal model of APC mutagenesis and provide strong evidence that APC in heterozygosity imparts a clear phenotypic and molecular defect, affecting basic cellular functions and integrity, providing novel insights in the earlier events of APC-mediated tumorigenesis.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli/genetics , Induced Pluripotent Stem Cells/metabolism , Mutagenesis , Mutation , Adenomatous Polyposis Coli/pathology , Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Cells, Cultured , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Intestines/pathology , Organoids/metabolism , Signal Transduction/genetics
2.
Nat Commun ; 8(1): 765, 2017 10 10.
Article in English | MEDLINE | ID: mdl-29018244

ABSTRACT

Patients with short bowel syndrome lack sufficient functional intestine to sustain themselves with enteral intake alone. Transplantable vascularized bioengineered intestine could restore nutrient absorption. Here we report the engineering of humanized intestinal grafts by repopulating decellularized rat intestinal matrix with human induced pluripotent stem cell-derived intestinal epithelium and human endothelium. After 28 days of in vitro culture, hiPSC-derived progenitor cells differentiate into a monolayer of polarized intestinal epithelium. Human endothelial cells seeded via native vasculature restore perfusability. Ex vivo isolated perfusion testing confirms transfer of glucose and medium-chain fatty acids from lumen to venous effluent. Four weeks after transplantation to RNU rats, grafts show survival and maturation of regenerated epithelium. Systemic venous sampling and positron emission tomography confirm uptake of glucose and fatty acids in vivo. Bioengineering intestine on vascularized native scaffolds could bridge the gap between cell/tissue-scale regeneration and whole organ-scale technology needed to treat intestinal failure patients.There is a need for humanised grafts to treat patients with intestinal failure. Here, the authors generate intestinal grafts by recellularizing native intestinal matrix with human induced pluripotent stem cell-derived epithelium and human endothelium, and show nutrient absorption after transplantation in rats.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Intestines/cytology , Short Bowel Syndrome/therapy , Animals , Bioengineering , Cell Differentiation , Cell Proliferation , Cells, Cultured , Endothelial Cells/cytology , Fatty Acids/metabolism , Glucose/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/transplantation , Male , Rats , Rats, Sprague-Dawley , Short Bowel Syndrome/metabolism , Tissue Engineering , Tissue Scaffolds , Transplants
3.
Nat Cell Biol ; 17(5): 545-57, 2015 May.
Article in English | MEDLINE | ID: mdl-25915124

ABSTRACT

How embryonic stem cells (ESCs) commit to specific cell lineages and yield all cell types of a fully formed organism remains a major question. ESC differentiation is accompanied by large-scale histone and DNA modifications, but the relations between these epigenetic categories are not understood. Here we demonstrate the interplay between the histone deacetylase sirtuin 6 (SIRT6) and the ten-eleven translocation enzymes (TETs). SIRT6 targets acetylated histone H3 at Lys 9 and 56 (H3K9ac and H3K56ac), while TETs convert 5-methylcytosine into 5-hydroxymethylcytosine (5hmC). ESCs derived from Sirt6 knockout (S6KO) mice are skewed towards neuroectoderm development. This phenotype involves derepression of OCT4, SOX2 and NANOG, which causes an upregulation of TET-dependent production of 5hmC. Genome-wide analysis revealed neural genes marked with 5hmC in S6KO ESCs, thereby implicating TET enzymes in the neuroectoderm-skewed differentiation phenotype. We demonstrate that SIRT6 functions as a chromatin regulator safeguarding the balance between pluripotency and differentiation through Tet-mediated production of 5hmC.


Subject(s)
Cell Differentiation , Cell Lineage , Cytosine/analogs & derivatives , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/enzymology , Proto-Oncogene Proteins/metabolism , Sirtuins/metabolism , 5-Methylcytosine/analogs & derivatives , Acetylation , Animals , Cells, Cultured , Chromatin Assembly and Disassembly , Cytosine/metabolism , DNA-Binding Proteins/genetics , Dioxygenases , Embryonic Stem Cells/pathology , Embryonic Stem Cells/transplantation , Gene Expression Regulation, Developmental , Genotype , Histones/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/enzymology , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, SCID , Nanog Homeobox Protein , Neurogenesis , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Phenotype , Proto-Oncogene Proteins/genetics , RNA Interference , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction , Sirtuins/deficiency , Sirtuins/genetics , Teratoma/enzymology , Teratoma/pathology , Transfection
4.
Mol Endocrinol ; 28(11): 1855-65, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25268051

ABSTRACT

In response to fat intake, enteroendocrine K cells release the hormone glucose-dependent insulinotropic polypeptide (GIP). GIP acts on adipocytes to increase lipid uptake and enhance adipokine secretion, promoting weight gain and insulin resistance. Modulation of intestinal GIP release could therefore represent a therapeutic strategy for the treatment and prevention of obesity and diabetes. However, the prospects of using drugs to effectively target specific enteroendocrine cell types have been tempered by the realization that these cells share similar transcriptional programs and frequently employ common mechanisms of hormone secretion. To gain novel insights into the regulation of GIP release, we generated knock-in mice expressing green fluorescent protein (GFP) under the control of the endogenous GIP promoter that enable the isolation of a purified population of small intestine K cells. Using RNA sequencing, we comprehensively characterized the transcriptomes of GIP(GFP) cells as well as the entire enteroendocrine lineage derived from Neurogenin3-expressing progenitors. Among the genes differentially expressed in GIP(GFP) cells, we identified and validated fatty acid-binding protein 5 (FABP5) as a highly expressed marker of GIP-producing cells that is absent in other enteroendocrine cell types. FABP5 promotes intracellular transport and inactivation of endocannabinoids, including anandamide, which inhibits GIP release. Remarkably, we found that circulating levels of GIP were significantly decreased in FABP5-deficient mice in the fasting state and in response to acute, oral fat diet administration. Our findings highlight the power of RNA sequencing to uncover molecular signatures of specific enteroendocrine cell types that can potentially be exploited for therapeutic purposes in the treatment of metabolic disorders.


Subject(s)
Enteroendocrine Cells/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Gastric Inhibitory Polypeptide/genetics , Gastric Inhibitory Polypeptide/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Animals , Diet , Endocannabinoids/genetics , Endocannabinoids/metabolism , Fasting/metabolism , Green Fluorescent Proteins/metabolism , Insulin Resistance/genetics , Intestine, Small , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Mice , Obesity/genetics , Obesity/metabolism , Promoter Regions, Genetic/genetics , RNA/genetics , Sequence Analysis, RNA/methods
5.
J Cell Physiol ; 228(2): 267-75, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22767332

ABSTRACT

The derivation of patient-specific pluripotent cell lines through the introduction of a few transcription factors into somatic cells has opened new avenues for the study and treatment of human disorders. Induced pluripotent stem cells (iPSCs) and their derivatives offer a unique platform for disease modeling, drug discovery and toxicology, as well as an invaluable source of cells for regenerative therapies. Here, we provide an overview of the various strategies currently available for iPSC generation, highlighting recent advances and discussing some of the challenges faced in harnessing the true potential of iPSCs for biomedical research and therapeutic applications.


Subject(s)
Cell Engineering/methods , Induced Pluripotent Stem Cells/physiology , Transcription Factors/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Genetic Vectors , Humans , Induced Pluripotent Stem Cells/cytology , Mice , Models, Biological , Rats , Transcription Factors/genetics
6.
PLoS One ; 7(12): e51711, 2012.
Article in English | MEDLINE | ID: mdl-23272148

ABSTRACT

Delivery of the transcription factors Oct4, Klf4, Sox2 and c-Myc via integrating viral vectors has been widely employed to generate induced pluripotent stem cell (iPSC) lines from both normal and disease-specific somatic tissues, providing an invaluable resource for medical research and drug development. Residual reprogramming transgene expression from integrated viruses nevertheless alters the biological properties of iPSCs and has been associated with a reduced developmental competence both in vivo and in vitro. We performed transcriptional profiling of mouse iPSC lines before and after excision of a polycistronic lentiviral reprogramming vector to systematically define the overall impact of persistent transgene expression on the molecular features of iPSCs. We demonstrate that residual expression of the Yamanaka factors prevents iPSCs from acquiring the transcriptional program exhibited by embryonic stem cells (ESCs) and that the expression profiles of iPSCs generated with and without c-Myc are indistinguishable. After vector excision, we find 36% of iPSC clones show normal methylation of the Gtl2 region, an imprinted locus that marks ESC-equivalent iPSC lines. Furthermore, we show that the reprogramming factor Klf4 binds to the promoter region of Gtl2. Regardless of Gtl2 methylation status, we find similar endodermal and hepatocyte differentiation potential comparing syngeneic Gtl2(ON) vs Gtl2(OFF) iPSC clones. Our findings provide new insights into the reprogramming process and emphasize the importance of generating iPSCs free of any residual transgene expression.


Subject(s)
Epigenesis, Genetic , Gene Expression Profiling , Induced Pluripotent Stem Cells/metabolism , Transcription Factors/genetics , Transcription, Genetic , Animals , Cell Differentiation/genetics , Cell Line , Cellular Reprogramming , Cluster Analysis , DNA Methylation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression , Gene Expression Regulation, Developmental , Gene Silencing , Hepatocytes/cytology , Hepatocytes/metabolism , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Male , Mice , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transcription Factors/metabolism , Transgenes
7.
J Clin Invest ; 121(6): 2313-25, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21537085

ABSTRACT

The directed differentiation of iPS and ES cells into definitive endoderm (DE) would allow the derivation of otherwise inaccessible progenitors for endodermal tissues. However, a global comparison of the relative equivalency of DE derived from iPS and ES populations has not been performed. Recent reports of molecular differences between iPS and ES cells have raised uncertainty as to whether iPS cells could generate autologous endodermal lineages in vitro. Here, we show that both mouse iPS and parental ES cells exhibited highly similar in vitro capacity to undergo directed differentiation into DE progenitors. With few exceptions, both cell types displayed similar surges in gene expression of specific master transcriptional regulators and global transcriptomes that define the developmental milestones of DE differentiation. Microarray analysis showed considerable overlap between the genetic programs of DE derived from ES/iPS cells in vitro and authentic DE from mouse embryos in vivo. Intriguingly, iPS cells exhibited aberrant silencing of imprinted genes known to participate in endoderm differentiation, yet retained a robust ability to differentiate into DE. Our results show that, despite some molecular differences, iPS cells can be efficiently differentiated into DE precursors, reinforcing their potential for development of cell-based therapies for diseased endoderm-derived tissues.


Subject(s)
Embryonic Stem Cells/cytology , Endoderm/cytology , Gene Expression Regulation, Developmental , Genomic Imprinting/physiology , Induced Pluripotent Stem Cells/cytology , Animals , Cell Differentiation/physiology , Cell Line/cytology , Cell Line/metabolism , Cell Lineage , Cell Separation , Chimera , Chromosome Mapping , Clone Cells/cytology , Clone Cells/metabolism , DNA Methylation , Embryonic Stem Cells/metabolism , Fetal Proteins/biosynthesis , Fetal Proteins/genetics , Genes, Reporter , Induced Pluripotent Stem Cells/metabolism , Liver/embryology , Mice , Mice, SCID , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Neoplasm Transplantation , Teratoma/pathology , Transcription Factors/biosynthesis , Transcription Factors/genetics
8.
Biomaterials ; 32(22): 5065-76, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21492931

ABSTRACT

Induced pluripotent stem cells (iPSCs) can differentiate into mineralizing cells and thus have a great potential in application in engineered bone substitutes with bioactive scaffolds in regeneration medicine. In the current study we characterized and demonstrated the pluripotency and osteogenic differentiation of mouse iPSCs. To enhance the osteogenic differentiation of iPSCs, we then transduced the iPSCs with the potent transcription factor, nuclear matrix protein SATB2. We observed that in SATB2-overexpressing iPSCs there were increased mineral nodule formation and elevated mRNA levels of key osteogenic genes, osterix (OSX), Runx2, bone sialoprotein (BSP) and osteocalcin (OCN). Moreover, the mRNA levels of HoxA2 was reduced after SATB2 overexpression in iPSCs. The SATB2-overexpressing iPSCs were then combined with silk scaffolds and transplanted into critical-size calvarial bone defects created in nude mice. Five weeks post-surgery, radiological and micro-CT analysis revealed enhanced new bone formation in calvarial defects in SATB2 group. Histological analysis also showed increased new bone formation and mineralization in the SATB2 group. In conclusion, the results demonstrate that SATB2 facilitates the differentiation of iPSCs towards osteoblast-lineage cells by repressing HoxA2 and augmenting the functions of the osteoblast determinants Runx2, BSP and OCN.


Subject(s)
Bone Regeneration/physiology , Induced Pluripotent Stem Cells/physiology , Matrix Attachment Region Binding Proteins/metabolism , Silk/chemistry , Skull/pathology , Tissue Engineering/instrumentation , Tissue Scaffolds/chemistry , Transcription Factors/metabolism , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Cell Differentiation , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Induced Pluripotent Stem Cells/cytology , Integrin-Binding Sialoprotein/genetics , Integrin-Binding Sialoprotein/metabolism , Materials Testing , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Nude , Osteocalcin/genetics , Osteocalcin/metabolism , Skull/cytology , Skull/metabolism , Sp7 Transcription Factor , Tissue Engineering/methods , Transcription Factors/genetics , Transduction, Genetic , Wound Healing/physiology
9.
Cell Stem Cell ; 8(2): 200-13, 2011 Feb 04.
Article in English | MEDLINE | ID: mdl-21295276

ABSTRACT

TET family enzymes convert 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) in DNA. Here, we show that Tet1 and Tet2 are Oct4-regulated enzymes that together sustain 5hmC in mouse embryonic stem cells (ESCs) and are induced concomitantly with 5hmC during reprogramming of fibroblasts to induced pluripotent stem cells. ESCs depleted of Tet1 by RNAi show diminished expression of the Nodal antagonist Lefty1 and display hyperactive Nodal signaling and skewed differentiation into the endoderm-mesoderm lineage in embryoid bodies in vitro. In Fgf4- and heparin-supplemented culture conditions, Tet1-depleted ESCs activate the trophoblast stem cell lineage determinant Elf5 and can colonize the placenta in midgestation embryo chimeras. Consistent with these findings, Tet1-depleted ESCs form aggressive hemorrhagic teratomas with increased endoderm, reduced neuroectoderm, and ectopic appearance of trophoblastic giant cells. Thus, 5hmC is an epigenetic modification associated with the pluripotent state, and Tet1 functions to regulate the lineage differentiation potential of ESCs.


Subject(s)
Cytosine/analogs & derivatives , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , Animals , Binding Sites/genetics , Binding Sites/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Lineage , Chromatin Immunoprecipitation , Computational Biology , Cytosine/metabolism , DNA-Binding Proteins/genetics , Dioxygenases , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins/genetics , Teratoma/genetics , Teratoma/metabolism
10.
Stem Cell Res Ther ; 1(3): 26, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20699015

ABSTRACT

Derivation of autologous induced pluripotent stem cells (iPSCs) through direct reprogramming of easily accessible somatic cells holds the potential to transform the field of regenerative medicine. Since Takahashi and Yamanaka's groundbreaking study describing the generation of iPSCs by retroviral-mediated delivery of defined transcription factors, substantial progress has been made to improve both the efficiency and safety of the method. These advances have provided new insights into the molecular mechanisms of reprogramming and promise to accelerate the clinical translation of iPSC technology. Here, we summarize current reprogramming methodologies with a focus on the production of transgene-free or genetically unmanipulated iPSCs and highlight important technical details that ultimately may influence the biological properties of pluripotent stem cells.


Subject(s)
Cellular Reprogramming/genetics , Genetic Vectors/genetics , Induced Pluripotent Stem Cells/cytology , Transcription Factors/metabolism , Cell Differentiation/genetics , Gene Transfer Techniques , Humans , Regenerative Medicine
11.
Stem Cells ; 28(10): 1728-40, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20715179

ABSTRACT

The development of methods to achieve efficient reprogramming of human cells while avoiding the permanent presence of reprogramming transgenes represents a critical step toward the use of induced pluripotent stem cells (iPSC) for clinical purposes, such as disease modeling or reconstituting therapies. Although several methods exist for generating iPSC free of reprogramming transgenes from mouse cells or neonatal normal human tissues, a sufficiently efficient reprogramming system is still needed to achieve the widespread derivation of disease-specific iPSC from humans with inherited or degenerative diseases. Here, we report the use of a humanized version of a single lentiviral "stem cell cassette" vector to accomplish efficient reprogramming of normal or diseased skin fibroblasts obtained from humans of virtually any age. Simultaneous transfer of either three or four reprogramming factors into human target cells using this single vector allows derivation of human iPSC containing a single excisable viral integration that on removal generates human iPSC free of integrated transgenes. As a proof of principle, here we apply this strategy to generate >100 lung disease-specific iPSC lines from individuals with a variety of diseases affecting the epithelial, endothelial, or interstitial compartments of the lung, including cystic fibrosis, α-1 antitrypsin deficiency-related emphysema, scleroderma, and sickle-cell disease. Moreover, we demonstrate that human iPSC generated with this approach have the ability to robustly differentiate into definitive endoderm in vitro, the developmental precursor tissue of lung epithelia.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cells, Cultured , Cellular Reprogramming/genetics , Cellular Reprogramming/physiology , Endoderm/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Genetic Vectors/genetics , Humans , Lentivirus/genetics , Reverse Transcriptase Polymerase Chain Reaction
13.
Stem Cells ; 28(1): 64-74, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19904830

ABSTRACT

The residual presence of integrated transgenes following the derivation of induced pluripotent stem (iPS) cells is highly undesirable. Here we demonstrate efficient derivation of iPS cells free of exogenous reprogramming transgenes using an excisable polycistronic lentiviral vector. A novel version of this vector containing a reporter fluorochrome allows direct visualization of vector excision in living iPS cells in real time. We find that removal of the reprogramming vector markedly improves the developmental potential of iPS cells and significantly augments their capacity to undergo directed differentiation in vitro. We further propose that methods to efficiently excise reprogramming transgenes with minimal culture passaging, such as those demonstrated here, are critical since we find that iPS cells may acquire chromosomal abnormalities, such as trisomy of chromosome 8, similar to embryonic stem cells after expansion in culture. Our findings illustrate an efficient method for the generation of transgene-free iPS cells and emphasize the potential beneficial effects that may result from elimination of integrated reprogramming factors. In addition, our results underscore the consequences of long-term culture that will need to be taken into account for the clinical application of iPS cells.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming , Gene Expression Regulation, Developmental , Genetic Vectors , Induced Pluripotent Stem Cells/metabolism , Lentivirus/genetics , Transduction, Genetic , Transgenes , Animals , Biomarkers/metabolism , Cell Proliferation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Integrases/metabolism , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Recombinant Fusion Proteins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Teratoma/genetics , Teratoma/metabolism , Time Factors , Trisomy , Virus Integration
14.
Stem Cells ; 27(3): 543-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19096035

ABSTRACT

Induced pluripotent stem (iPS) cells can be generated using retroviral vectors expressing Oct4, Klf4, Sox2, and cMyc. Most prior studies have required multiple retroviral vectors for reprogramming, resulting in high numbers of genomic integrations in iPS cells and limiting their use for therapeutic applications. Here we describe the use of a single lentiviral vector expressing a "stem cell cassette" composed of the four transcription factors and a combination of 2A peptide and internal ribosome entry site technology, generating iPS cells from postnatal fibroblasts. iPS cells generated in this manner display embryonic stem cell-like morphology, express stem cell markers, and exhibit in vivo pluripotency, as evidenced by their ability to differentiate in teratoma assays and their robust contribution to mouse chimeras. Combining all factors into a single transcript achieves the most efficient reprogramming system to date and allows derivation of iPS cells with a single viral integration. The use of a single lentiviral vector for reprogramming represents a powerful laboratory tool and a significant step toward the application of iPS technology for clinical purposes.


Subject(s)
Lentivirus/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Blotting, Southern , Cells, Cultured , Genetic Vectors/genetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/physiology , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/physiology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/physiology , Reverse Transcriptase Polymerase Chain Reaction , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/physiology
15.
Genome ; 51(1): 19-29, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18356936

ABSTRACT

The molecular mechanisms by which trisomy of human chromosome 21 disrupts normal development are not well understood. Global transcriptome studies attempting to analyze the consequences of trisomy in Down syndrome (DS) tissues have reported conflicting results, which have led to the suggestion that the analysis of specific tissues or cell types may be more productive. In the present study, we set out to analyze global changes of gene expression in lymphocytes from children with trisomy 21 by means of the serial analysis of gene expression (SAGE) methodology. Two SAGE libraries were constructed using pooled RNA of normal and Down syndrome children. Comparison between DS and normal profiles revealed that most of the transcripts were expressed at similar levels and functional classes of abundant genes were equally represented. Among the 242 significantly differentially expressed SAGE tags, several transcripts downregulated in DS code for proteins involved in T-cell and B-cell receptor signaling (e.g., PI3Kdelta, RGS2, LY6E, FOS, TAGAP, CD46). The SAGE data and interindividual variability were validated by real-time quantitative PCR. Our results indicate that trisomy 21 induces a modest dysregulation of disomic genes that may be related to the immunological perturbations seen in DS.


Subject(s)
Down Syndrome/genetics , Gene Expression Regulation , Child , Chromosome Mapping , Down Syndrome/immunology , Gene Expression Profiling , Humans , Lymphocytes/immunology , Reverse Transcriptase Polymerase Chain Reaction , Sequence Tagged Sites
16.
Biol Chem ; 389(4): 447-53, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18208350

ABSTRACT

The lysosomal cysteine proteases cathepsin B and L play important roles in tumor cell invasion. An imbalance between these cathepsins and their endogenous inhibitors, the cystatins, has been associated with development of the metastatic phenotype. Accordingly, many studies have indicated potential use of cystatins in therapeutic approaches. We report a novel cystatin from sugarcane (Saccharum officinarum), CaneCPI-4, with strong inhibitory activity against cathepsins B (K(i) = 0.83 nM) and L (K(i) = 0.021 nM). The invasive ability of MDA-MB-231 human breast cancer cells expressing CaneCPI-4 was only slightly decreased. In contrast, addition of low, non-toxic concentrations of recombinant His-tagged CaneCPI-4 significantly reduced invasion through a Matrigel matrix. Immunoblot analyses failed to detect the recombinant protein inside cells, indicating that the cystatin was not internalized by endocytosis, but exerted its anti-invasive effect mainly through inhibition of extracellular cathepsins. Our findings open the possibility of considering phytocystatins for anti-cancer strategies.


Subject(s)
Cathepsin B/metabolism , Cathepsins/metabolism , Cystatins/metabolism , Cystatins/pharmacology , Saccharum/metabolism , Amino Acid Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cathepsin B/antagonists & inhibitors , Cathepsins/antagonists & inhibitors , Cell Line, Tumor , Cystatins/genetics , Humans , Immunoblotting , Molecular Sequence Data , Neoplasm Invasiveness/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
17.
Protein Expr Purif ; 45(2): 324-8, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16214370

ABSTRACT

The ubiquitin system represents a selective mechanism for intracellular proteolysis in eukaryotic cells that involves the sequential activity of three enzymes, ubiquitin-activating enzyme (E1), ubiquitin-conjugating enzyme (E2), and ubiquitin-protein ligase (E3). The identification of these proteins and their cellular targets, as well as structural data, are essential to understanding how this system operates in the eukaryotic cell. In the present study, the open reading frame of the human ubiquitin-conjugating enzyme UBE2G2 was isolated from a human brain cDNA panel, cloned into pET28a vector and expressed in Escherichia coli. The His-tagged protein was then purified through nickel-affinity chromatography and subjected to structural and functional studies using circular dichroism (CD) and an in vitro ubiquitin-binding assay, respectively. Our results showed that the production of the HISUBE2G2 protein in bacteria, carried out with 0.1 mM of IPTG at 30 degrees C, was successfully achieved, rendering high concentrations of soluble, pure and stable enzyme after a single purification step. The recombinant protein was able to bind ubiquitin molecules when exposed to a HeLa cell extract during the ubiquitin assay. Moreover, the fact that HISUBE2G2 was expressed in its active form is supported by the typical alpha/beta secondary structure specific to other class I E2 enzymes displayed during the CD assay.


Subject(s)
Ubiquitin-Conjugating Enzymes , Circular Dichroism , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , HeLa Cells , Humans , Open Reading Frames , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Ubiquitin-Conjugating Enzymes/chemistry , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/isolation & purification , Ubiquitin-Conjugating Enzymes/metabolism
18.
Croat Med J ; 46(4): 647-56, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16100769

ABSTRACT

AIM: Identification of differences in the gene expression patterns of Down syndrome and normal leukocytes. METHODS: We constructed the first Down syndrome leukocyte serial analysis of gene expression (SAGE) library from a 28 year-old patient. This library was analyzed and compared with a normal leukocyte SAGE library using the eSAGE software. Reverse transcriptase polymerase chain reaction (RT-PCR) was used to validate the results. RESULTS: We found that a large number of unidentified transcripts were overexpressed in Down syndrome leukocytes and some transcripts coding for growth factors (e.g. interleukin 8, IL-8), ribosomaproteins (e.g. L13a, L29, and L37), and transcription factors (e.g., Jun B, Jun D, and C/EBP beta) were underexpressed. The SAGE data were successfully validated for the genes IL-8, CXCR4, BCL2A1, L13a, L29, L37, and GTF3A using RT-PCR. CONCLUSION: Our analysis identified significant changes in the expression pattern of Down syndrome leukocytes compared with normal ones, including key regulators of growth and proliferation, ribosomal proteins, and a large number of overexpressed transcripts that were not matched in UniGene clusters and that may represent novel genes related to Down syndrome. This study offers a new insight into transcriptional changes in Down syndrome leukocytes and indicates candidate genes for further investigations into the molecular mechanism of Down syndrome pathology.


Subject(s)
Down Syndrome/genetics , Gene Expression Profiling , Leukocytes/metabolism , Adult , Base Sequence , DNA Primers , Down Syndrome/blood , Humans , Male , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...