Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 20
1.
Nat Genet ; 55(1): 66-77, 2023 01.
Article En | MEDLINE | ID: mdl-36543915

Single-cell transcriptomics has allowed unprecedented resolution of cell types/states in the human lung, but their spatial context is less well defined. To (re)define tissue architecture of lung and airways, we profiled five proximal-to-distal locations of healthy human lungs in depth using multi-omic single cell/nuclei and spatial transcriptomics (queryable at lungcellatlas.org ). Using computational data integration and analysis, we extend beyond the suspension cell paradigm and discover macro and micro-anatomical tissue compartments including previously unannotated cell types in the epithelial, vascular, stromal and nerve bundle micro-environments. We identify and implicate peribronchial fibroblasts in lung disease. Importantly, we discover and validate a survival niche for IgA plasma cells in the airway submucosal glands (SMG). We show that gland epithelial cells recruit B cells and IgA plasma cells, and promote longevity and antibody secretion locally through expression of CCL28, APRIL and IL-6. This new 'gland-associated immune niche' has implications for respiratory health.


Lung , Respiratory Mucosa , Humans , Respiratory Mucosa/metabolism , Epithelial Cells/metabolism , B-Lymphocytes , Immunoglobulin A/metabolism
2.
Braz J Psychiatry ; 44(1): 94-102, 2022.
Article En | MEDLINE | ID: mdl-35170672

OBJECTIVE: To examine personality/temperament features and mental health vulnerability in offspring of mothers with bipolar disorders (BD), including dimensions which may impact psychological characteristics or therapeutic measures. METHODS: A systematic review, following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines, was conducted to search for original articles that investigated personality/temperament features of offspring of women with BD and emotional factors involved in the mother-child relationship. The electronic search was performed in the PubMed, Web of Science, and PsycINFO databases from February 2010 to February 2017. RESULTS: Ten quantitative studies were included in the analysis: seven from the United States, two from Brazil, and one from Canada. The narrative synthesis was categorized into three dimensions: 1) reliability of instruments for prediction of future psychopathology in offspring; 2) environmental risk factors for offspring; and 3) early interventions. The findings showed impairments in the offspring's lives, high rates of behavior and temperament problems, and psychiatric disorders. CONCLUSION: BD is a frequent psychiatric disorder, and the offspring of mothers with this condition are exposed to complex family relationships and psychosocial difficulties. If they are to ensure a good provision of mental health and psychosocial care to this unique population, early interventions must not neglect their contextual specificities. SYSTEMATIC REVIEW REGISTRATION: PROSPERO CRD-42017039010.


Bipolar Disorder , Female , Humans , Mothers , Personality Disorders , Reproducibility of Results , Temperament
3.
Braz. J. Psychiatry (São Paulo, 1999, Impr.) ; 44(1): 94-102, Jan.-Feb. 2022. tab, graf
Article En | LILACS-Express | LILACS | ID: biblio-1360170

Objective: To examine personality/temperament features and mental health vulnerability in offspring of mothers with bipolar disorders (BD), including dimensions which may impact psychological characteristics or therapeutic measures. Methods: A systematic review, following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines, was conducted to search for original articles that investigated personality/temperament features of offspring of women with BD and emotional factors involved in the mother-child relationship. The electronic search was performed in the PubMed, Web of Science, and PsycINFO databases from February 2010 to February 2017. Results: Ten quantitative studies were included in the analysis: seven from the United States, two from Brazil, and one from Canada. The narrative synthesis was categorized into three dimensions: 1) reliability of instruments for prediction of future psychopathology in offspring; 2) environmental risk factors for offspring; and 3) early interventions. The findings showed impairments in the offspring's lives, high rates of behavior and temperament problems, and psychiatric disorders. Conclusion: BD is a frequent psychiatric disorder, and the offspring of mothers with this condition are exposed to complex family relationships and psychosocial difficulties. If they are to ensure a good provision of mental health and psychosocial care to this unique population, early interventions must not neglect their contextual specificities. Systematic review registration: PROSPERO CRD-42017039010

4.
Nat Genet ; 53(11): 1553-1563, 2021 11.
Article En | MEDLINE | ID: mdl-34663923

Esophageal squamous cell carcinoma (ESCC) shows remarkable variation in incidence that is not fully explained by known lifestyle and environmental risk factors. It has been speculated that an unknown exogenous exposure(s) could be responsible. Here we combine the fields of mutational signature analysis with cancer epidemiology to study 552 ESCC genomes from eight countries with varying incidence rates. Mutational profiles were similar across all countries studied. Associations between specific mutational signatures and ESCC risk factors were identified for tobacco, alcohol, opium and germline variants, with modest impacts on mutation burden. We find no evidence of a mutational signature indicative of an exogenous exposure capable of explaining differences in ESCC incidence. Apolipoprotein B mRNA-editing enzyme, catalytic polypeptide-like (APOBEC)-associated mutational signatures single-base substitution (SBS)2 and SBS13 were present in 88% and 91% of cases, respectively, and accounted for 25% of the mutation burden on average, indicating that APOBEC activation is a crucial step in ESCC tumor development.


Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/epidemiology , Esophageal Squamous Cell Carcinoma/genetics , Mutation , APOBEC Deaminases/genetics , Adult , Aged , Aged, 80 and over , Aldehyde Dehydrogenase, Mitochondrial/genetics , Brazil/epidemiology , China/epidemiology , Female , Humans , Incidence , Iran/epidemiology , Male , Middle Aged , Tumor Suppressor Protein p53/genetics , United Kingdom/epidemiology , Whole Genome Sequencing
5.
Nature ; 597(7875): 250-255, 2021 09.
Article En | MEDLINE | ID: mdl-34497389

The cellular landscape of the human intestinal tract is dynamic throughout life, developing in utero and changing in response to functional requirements and environmental exposures. Here, to comprehensively map cell lineages, we use single-cell RNA sequencing and antigen receptor analysis of almost half a million cells from up to 5 anatomical regions in the developing and up to 11 distinct anatomical regions in the healthy paediatric and adult human gut. This reveals the existence of transcriptionally distinct BEST4 epithelial cells throughout the human intestinal tract. Furthermore, we implicate IgG sensing as a function of intestinal tuft cells. We describe neural cell populations in the developing enteric nervous system, and predict cell-type-specific expression of genes associated with Hirschsprung's disease. Finally, using a systems approach, we identify key cell players that drive the formation of secondary lymphoid tissue in early human development. We show that these programs are adopted in inflammatory bowel disease to recruit and retain immune cells at the site of inflammation. This catalogue of intestinal cells will provide new insights into cellular programs in development, homeostasis and disease.


Aging , Enteric Nervous System/cytology , Fetus/cytology , Health , Intestines/cytology , Intestines/growth & development , Lymph Nodes/cytology , Lymph Nodes/growth & development , Adult , Animals , Child , Crohn Disease/pathology , Datasets as Topic , Enteric Nervous System/anatomy & histology , Enteric Nervous System/embryology , Enteric Nervous System/growth & development , Epithelial Cells/cytology , Female , Fetus/anatomy & histology , Fetus/embryology , Humans , Intestines/embryology , Intestines/innervation , Lymph Nodes/embryology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Organogenesis , Receptors, IgG/metabolism , Signal Transduction , Spatio-Temporal Analysis , Time Factors
6.
Nat Commun ; 11(1): 3588, 2020 07 17.
Article En | MEDLINE | ID: mdl-32680985

Tumors subvert immune cell function to evade immune responses, yet the complex mechanisms driving immune evasion remain poorly understood. Here we show that tumors induce de novo steroidogenesis in T lymphocytes to evade anti-tumor immunity. Using a transgenic steroidogenesis-reporter mouse line we identify and characterize de novo steroidogenic immune cells, defining the global gene expression identity of these steroid-producing immune cells and gene regulatory networks by using single-cell transcriptomics. Genetic ablation of T cell steroidogenesis restricts primary tumor growth and metastatic dissemination in mouse models. Steroidogenic T cells dysregulate anti-tumor immunity, and inhibition of the steroidogenesis pathway is sufficient to restore anti-tumor immunity. This study demonstrates T cell de novo steroidogenesis as a mechanism of anti-tumor immunosuppression and a potential druggable target.


CD4-Positive T-Lymphocytes/immunology , Melanoma/immunology , Steroids/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cholesterol Side-Chain Cleavage Enzyme/genetics , Cholesterol Side-Chain Cleavage Enzyme/immunology , Humans , Immune Evasion , Melanoma/genetics , Melanoma/metabolism , Mice , Mice, Knockout , Steroids/biosynthesis
7.
Exp Hematol ; 76: 1-12.e5, 2019 08.
Article En | MEDLINE | ID: mdl-31326613

Pluripotent stem cell (PSC) differentiation in vitro represents a powerful and tractable model to study mammalian development and an unlimited source of cells for regenerative medicine. Within hematology, in vitro PSC hematopoiesis affords novel insights into blood formation and represents an exciting potential approach to generate hematopoietic and immune cell types for transplantation and transfusion. Most studies to date have focused on in vitro hematopoiesis from mouse PSCs and human PSCs. However, differences in mouse and human PSC culture protocols have complicated the translation of discoveries between these systems. We recently developed a novel chemical media formulation, expanded potential stem cell medium (EPSCM), that maintains mouse PSCs in a unique cellular state and extraembryonic differentiation capacity. Herein, we describe how EPSCM can be directly used to stably maintain human PSCs. We further demonstrate that human PSCs maintained in EPSCM can spontaneously form embryoid bodies and undergo in vitro hematopoiesis using a simple differentiation protocol, similar to mouse PSC differentiation. EPSCM-maintained human PSCs generated at least two hematopoietic cell populations, which displayed distinct transcriptional profiles by RNA-sequencing (RNA-seq) analysis. EPSCM also supports gene targeting using homologous recombination, affording generation of an SPI1 (PU.1) reporter PSC line to study and track in vitro hematopoiesis. EPSCM therefore provides a useful tool not only to study pluripotency but also hematopoietic cell specification and developmental-lineage commitment.


Culture Media/pharmacology , Hematopoiesis/drug effects , Human Embryonic Stem Cells/drug effects , Pluripotent Stem Cells/drug effects , Animals , Cell Culture Techniques/methods , Cell Cycle , Cell Lineage , Cells, Cultured , Cellular Reprogramming Techniques , Embryoid Bodies/drug effects , Fibroblasts/cytology , Genes, Reporter , Human Embryonic Stem Cells/cytology , Humans , Mice , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/transplantation , Sequence Analysis, RNA , Species Specificity , Stem Cell Transplantation/adverse effects , Teratoma/etiology
8.
Nat Cell Biol ; 21(6): 687-699, 2019 06.
Article En | MEDLINE | ID: mdl-31160711

We recently derived mouse expanded potential stem cells (EPSCs) from individual blastomeres by inhibiting the critical molecular pathways that predispose their differentiation. EPSCs had enriched molecular signatures of blastomeres and possessed developmental potency for all embryonic and extra-embryonic cell lineages. Here, we report the derivation of porcine EPSCs, which express key pluripotency genes, are genetically stable, permit genome editing, differentiate to derivatives of the three germ layers in chimeras and produce primordial germ cell-like cells in vitro. Under similar conditions, human embryonic stem cells and induced pluripotent stem cells can be converted, or somatic cells directly reprogrammed, to EPSCs that display the molecular and functional attributes reminiscent of porcine EPSCs. Importantly, trophoblast stem-cell-like cells can be generated from both human and porcine EPSCs. Our pathway-inhibition paradigm thus opens an avenue for generating mammalian pluripotent stem cells, and EPSCs present a unique cellular platform for translational research in biotechnology and regenerative medicine.


Cell Differentiation/genetics , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Blastomeres/cytology , Blastomeres/metabolism , Cell Lineage/genetics , Embryonic Stem Cells/cytology , Germ Layers/growth & development , Germ Layers/metabolism , Humans , Mice , Regenerative Medicine , Signal Transduction/genetics , Swine , Trophoblasts/cytology , Trophoblasts/metabolism
9.
Nat Commun ; 9(1): 3327, 2018 08 20.
Article En | MEDLINE | ID: mdl-30127402

Patients diagnosed with lung squamous cell carcinoma (LUSC) have limited targeted therapies. We report here the identification and characterisation of BCL11A, as a LUSC oncogene. Analysis of cancer genomics datasets revealed BCL11A to be upregulated in LUSC but not in lung adenocarcinoma (LUAD). Experimentally we demonstrate that non-physiological levels of BCL11A in vitro and in vivo promote squamous-like phenotypes, while its knockdown abolishes xenograft tumour formation. At the molecular level we found that BCL11A is transcriptionally regulated by SOX2 and is required for its oncogenic functions. Furthermore, we show that BCL11A and SOX2 regulate the expression of several transcription factors, including SETD8. We demonstrate that shRNA-mediated or pharmacological inhibition of SETD8 selectively inhibits LUSC growth. Collectively, our study indicates that BCL11A is integral to LUSC pathology and highlights the disruption of the BCL11A-SOX2 transcriptional programme as a novel candidate for drug development.


Carcinoma, Squamous Cell/genetics , Carrier Proteins/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Nuclear Proteins/metabolism , SOXB1 Transcription Factors/metabolism , Animals , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Cell Proliferation/genetics , Gene Knockdown Techniques , Genetic Loci , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Lung/pathology , Lung Neoplasms/drug therapy , Mice , Oncogenes , Organoids/pathology , Protein Binding , Repressor Proteins
10.
Nature ; 550(7676): 393-397, 2017 10 19.
Article En | MEDLINE | ID: mdl-29019987

Mouse embryonic stem cells derived from the epiblast contribute to the somatic lineages and the germline but are excluded from the extra-embryonic tissues that are derived from the trophectoderm and the primitive endoderm upon reintroduction to the blastocyst. Here we report that cultures of expanded potential stem cells can be established from individual eight-cell blastomeres, and by direct conversion of mouse embryonic stem cells and induced pluripotent stem cells. Remarkably, a single expanded potential stem cell can contribute both to the embryo proper and to the trophectoderm lineages in a chimaera assay. Bona fide trophoblast stem cell lines and extra-embryonic endoderm stem cells can be directly derived from expanded potential stem cells in vitro. Molecular analyses of the epigenome and single-cell transcriptome reveal enrichment for blastomere-specific signature and a dynamic DNA methylome in expanded potential stem cells. The generation of mouse expanded potential stem cells highlights the feasibility of establishing expanded potential stem cells for other mammalian species.


Blastomeres/cytology , Mouse Embryonic Stem Cells/cytology , Animals , Blastocyst/cytology , Blastomeres/metabolism , Cell Lineage , Cells, Cultured , Chimera , Embryo, Mammalian/cytology , Endoderm/cytology , Epigenesis, Genetic , Epigenomics , Female , Male , Mice , Mouse Embryonic Stem Cells/metabolism , Placenta/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Pregnancy , Single-Cell Analysis , Transcriptome , Trophoblasts/cytology
11.
Nature ; 539(7627): 102-106, 2016 11 03.
Article En | MEDLINE | ID: mdl-27749818

Innate lymphoid cells (ILCs) functionally resemble T lymphocytes in cytotoxicity and cytokine production but lack antigen-specific receptors, and they are important regulators of immune responses and tissue homeostasis. ILCs are generated from common lymphoid progenitors, which are subsequently committed to innate lymphoid lineages in the α-lymphoid progenitor, early innate lymphoid progenitor, common helper innate lymphoid progenitor and innate lymphoid cell progenitor compartments. ILCs consist of conventional natural killer cells and helper-like cells (ILC1, ILC2 and ILC3). Despite recent advances, the cellular heterogeneity, developmental trajectory and signalling dependence of ILC progenitors are not fully understood. Here, using single-cell RNA-sequencing (scRNA-seq) of mouse bone marrow progenitors, we reveal ILC precursor subsets, delineate distinct ILC development stages and pathways, and report that high expression of programmed death 1 (PD-1hi) marked a committed ILC progenitor that was essentially identical to an innate lymphoid cell progenitor. Our data defined PD-1hiIL-25Rhi as an early checkpoint in ILC2 development, which was abolished by deficiency in the zinc-finger protein Bcl11b but restored by IL-25R overexpression. Similar to T lymphocytes, PD-1 was upregulated on activated ILCs. Administration of a PD-1 antibody depleted PD-1hi ILCs and reduced cytokine levels in an influenza infection model in mice, and blocked papain-induced acute lung inflammation. These results provide a perspective for exploring PD-1 and its ligand (PD-L1) in immunotherapy, and allow effective manipulation of the immune system for disease prevention and therapy.


Base Sequence , Cell Lineage , Immunity, Innate , Lymphocytes/cytology , Lymphoid Progenitor Cells/cytology , Programmed Cell Death 1 Receptor/metabolism , Single-Cell Analysis , Animals , Antibodies/immunology , Cell Differentiation , Cell Lineage/genetics , Cell Separation , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Humans , Immunotherapy/trends , Influenza, Human/immunology , Influenza, Human/metabolism , Killer Cells, Natural/cytology , Lymphocyte Activation , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Inbred C57BL , Pneumonia/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Receptors, Interleukin/metabolism , Repressor Proteins/deficiency , Repressor Proteins/metabolism , T-Lymphocytes/metabolism , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/metabolism
12.
Stem Cells ; 33(5): 1390-404, 2015 May.
Article En | MEDLINE | ID: mdl-25546009

We previously demonstrated that coexpressing retinoic acid (RA) receptor gamma and liver receptor homolog-1 (LRH1 or NR5A2) with OCT4, MYC, KLF4, and SOX2 (4F) rapidly reprograms mouse embryonic fibroblast cells (MEFs) into induced pluripotent stem cells (iPSCs). Here, we further explore the role of RA in reprogramming and report that the six factors (6F) efficiently and directly reprogram MEFs into integration-free iPSCs in defined medium (N2B27) in the absence of feeder cells. Through genetic and chemical approaches, we find that RA signalling is essential, in a highly dose-sensitive manner, for MEF reprogramming. The removal of exogenous RA from N2B27, the inhibition of endogenous RA synthesis or the expression of a dominant-negative form of RARA severely impedes reprogramming. By contrast, supplementing N2B27 with various retinoids substantially boosts reprogramming. In addition, when coexpressed with LRH1, RA receptors (RARs) can promote reprogramming in the absence of both exogenous and endogenously synthesized RA. Remarkably, the reprogramming of epiblast stem cells into embryonic stem cell-like cells also requires low levels of RA, which can modulate Wnt signalling through physical interactions of RARs with ß-catenin. These results highlight the important functions of RA signalling in reprogramming somatic cells and primed stem cells to naïve pluripotency. Stem Cells 2015;33:1390-1404.


Cellular Reprogramming , Embryo, Mammalian/cytology , Fibroblasts/cytology , Germ Layers/cytology , Induced Pluripotent Stem Cells/cytology , Receptors, Retinoic Acid/metabolism , Signal Transduction , Animals , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Ligands , Mice , Transcription Factors , Tretinoin/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism , Retinoic Acid Receptor gamma
13.
Genome Biol ; 15(9): 455, 2014 Sep 27.
Article En | MEDLINE | ID: mdl-25260652

The in vivo validation of cancer mutations and genes identified in cancer genomics is resource-intensive because of the low throughput of animal experiments. We describe a mouse model that allows multiple cancer mutations to be validated in each animal line. Animal lines are generated with multiple candidate cancer mutations using transposons. The candidate cancer genes are tagged and randomly expressed in somatic cells, allowing easy identification of the cancer genes involved in the generated tumours. This system presents a useful, generalised and efficient means for animal validation of cancer genes.


Genetic Association Studies/methods , Neoplasms/genetics , Animals , Carcinogenesis/genetics , Cells, Cultured , Coculture Techniques , DNA Transposable Elements , Genetic Predisposition to Disease , Humans , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Multifactorial Inheritance , Mutation , Neoplasm Transplantation
14.
PLoS One ; 7(8): e40938, 2012.
Article En | MEDLINE | ID: mdl-22912667

BACKGROUND: miRNAs are a class of small non-coding RNAs that regulate gene expression and have critical functions in various biological processes. Hundreds of miRNAs have been identified in mammalian genomes but only a small number of them have been functionally characterized. Recent studies also demonstrate that some miRNAs have important roles in reprogramming somatic cells to induced pluripotent stem cells (iPSCs). METHODS: We screened 52 miRNAs cloned in a piggybac (PB) vector for their roles in reprogramming of mouse embryonic fibroblast cells to iPSCs. To identify targets of miRNAs, we made Dgcr8-deficient embryonic stem (ES) cells and introduced miRNA mimics to these cells, which lack miRNA biogenesis. The direct target genes of miRNA were identified through global gene expression analysis and target validation. RESULTS AND CONCLUSION: We found that over-expressing miR-25 or introducing miR-25 mimics enhanced production of iPSCs. We identified a number of miR-25 candidate gene targets. Of particular interest were two ubiquitin ligases, Wwp2 and Fbxw7, which have been proposed to regulate Oct4, c-Myc and Klf5, respectively. Our findings thus highlight the complex interplay between miRNAs and transcription factors involved in reprogramming, stem cell self-renewal and maintenance of pluripotency.


Cellular Reprogramming/genetics , F-Box Proteins/genetics , Fibroblasts/metabolism , Induced Pluripotent Stem Cells/cytology , MicroRNAs/genetics , MicroRNAs/metabolism , Ubiquitin-Protein Ligases/genetics , Animals , Base Sequence , Computational Biology , F-Box-WD Repeat-Containing Protein 7 , Fibroblasts/cytology , Gene Expression Regulation , Mice
15.
Proc Natl Acad Sci U S A ; 108(45): 18283-8, 2011 Nov 08.
Article En | MEDLINE | ID: mdl-21990348

Somatic cells can be reprogrammed to induced pluripotent stem cells (iPSCs) by expressing four transcription factors: Oct4, Sox2, Klf4, and c-Myc. Here we report that enhancing RA signaling by expressing RA receptors (RARs) or by RA agonists profoundly promoted reprogramming, but inhibiting it using a RAR-α dominant-negative form completely blocked it. Coexpressing Rarg (RAR-γ) and Lrh-1 (liver receptor homologue 1; Nr5a2) with the four factors greatly accelerated reprogramming so that reprogramming of mouse embryonic fibroblast cells to ground-state iPSCs requires only 4 d induction of these six factors. The six-factor combination readily reprogrammed primary human neonatal and adult fibroblast cells to exogenous factor-independent iPSCs, which resembled ground-state mouse ES cells in growth properties, gene expression, and signaling dependency. Our findings demonstrate that signaling through RARs has critical roles in molecular reprogramming and that the synergistic interaction between Rarg and Lrh1 directs reprogramming toward ground-state pluripotency. The human iPSCs described here should facilitate functional analysis of the human genome.


Cell Differentiation/physiology , Pluripotent Stem Cells/cytology , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Retinoic Acid/physiology , Animals , Cells, Cultured , Female , Humans , Kruppel-Like Factor 4 , Mice , Signal Transduction , Retinoic Acid Receptor gamma
16.
Science ; 330(6007): 1104-7, 2010 Nov 19.
Article En | MEDLINE | ID: mdl-20947725

Transposons are mobile DNA segments that can disrupt gene function by inserting in or near genes. Here, we show that insertional mutagenesis by the PiggyBac transposon can be used for cancer gene discovery in mice. PiggyBac transposition in genetically engineered transposon-transposase mice induced cancers whose type (hematopoietic versus solid) and latency were dependent on the regulatory elements introduced into transposons. Analysis of 63 hematopoietic tumors revealed that PiggyBac is capable of genome-wide mutagenesis. The PiggyBac screen uncovered many cancer genes not identified in previous retroviral or Sleeping Beauty transposon screens, including Spic, which encodes a PU.1-related transcription factor, and Hdac7, a histone deacetylase gene. PiggyBac and Sleeping Beauty have different integration preferences. To maximize the utility of the tool, we engineered 21 mouse lines to be compatible with both transposon systems in constitutive, tissue- or temporal-specific mutagenesis. Mice with different transposon types, copy numbers, and chromosomal locations support wide applicability.


DNA Transposable Elements , Genes, Neoplasm , Genetic Testing/methods , Mutagenesis, Insertional , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/genetics , Oncogenes , Promoter Regions, Genetic
17.
Epigenetics ; 4(4): 248-54, 2009 May 16.
Article En | MEDLINE | ID: mdl-19535899

DNA methylation is an important epigenetic mark that is involved in the regulation of many cellular processes such as gene expression, genomic imprinting and silencing of repetitive elements. Because of their ability to cause and capture phenotypic plasticity, epigenetic marks such as DNA methylation represent potential biomarkers to distinguish between different types of tissues and stages of differentiation. Here, we have identified differential DNA methylation in the gene body of the nitric oxide inhibitor Ddah2 that discriminates embryonic stem cells from neural stem cells and is positively correlated with differential gene expression.


Amidohydrolases/genetics , Cell Differentiation/genetics , DNA Methylation , Epigenesis, Genetic , Neurons/cytology , Stem Cells/cytology , Amidohydrolases/metabolism , Biomarkers , Cell Line , Gene Expression
18.
Proc Natl Acad Sci U S A ; 105(50): 19904-9, 2008 Dec 16.
Article En | MEDLINE | ID: mdl-19064922

Large-scale cancer genome projects will soon be able to sequence many cancer genomes to comprehensively identify genetic changes in human cancer. Genome-wide association studies have also identified putative cancer associated loci. Functional validation of these genetic mutations in vivo is becoming a challenge. We describe here a DNA transposon-based platform that permits us to explore the oncogenic potential of genetic mutations in the mouse. Briefly, promoter-less human cancer gene cDNAs were first cloned into Sleeping Beauty (SB) transposons. DNA transposition in the mouse that carried both the transposons and the SB transposase made it possible for the cDNAs to be expressed from an appropriate endogenous genomic locus and in the relevant cell types for tumor development. Consequently, these mice developed a broad spectrum of tumors at very early postnatal stages. This technology thus complements the large-scale cancer genome projects.


DNA Mutational Analysis/methods , DNA Transposable Elements/genetics , DNA, Neoplasm/genetics , Neoplasms/genetics , Oncogenes , Animals , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , Genetic Complementation Test , Genome, Human , Humans , Mice , Mice, Transgenic , Mutation , Neoplasms/pathology , Oligonucleotide Array Sequence Analysis , Transposases/genetics
19.
J Cell Sci ; 118(Pt 12): 2589-99, 2005 Jun 15.
Article En | MEDLINE | ID: mdl-15928047

Neural stem cells give rise to undifferentiated nestin-positive progenitors that undergo extensive cell division before differentiating into neuronal and glial cells. The precise control of this process is likely to be, at least in part, controlled by instructive cues originating from the extracellular environment. Some of these cues are interpreted by the integrin family of extracellular matrix receptors. Using neurosphere cell cultures as a model system, we show that beta1-integrin signalling plays a crucial role in the regulation of progenitor cell proliferation, survival and migration. Following conditional genetic ablation of the beta1-integrin allele, and consequent loss of beta1-integrin cell surface protein, mutant nestin-positive progenitor cells proliferate less and die in higher numbers than their wild-type counterparts. Mutant progenitor cell migration on different ECM substrates is also impaired. These effects can be partially compensated by the addition of exogenous growth factors. Thus, beta1-integrin signalling and growth factor signalling tightly interact to control the number and migratory capacity of nestin-positive progenitor cells.


Integrin beta1/metabolism , Neurons/cytology , Neurons/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Adhesion , Cell Death , Cell Movement/drug effects , Cell Proliferation , Cell Survival , Cells, Cultured , Female , Fibronectins/metabolism , Growth Substances/pharmacology , Integrin beta1/genetics , Intermediate Filament Proteins/metabolism , Laminin/metabolism , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nestin , Signal Transduction
20.
Development ; 131(14): 3433-44, 2004 Jul.
Article En | MEDLINE | ID: mdl-15226259

The emerging evidence that stem cells develop in specialised niches highlights the potential role of environmental factors in their regulation. Here we examine the role of beta1 integrin/extracellular matrix interactions in neural stem cells. We find high levels of beta1 integrin expression in the stem-cell containing regions of the embryonic CNS, with associated expression of the laminin alpha2 chain. Expression levels of laminin alpha2 are reduced in the postnatal CNS, but a population of cells expressing high levels of beta1 remains. Using neurospheres - aggregate cultures, derived from single stem cells, that have a three-dimensional architecture that results in the localisation of the stem cell population around the edge of the sphere - we show directly that beta1 integrins are expressed at high levels on neural stem cells and can be used for their selection. MAPK, but not PI3K, signalling is required for neural stem cell maintenance, as assessed by neurosphere formation, and inhibition or genetic ablation of beta1 integrin using cre/lox technology reduces the level of MAPK activity. We conclude that integrins are therefore an important part of the signalling mechanisms that control neural stem cell behaviour in specific areas of the CNS.


Integrin beta1/physiology , MAP Kinase Signaling System , Neurons/cytology , Stem Cells/cytology , Animals , Blotting, Western , Bromodeoxyuridine/pharmacology , Cell Culture Techniques/methods , Cell Separation , Cells, Cultured , Coloring Agents/pharmacology , Epithelial Cells/metabolism , Epithelium/metabolism , Extracellular Matrix/metabolism , Fibronectins/biosynthesis , Fibronectins/metabolism , Flow Cytometry , Immunohistochemistry , Laminin/biosynthesis , Laminin/metabolism , Mice , Mice, Knockout , Models, Biological , Neurons/metabolism , Phosphorylation , Signal Transduction , Time Factors
...