Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(26): e2214842120, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37339216

ABSTRACT

Transplantation of stem cell-derived retinal pigment epithelial (RPE) cells is considered a viable therapeutic option for age-related macular degeneration (AMD). Several landmark Phase I/II clinical trials have demonstrated safety and tolerability of RPE transplants in AMD patients, albeit with limited efficacy. Currently, there is limited understanding of how the recipient retina regulates the survival, maturation, and fate specification of transplanted RPE cells. To address this, we transplanted stem cell-derived RPE into the subretinal space of immunocompetent rabbits for 1 mo and conducted single-cell RNA sequencing analyses on the explanted RPE monolayers, compared to their age-matched in vitro counterparts. We observed an unequivocal retention of RPE identity, and a trajectory-inferred survival of all in vitro RPE populations after transplantation. Furthermore, there was a unidirectional maturation toward the native adult human RPE state in all transplanted RPE, regardless of stem cell resource. Gene regulatory network analysis suggests that tripartite transcription factors (FOS, JUND, and MAFF) may be specifically activated in posttransplanted RPE cells, to regulate canonical RPE signature gene expression crucial for supporting host photoreceptor function, and to regulate prosurvival genes required for transplanted RPE's adaptation to the host subretinal microenvironment. These findings shed insights into the transcriptional landscape of RPE cells after subretinal transplantation, with important implications for cell-based therapy for AMD.


Subject(s)
Macular Degeneration , Transcriptome , Adult , Animals , Humans , Rabbits , Macular Degeneration/genetics , Macular Degeneration/therapy , Stem Cells , Epithelial Cells , Retinal Pigments
2.
Nat Commun ; 13(1): 2796, 2022 05 19.
Article in English | MEDLINE | ID: mdl-35589753

ABSTRACT

One common cause of vision loss after retinal detachment surgery is the formation of proliferative and contractile fibrocellular membranes. This aberrant wound healing process is mediated by epithelial-mesenchymal transition (EMT) and hyper-proliferation of retinal pigment epithelial (RPE) cells. Current treatment relies primarily on surgical removal of these membranes. Here, we demonstrate that a bio-functional polymer by itself is able to prevent retinal scarring in an experimental rabbit model of proliferative vitreoretinopathy. This is mediated primarily via clathrin-dependent internalisation of polymeric micelles, downstream suppression of canonical EMT transcription factors, reduction of RPE cell hyper-proliferation and migration. Nuclear factor erythroid 2-related factor 2 signalling pathway was identified in a genome-wide transcriptomic profiling as a key sensor and effector. This study highlights the potential of using synthetic bio-functional polymer to modulate RPE cellular behaviour and offers a potential therapy for retinal scarring prevention.


Subject(s)
NF-E2-Related Factor 2 , Retinal Pigment Epithelium , Animals , Cell Line , Cell Movement , Cicatrix/metabolism , Epithelial-Mesenchymal Transition , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Polymers/metabolism , Rabbits , Retinal Pigment Epithelium/metabolism
3.
J Clin Med ; 9(9)2020 Sep 10.
Article in English | MEDLINE | ID: mdl-32927780

ABSTRACT

(1) Background: Intravitreal anti-vascular endothelial growth factor (anti-VEGF) is an established treatment for center-involving diabetic macular edema (ci-DME). However, the clinical response is heterogeneous. This study investigated miRNAs as a biomarker to predict treatment response to anti-VEGF in DME. (2) Methods: Tear fluid, aqueous, and blood were collected from patients with treatment-naïve DME for miRNA expression profiling with quantitative polymerase chain reaction. Differentially expressed miRNAs between good and poor responders were identified from tear fluid. Bioinformatics analysis with the miEAA tool, miRTarBase Annotations, Gene Ontology categories, KEGG, and miRWalk pathways identified interactions between enriched miRNAs and biological pathways. (3) Results: Of 24 participants, 28 eyes received bevacizumab (15 eyes) or aflibercept (13 eyes). Tear fluid had the most detectable miRNA species (N = 315), followed by serum (N = 309), then aqueous humor (N = 134). MiRNAs that correlated with change in macular thickness were miR-214-3p, miR-320d, and hsa-miR-874-3p in good responders; and miR-98-5p, miR-196b-5p, and miR-454-3p in poor responders. VEGF-related pathways and the angiogenin-PRI complex were enriched in good responders, while transforming growth factor-ß and insulin-like growth factor pathways were enriched in poor responders. (4) Conclusions: We reported a panel of novel miRNAs that provide insight into biological pathways in DME. Validation in larger independent cohorts is needed to determine the predictive performance of these miRNA candidate biomarkers.

4.
Nat Commun ; 10(1): 3653, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31409800

ABSTRACT

Urinary sodium and potassium excretion are associated with blood pressure (BP) and cardiovascular disease (CVD). The exact biological link between these traits is yet to be elucidated. Here, we identify 50 loci for sodium and 13 for potassium excretion in a large-scale genome-wide association study (GWAS) on urinary sodium and potassium excretion using data from 446,237 individuals of European descent from the UK Biobank study. We extensively interrogate the results using multiple analyses such as Mendelian randomization, functional assessment, co localization, genetic risk score, and pathway analyses. We identify a shared genetic component between urinary sodium and potassium expression and cardiovascular traits. Ingenuity pathway analysis shows that urinary sodium and potassium excretion loci are over-represented in behavioural response to stimuli. Our study highlights pathways that are shared between urinary sodium and potassium excretion and cardiovascular traits.


Subject(s)
Cardiovascular Diseases/genetics , Genome-Wide Association Study , Potassium/urine , Sodium/urine , Blood Pressure , Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/urine , Female , Humans , Male , Polymorphism, Single Nucleotide
5.
Nat Hum Behav ; 3(9): 950-961, 2019 09.
Article in English | MEDLINE | ID: mdl-31358974

ABSTRACT

Excessive alcohol consumption is one of the main causes of death and disability worldwide. Alcohol consumption is a heritable complex trait. Here we conducted a meta-analysis of genome-wide association studies of alcohol consumption (g d-1) from the UK Biobank, the Alcohol Genome-Wide Consortium and the Cohorts for Heart and Aging Research in Genomic Epidemiology Plus consortia, collecting data from 480,842 people of European descent to decipher the genetic architecture of alcohol intake. We identified 46 new common loci and investigated their potential functional importance using magnetic resonance imaging data and gene expression studies. We identify genetic pathways associated with alcohol consumption and suggest genetic mechanisms that are shared with neuropsychiatric disorders such as schizophrenia.


Subject(s)
Alcohol Drinking/genetics , Genes/genetics , Genetic Predisposition to Disease/genetics , Mental Disorders/genetics , Adult , Aged , Alcoholism/genetics , Brain/physiopathology , Female , Genes/physiology , Genome-Wide Association Study , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neuroimaging , Polymorphism, Single Nucleotide/genetics , Quantitative Trait Loci/genetics , Schizophrenia/genetics , White People/genetics
6.
Dev Cell ; 47(5): 645-659.e6, 2018 12 03.
Article in English | MEDLINE | ID: mdl-30393076

ABSTRACT

Meiotic synapsis and recombination ensure correct homologous segregation and genetic diversity. Asynapsed homologs are transcriptionally inactivated by meiotic silencing, which serves a surveillance function and in males drives meiotic sex chromosome inactivation. Silencing depends on the DNA damage response (DDR) network, but how DDR proteins engage repressive chromatin marks is unknown. We identify the histone H3-lysine-9 methyltransferase SETDB1 as the bridge linking the DDR to silencing in male mice. At the onset of silencing, X chromosome H3K9 trimethylation (H3K9me3) enrichment is downstream of DDR factors. Without Setdb1, the X chromosome accrues DDR proteins but not H3K9me3. Consequently, sex chromosome remodeling and silencing fail, causing germ cell apoptosis. Our data implicate TRIM28 in linking the DDR to SETDB1 and uncover additional factors with putative meiotic XY-silencing functions. Furthermore, we show that SETDB1 imposes timely expression of meiotic and post-meiotic genes. Setdb1 thus unites the DDR network, asynapsis, and meiotic chromosome silencing.


Subject(s)
Chromosome Pairing , DNA Damage , Gene Silencing , Histone Code , Histone-Lysine N-Methyltransferase/metabolism , Animals , Apoptosis , DNA Repair , Histone-Lysine N-Methyltransferase/genetics , Histones/metabolism , Male , Mice , Mice, Inbred C57BL , Tripartite Motif-Containing Protein 28/genetics , Tripartite Motif-Containing Protein 28/metabolism
8.
Nature ; 550(7674): 67-73, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28953884

ABSTRACT

Despite their fundamental biological and clinical importance, the molecular mechanisms that regulate the first cell fate decisions in the human embryo are not well understood. Here we use CRISPR-Cas9-mediated genome editing to investigate the function of the pluripotency transcription factor OCT4 during human embryogenesis. We identified an efficient OCT4-targeting guide RNA using an inducible human embryonic stem cell-based system and microinjection of mouse zygotes. Using these refined methods, we efficiently and specifically targeted the gene encoding OCT4 (POU5F1) in diploid human zygotes and found that blastocyst development was compromised. Transcriptomics analysis revealed that, in POU5F1-null cells, gene expression was downregulated not only for extra-embryonic trophectoderm genes, such as CDX2, but also for regulators of the pluripotent epiblast, including NANOG. By contrast, Pou5f1-null mouse embryos maintained the expression of orthologous genes, and blastocyst development was established, but maintenance was compromised. We conclude that CRISPR-Cas9-mediated genome editing is a powerful method for investigating gene function in the context of human development.


Subject(s)
Embryonic Development/genetics , Gene Editing , Gene Expression Regulation, Developmental , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Animals , Blastocyst/metabolism , CRISPR-Cas Systems/genetics , Cell Lineage , Ectoderm/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Germ Layers/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Male , Mice , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/deficiency , Substrate Specificity , Zygote/metabolism
10.
Nature ; 534(7607): 383-6, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27281217

ABSTRACT

Mitochondrial DNA (mtDNA) mutations are maternally inherited and are associated with a broad range of debilitating and fatal diseases. Reproductive technologies designed to uncouple the inheritance of mtDNA from nuclear DNA may enable affected women to have a genetically related child with a greatly reduced risk of mtDNA disease. Here we report the first preclinical studies on pronuclear transplantation (PNT). Surprisingly, techniques used in proof-of-concept studies involving abnormally fertilized human zygotes were not well tolerated by normally fertilized zygotes. We have therefore developed an alternative approach based on transplanting pronuclei shortly after completion of meiosis rather than shortly before the first mitotic division. This promotes efficient development to the blastocyst stage with no detectable effect on aneuploidy or gene expression. After optimization, mtDNA carryover was reduced to <2% in the majority (79%) of PNT blastocysts. The importance of reducing carryover to the lowest possible levels is highlighted by a progressive increase in heteroplasmy in a stem cell line derived from a PNT blastocyst with 4% mtDNA carryover. We conclude that PNT has the potential to reduce the risk of mtDNA disease, but it may not guarantee prevention.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondrial Diseases/genetics , Mitochondrial Diseases/prevention & control , Mitochondrial Replacement Therapy/methods , Nuclear Transfer Techniques , Adult , Blastocyst/cytology , Blastocyst/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , DNA, Mitochondrial/analysis , Female , Gene Expression Profiling , Humans , Male , Meiosis , Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Diseases/pathology , Stem Cells/cytology , Stem Cells/metabolism , Translational Research, Biomedical , Young Adult , Zygote/cytology , Zygote/metabolism
11.
Development ; 142(20): 3613, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26487783

ABSTRACT

There were errors published in Development 142, 3151-3165.In the issue published online on 22 September 2015, Fig. 3 was mislabelled: panels A, B, C and D should have been B, C, D and A, respectively. In the legend, the text prior to '(A) Cytoscape enrichment map…' should not have been included. The correct version of the figure and legend now appear online and in print.We apologise to the authors and readers for this mistake.

12.
Development ; 142(18): 3151-65, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26293300

ABSTRACT

Here, we provide fundamental insights into early human development by single-cell RNA-sequencing of human and mouse preimplantation embryos. We elucidate conserved transcriptional programs along with those that are human specific. Importantly, we validate our RNA-sequencing findings at the protein level, which further reveals differences in human and mouse embryo gene expression. For example, we identify several genes exclusively expressed in the human pluripotent epiblast, including the transcription factor KLF17. Key components of the TGF-ß signalling pathway, including NODAL, GDF3, TGFBR1/ALK5, LEFTY1, SMAD2, SMAD4 and TDGF1, are also enriched in the human epiblast. Intriguingly, inhibition of TGF-ß signalling abrogates NANOG expression in human epiblast cells, consistent with a requirement for this pathway in pluripotency. Although the key trophectoderm factors Id2, Elf5 and Eomes are exclusively localized to this lineage in the mouse, the human orthologues are either absent or expressed in alternative lineages. Importantly, we also identify genes with conserved expression dynamics, including Foxa2/FOXA2, which we show is restricted to the primitive endoderm in both human and mouse embryos. Comparison of the human epiblast to existing embryonic stem cells (hESCs) reveals conservation of pluripotency but also additional pathways more enriched in hESCs. Our analysis highlights significant differences in human preimplantation development compared with mouse and provides a molecular blueprint to understand human embryogenesis and its relationship to stem cells.


Subject(s)
Blastocyst/cytology , Cell Lineage/physiology , Gene Expression Regulation, Developmental/physiology , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Animals , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Humans , Mice , Principal Component Analysis , Species Specificity
13.
Genes Dev ; 29(12): 1239-55, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-26109048

ABSTRACT

Transcription factor-mediated reprograming is a powerful method to study cell fate changes. In this study, we demonstrate that the transcription factor Gata6 can initiate reprograming of multiple cell types to induced extraembryonic endoderm stem (iXEN) cells. Intriguingly, Gata6 is sufficient to drive iXEN cells from mouse pluripotent cells and differentiated neural cells. Furthermore, GATA6 induction in human embryonic stem (hES) cells also down-regulates pluripotency gene expression and up-regulates extraembryonic endoderm (ExEn) genes, revealing a conserved function in mediating this cell fate switch. Profiling transcriptional changes following Gata6 induction in mES cells reveals step-wise pluripotency factor disengagement, with initial repression of Nanog and Esrrb, then Sox2, and finally Oct4, alongside step-wise activation of ExEn genes. Chromatin immunoprecipitation and subsequent high-throughput sequencing analysis shows Gata6 enrichment near pluripotency and endoderm genes, suggesting that Gata6 functions as both a direct repressor and activator. Together, this demonstrates that Gata6 is a versatile and potent reprograming factor that can act alone to drive a cell fate switch from diverse cell types.


Subject(s)
Cellular Reprogramming/genetics , Embryonic Stem Cells/cytology , Endoderm/cytology , GATA6 Transcription Factor/metabolism , Pluripotent Stem Cells/cytology , Animals , Binding Sites , Cell Differentiation , Fibroblast Growth Factor 4/genetics , Fibroblast Growth Factor 4/metabolism , GATA4 Transcription Factor/genetics , GATA4 Transcription Factor/metabolism , GATA6 Transcription Factor/genetics , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Humans , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Protein Binding , Signal Transduction
14.
J Psychosom Res ; 75(4): 341-5, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24119940

ABSTRACT

OBJECTIVE: Maternal prenatal symptoms of depression and anxiety have been associated with altered neurodevelopmental outcomes in the child. These effects may be mediated in part by altered placental function, with increased fetal 5-hydroxytryptamine (5-HT) exposure being one possible mechanism. The current study aimed to determine whether maternal symptoms of depression or anxiety were associated with decreased placental expression of monoamine oxidase A (MAO A), the enzyme which metabolises 5-HT into 5-hydroxyindoleacetic acid. The localisation of MAO A in the placenta was also investigated. METHODS: Pregnant women were recruited one day prior to elective caesarean and assessed using psychometric tests for symptoms of depression (Edinburgh Depression Scale) and anxiety (Spielberger State/Trait Index). Villous trophoblast tissue was extracted from each placenta and used for subsequent gene expression analysis (N=62). Localisation was studied using immunohistochemistry, with a specific polyclonal antibody. RESULTS: Increasing symptoms of maternal depression were associated with a reduction in placental MAO A expression (r=-0.339, p=0.007, N=62). There was a trend for a similar correlation with symptoms of maternal trait anxiety, but not with state anxiety. MAO A was localised to the syncytiotrophoblast, the tissue between maternal and fetal blood. CONCLUSIONS: These findings support the hypothesis that maternal mood is associated with altered placental function. A reduction in placental MAO A expression is consistent with a subsequent increase in fetal exposure to 5-HT.


Subject(s)
Anxiety/enzymology , Depression/enzymology , Monoamine Oxidase/metabolism , Placenta/enzymology , Pregnant Women/psychology , Serotonin/metabolism , Adult , Affect , Child , Down-Regulation , Female , Humans , Immunohistochemistry , Monoamine Oxidase/analysis , Monoamine Oxidase/genetics , Pregnancy , Psychiatric Status Rating Scales , Trophoblasts/enzymology
15.
J Proteome Res ; 11(11): 5221-34, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-23025403

ABSTRACT

Proteogenomics has the potential to advance genome annotation through high quality peptide identifications derived from mass spectrometry experiments, which demonstrate a given gene or isoform is expressed and translated at the protein level. This can advance our understanding of genome function, discovering novel genes and gene structure that have not yet been identified or validated. Because of the high-throughput shotgun nature of most proteomics experiments, it is essential to carefully control for false positives and prevent any potential misannotation. A number of statistical procedures to deal with this are in wide use in proteomics, calculating false discovery rate (FDR) and posterior error probability (PEP) values for groups and individual peptide spectrum matches (PSMs). These methods control for multiple testing and exploit decoy databases to estimate statistical significance. Here, we show that database choice has a major effect on these confidence estimates leading to significant differences in the number of PSMs reported. We note that standard target:decoy approaches using six-frame translations of nucleotide sequences, such as assembled transcriptome data, apparently underestimate the confidence assigned to the PSMs. The source of this error stems from the inflated and unusual nature of the six-frame database, where for every target sequence there exists five "incorrect" targets that are unlikely to code for protein. The attendant FDR and PEP estimates lead to fewer accepted PSMs at fixed thresholds, and we show that this effect is a product of the database and statistical modeling and not the search engine. A variety of approaches to limit database size and remove noncoding target sequences are examined and discussed in terms of the altered statistical estimates generated and PSMs reported. These results are of importance to groups carrying out proteogenomics, aiming to maximize the validation and discovery of gene structure in sequenced genomes, while still controlling for false positives.


Subject(s)
Databases, Protein , Genomics , Nucleotides/chemistry , Proteomics , Base Sequence , Expressed Sequence Tags , Mass Spectrometry , Probability
16.
Proteomics ; 10(6): 1127-40, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20077415

ABSTRACT

Alternative splicing (AS) and processing of pre-messenger RNAs explains the discrepancy between the number of genes and proteome complexity in multicellular eukaryotic organisms. However, relatively few alternative protein isoforms have been experimentally identified, particularly at the protein level. In this study, we assess the ability of proteomics to inform on differently spliced protein isoforms in human and four other model eukaryotes. The number of Ensembl-annotated genes for which proteomic data exists that informs on AS exceeds 33% of the alternately spliced genes in the human and worm genomes. Examining AS in chicken via proteomics for the first time, we find support for over 600 AS genes. However, although peptide identifications support only a small fraction of alternative protein isoforms that are annotated in Ensembl, many more variants are amenable to proteomic identification. There remains a sizeable gap between these existing identifications (10-52% of AS genes) and those that are theoretically feasible (90-99%). We also compare annotations between Swiss-Prot and Ensembl, recommending use of both to maximize coverage of AS. We propose that targeted proteomic experiments using selected reactions and standards are essential to uncover further alternative isoforms and discuss the issues surrounding these strategies.


Subject(s)
Peptides/chemistry , Protein Isoforms/genetics , Proteomics/methods , Alternative Splicing , Amino Acid Sequence , Animals , Chickens , Computational Biology , Databases, Protein , Feasibility Studies , Genome, Human , Humans , Molecular Sequence Data , NADH Dehydrogenase/genetics , Proteome/genetics , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...