32 results on '"Sue Mei Lim"'
Search Results
2. Cell Type of Origin Dictates the Route to Pluripotency
- Author
-
Christian M. Nefzger, Fernando J. Rossello, Joseph Chen, Xiaodong Liu, Anja S. Knaupp, Jaber Firas, Jacob M. Paynter, Jahnvi Pflueger, Sam Buckberry, Sue Mei Lim, Brenda Williams, Sara Alaei, Keshav Faye-Chauhan, Enrico Petretto, Susan K. Nilsson, Ryan Lister, Mirana Ramialison, David R. Powell, Owen J.L. Rackham, and Jose M. Polo
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: Our current understanding of induced pluripotent stem cell (iPSC) generation has almost entirely been shaped by studies performed on reprogramming fibroblasts. However, whether the resulting model universally applies to the reprogramming process of other cell types is still largely unknown. By characterizing and profiling the reprogramming pathways of fibroblasts, neutrophils, and keratinocytes, we unveil that key events of the process, including loss of original cell identity, mesenchymal to epithelial transition, the extent of developmental reversion, and reactivation of the pluripotency network, are to a large degree cell-type specific. Thus, we reveal limitations for the use of fibroblasts as a universal model for the study of the reprogramming process and provide crucial insights about iPSC generation from alternative cell sources. : Nefzger et al. find that the molecular reprogramming trajectories of fibroblasts, neutrophils, and keratinocytes have a cell-type-specific component that only fully converges in induced pluripotent stem cells. The authors also identify universal changes shared by all three cell types, including two transcriptional waves and a conserved transcriptional program involving Egr1 downregulation. Keywords: reprogramming, induced pluripotent stem cells, fibroblasts, neutrophils, keratinocytes, transcriptional dynamics, Egr1
- Published
- 2017
- Full Text
- View/download PDF
3. Brachyury and related Tbx proteins interact with the Mixl1 homeodomain protein and negatively regulate Mixl1 transcriptional activity.
- Author
-
Lloyd A Pereira, Michael S Wong, Sue Mei Lim, Alexandra Sides, Edouard G Stanley, and Andrew G Elefanty
- Subjects
Medicine ,Science - Abstract
Mixl1 is a homeodomain transcription factor required for mesoderm and endoderm patterning during mammalian embryogenesis. Despite its crucial function in development, co-factors that modulate the activity of Mixl1 remain poorly defined. Here we report that Mixl1 interacts physically and functionally with the T-box protein Brachyury and related members of the T-box family of transcription factors. Transcriptional and protein analyses demonstrated overlapping expression of Mixl1 and Brachyury during embryonic stem cell differentiation. In vitro protein interaction studies showed that the Mixl1 with Brachyury associated via their DNA-binding domains and gel shift assays revealed that the Brachyury T-box domain bound to Mixl1-DNA complexes. Furthermore, luciferase reporter experiments indicated that association of Mixl1 with Brachyury and related T-box factors inhibited the transactivating potential of Mixl1 on the Gsc and Pdgfrα promoters. Our results indicate that the activity of Mixl1 can be modulated by protein-protein interactions and that T-box factors can function as negative regulators of Mixl1 activity.
- Published
- 2011
- Full Text
- View/download PDF
4. Safety and side effect profile of Pfizer-BioNTech COVID-19 vaccination among healthcare workers: A tertiary hospital experience in Singapore
- Author
-
Swee Chye Quek, Amelia Santosa, Jyoti Somani, E. H. C. Liu, Hwang Ching Chan, and Sue Mei Lim
- Subjects
Singapore ,Allergy ,medicine.medical_specialty ,COVID-19 Vaccines ,Side effect ,SARS-CoV-2 ,Cross-sectional study ,business.industry ,Health Personnel ,Vaccination ,COVID-19 ,General Medicine ,Odds ratio ,medicine.disease ,Occupational safety and health ,Tertiary Care Centers ,Cross-Sectional Studies ,Emergency medicine ,medicine ,Humans ,Chills ,medicine.symptom ,Adverse effect ,business - Abstract
Introduction: Vaccination remains a key strategy to living endemically with COVID-19. The Pfizer-BioNTech COVID-19 vaccine was first granted interim authorisation for use in Singapore in December 2020. With overseas studies published about the safety and side effect profiles of mRNA COVID-19 vaccines focusing mainly on non-Asian populations, we described the side effects of Pfizer-BioNTech COVID-19 vaccination experienced by the healthcare workers (HCWs) in a tertiary hospital in Singapore. Methods: Data were obtained from the Occupational Health Clinic (OHC) at the National University Hospital in Singapore, which monitored staff for any adverse effects within 30 minutes postvaccination on-site and any adverse effects after that. A cross-sectional study among the vaccinated HCWs was conducted using an online survey, which established basic demographics, histories of allergies or atopic disorders, and adverse events encountered after dose 1 and dose 2 of vaccination. Results: No anaphylaxis was reported. Most common symptom was giddiness (32.7%) experienced by HCWs within 30 minutes. Adverse events attended post-vaccination by OHC were generally mild and self-limiting. From the survey, odds of experiencing an adverse event after dose 2 was significantly higher than after the first dose, especially for fever/chills (odds ratio [OR] 22.5). Fever/chills, injection site reactions, headache, aches and pains, and feeling unwell were significantly more common in HCWs below 60 years compared to those ≥60 years. An allergy to food (adjusted OR 2.7) and a history of eczema/sensitive skin (adjusted OR 2.6) were associated with a skin reaction not at injection site. Conclusion: The side effects experienced after Pfizer-BioNTech COVID-19 vaccines are generally self-limiting and mild, with no anaphylaxis reported. Keywords: COVID-19, infectious diseases, occupational medicine, side effects, vaccination
- Published
- 2021
5. Modelling human blastocysts by reprogramming fibroblasts into iBlastoids
- Author
-
Amander T. Clark, Jan Schröder, Daniel Poppe, Joseph Chen, Yu Bo Yang Sun, Ryan Lister, Sue Mei Lim, Jose M. Polo, Jia Ping Tan, Xiaodong Liu, Monika Mohenska, Jennifer Zenker, Owen J. L. Rackham, Guizhi Sun, Yichen Zhou, John F. Ouyang, and Asma Aberkane
- Subjects
0303 health sciences ,Multidisciplinary ,Embryogenesis ,Trophoblast ,Embryo ,Gene mutation ,Biology ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Epiblast ,embryonic structures ,medicine ,Blastocyst ,Stem cell ,Reprogramming ,reproductive and urinary physiology ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
Human pluripotent and trophoblast stem cells have been essential alternatives to blastocysts for understanding early human development1–4. However, these simple culture systems lack the complexity to adequately model the spatiotemporal cellular and molecular dynamics that occur during early embryonic development. Here we describe the reprogramming of fibroblasts into in vitro three-dimensional models of the human blastocyst, termed iBlastoids. Characterization of iBlastoids shows that they model the overall architecture of blastocysts, presenting an inner cell mass-like structure, with epiblast- and primitive endoderm-like cells, a blastocoel-like cavity and a trophectoderm-like outer layer of cells. Single-cell transcriptomics further confirmed the presence of epiblast-, primitive endoderm-, and trophectoderm-like cells. Moreover, iBlastoids can give rise to pluripotent and trophoblast stem cells and are capable of modelling, in vitro, several aspects of the early stage of implantation. In summary, we have developed a scalable and tractable system to model human blastocyst biology; we envision that this will facilitate the study of early human development and the effects of gene mutations and toxins during early embryogenesis, as well as aiding in the development of new therapies associated with in vitro fertilization. Human fibroblasts are reprogrammed to generate blastocyst-like structures called iBlastoids, which recapitulate aspects of embryo implantation.
- Published
- 2021
6. The nuclear transporter importin 13 is critical for cell survival during embryonic stem cell differentiation
- Author
-
Shadma Fatima, Kylie M. Wagstaff, Julia C. Young, Sue Mei Lim, David A. Jans, and Jose M. Polo
- Subjects
0301 basic medicine ,Brachyury ,Mesoderm ,Cell Survival ,Biophysics ,Apoptosis ,Ectoderm ,Importin ,Karyopherins ,Biology ,Biochemistry ,Gene Knockout Techniques ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Proliferation Marker ,Molecular Biology ,Embryoid Bodies ,Embryonic Stem Cells ,Cell Proliferation ,Cell Differentiation ,Cell Biology ,Embryonic stem cell ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,embryonic structures ,Nuclear transport ,Endoderm ,Octamer Transcription Factor-3 - Abstract
Nuclear transporter Importin (Imp, Ipo) 13 is known to transport various mammalian cargoes into/out of the nucleus, but its role in directing cell-fate is unclear. Here we examine the role of Imp13 in the maintenance of pluripotency and differentiation of embryonic stem cells (ESCs) for the first time, using an embryonic body (EB)-based model. When induced to differentiate, Ipo13-/- ESCs displayed slow proliferation, reduced EB size, and lower expression of the proliferation marker KI67, concomitant with an increase in the number of TUNEL+ nuclei compared to wildtype ESCs. At days 5 and 10 of differentiation, Ipo13-/- EBs also showed enhanced loss of the pluripotency transcript OCT3/4, and barely detectable clusters of OCT3/4 positive cells. Day 5 Ipo13-/- EBs further exhibited reduced levels of the mesodermal markers Brachyury and Mixl1, correlating with reduced numbers of haemoglobinised cells generated. Our findings suggest that Imp13 is critical to ESC survival as well as early post-gastrulation differentiation.
- Published
- 2021
7. TINC— A Method to Dissect Regulatory Complexes at Single-Locus Resolution— Reveals an Extensive Protein Complex at the Nanog Promoter
- Author
-
Cheng Huang, Jaber Firas, Partha Pratim Das, Yu Bo Yang Sun, Anja S Knaupp, Joseph Chen, Melissa L. Holmes, Christian M. Nefzger, Fernando J. Rossello, Pratibha Tripathi, Trung V. Nguyen, Ralf B. Schittenhelm, Sue Mei Lim, Kayla Wong, Xiaodong Liu, Monika Mohenska, Jody J. Haigh, Jan Schröder, Ryan Lister, Jahnvi Pflueger, Kathryn C. Davidson, Ethan Ford, Jose M. Polo, and Michael R. Larcombe
- Subjects
0301 basic medicine ,Homeobox protein NANOG ,Human Embryonic Stem Cells ,iPSCs ,TINC ,Biology ,Nanog ,Biochemistry ,Article ,single-locus pull-down ,03 medical and health sciences ,0302 clinical medicine ,Genetics ,Humans ,Epigenetics ,Induced pluripotent stem cell ,Gene ,Transcription factor ,RCOR2 ,reprogramming ,Nanog Homeobox Protein ,Cell Biology ,Epigenome ,pluripotency ,3. Good health ,Cell biology ,030104 developmental biology ,Genetic Loci ,Multiprotein Complexes ,transcriptional complex ,Co-Repressor Proteins ,Reprogramming ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Summary Cellular identity is ultimately dictated by the interaction of transcription factors with regulatory elements (REs) to control gene expression. Advances in epigenome profiling techniques have significantly increased our understanding of cell-specific utilization of REs. However, it remains difficult to dissect the majority of factors that interact with these REs due to the lack of appropriate techniques. Therefore, we developed TINC: TALE-mediated isolation of nuclear chromatin. Using this new method, we interrogated the protein complex formed at the Nanog promoter in embryonic stem cells (ESCs) and identified many known and previously unknown interactors, including RCOR2. Further interrogation of the role of RCOR2 in ESCs revealed its involvement in the repression of lineage genes and the fine-tuning of pluripotency genes. Consequently, using the Nanog promoter as a paradigm, we demonstrated the power of TINC to provide insight into the molecular makeup of specific transcriptional complexes at individual REs as well as into cellular identity control in general., Graphical Abstract, Highlights • TINC allows the isolation of a specific locus for molecular analyses • TINC identified hundreds of proteins at the Nanog promoter • RCOR2 is a component of the pluripotency network in embryonic stem cells • RCOR2 is required for efficient differentiation, Here, Knaupp and colleagues describe TINC, an epigenetic method that allows interrogation of mammalian regulatory complexes at a single-locus resolution. TINC was applied to dissect the transcriptional complex at the Nanog promoter in embryonic stem cells, revealing hundreds of interactors, including RCOR2, hence redefining how this gene is regulated in pluripotency.
- Published
- 2020
8. An Efficient CRISPR-Cas9 DNA Editing Methodology Applicable for iPSC Disease Modelling
- Author
-
Atefeh Namipashaki, Xiaodong Liu, Kealan Pugsley, Sue Mei Lim, Guizhi Sun, Marco J. Herold, Jose M. Polo, Mark A. Bellgrove, and Ziarih Hawi
- Abstract
The capability to generate induced pluripotent cell (iPSC) lines, in combination with the CRISPR-Cas9 DNA editing technology, offers great promise to understand the underlying genetic mechanisms of human disease. However, technical impediments including, but not limited to, low transfection efficiency, single-cell survival, and high clonal heterogeneity, limit the potential of these techniques. Here we provide an efficient methodology addressing these challenges, resulting in high transfection efficiency exceeding 97% with an increased single cell clone survival rate (up to 70%). These enhancements were accompanied by a high editing efficiency in the range of 48.6 to 57.5%, comparable to existing methods.
- Published
- 2022
9. Modelling human blastocysts by reprogramming fibroblasts into iBlastoids
- Author
-
Xiaodong, Liu, Jia Ping, Tan, Jan, Schröder, Asma, Aberkane, John F, Ouyang, Monika, Mohenska, Sue Mei, Lim, Yu B Y, Sun, Joseph, Chen, Guizhi, Sun, Yichen, Zhou, Daniel, Poppe, Ryan, Lister, Amander T, Clark, Owen J L, Rackham, Jennifer, Zenker, and Jose M, Polo
- Subjects
Pluripotency ,Stem Cells ,Cell Culture Techniques ,Fibroblasts ,In Vitro Techniques ,Cellular Reprogramming ,Models, Biological ,Research Highlight ,Trophoblasts ,Blastocyst ,Humans ,Female ,Single-Cell Analysis ,Transcriptome - Abstract
Human pluripotent and trophoblast stem cells have been essential alternatives to blastocysts for understanding early human development
- Published
- 2020
10. DNA Hypermethylation Encroachment at CpG Island Borders in Cancer Is Predisposed by H3K4 Monomethylation Patterns
- Author
-
Lisa A. Miosge, Grady C. Smith, Jenny Z. Song, Amanda Khoury, Shalima S. Nair, Sue Mei Lim, Ozren Bogdanovic, Clare Stirzaker, Qian Du, Ruth Pidsley, Tim J Peters, Christopher C. Goodnow, Etienne Masle-Farquhar, Ksenia Skvortsova, James G. Kench, Elena Zotenko, Wenjia Qu, Susan J. Clark, Phuc-Loi Luu, Mark A. Rubin, Cathryn M. Gould, Joanne H. Reed, Lisa G. Horvath, Jose M. Polo, and Yolanda Colino-Sanguino
- Subjects
0301 basic medicine ,Cpg island methylation ,Male ,Cancer Research ,CpG island hypermethylation ,Dna hypermethylation ,Biology ,Methylation ,Histones ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Promoter Regions, Genetic ,610 Medicine & health ,Gene ,Mice, Knockout ,Cancer ,Cell Biology ,DNA, Neoplasm ,Histone-Lysine N-Methyltransferase ,DNA Methylation ,medicine.disease ,Neoplasm Proteins ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,030104 developmental biology ,Oncology ,CpG site ,030220 oncology & carcinogenesis ,DNA methylation ,Cancer research ,5-Methylcytosine ,CpG Islands ,Female ,Myeloid-Lymphoid Leukemia Protein - Abstract
Promoter CpG islands are typically unmethylated in normal cells, but in cancer a proportion are subject to hypermethylation. Using methylome sequencing we identified CpG islands that display partial methylation encroachment across the 5' or 3' CpG island borders. CpG island methylation encroachment is widespread in prostate and breast cancer and commonly associates with gene suppression. We show that the pattern of H3K4me1 at CpG island borders in normal cells predicts the different modes of cancer CpG island hypermethylation. Notably, genetic manipulation of Kmt2d results in concordant alterations in H3K4me1 levels and CpG island border DNA methylation encroachment. Our findings suggest a role for H3K4me1 in the demarcation of CpG island methylation borders in normal cells, which become eroded in cancer.
- Published
- 2019
- Full Text
- View/download PDF
11. An improved reprogrammable mouse model harbouring the reverse tetracycline-controlled transcriptional transactivator 3
- Author
-
Sara Alaei, Minna-Liisa Änkö, Melissa L. Holmes, Anja S Knaupp, Jose M. Polo, Sue Mei Lim, Christian M. Nefzger, and Joseph Chen
- Subjects
Transcriptional Activation ,0301 basic medicine ,Octamer Transcription Factor-3 ,Cellular differentiation ,Induced Pluripotent Stem Cells ,Kruppel-Like Transcription Factors ,Biology ,Proto-Oncogene Proteins c-myc ,Kruppel-Like Factor 4 ,Mice ,03 medical and health sciences ,Transactivation ,Plasmid ,Animals ,Induced pluripotent stem cell ,Gene ,lcsh:QH301-705.5 ,Cells, Cultured ,Genetics ,Medicine(all) ,SOXB1 Transcription Factors ,Teratoma ,Cell Differentiation ,General Medicine ,Cell Biology ,Fibroblasts ,Cellular Reprogramming ,Cell biology ,030104 developmental biology ,lcsh:Biology (General) ,Tetracyclines ,Reprogramming ,Plasmids ,Developmental Biology - Abstract
Reprogrammable mouse models engineered to conditionally express Oct-4, Klf-4, Sox-2 and c-Myc (OKSM) have been instrumental in dissecting molecular events underpinning the generation of induced pluripotent stem cells. However, until now these models have been reported in the context of the m2 reverse tetracycline-controlled transactivator, which results in low reprogramming efficiency and consequently limits the number of reprogramming intermediates that can be isolated for downstream profiling. Here, we describe an improved OKSM mouse model in the context of the reverse tetracycline-controlled transactivator 3 with enhanced reprogramming efficiency (>9-fold) and increased numbers of reprogramming intermediate cells albeit with similar kinetics, which we believe will facilitate mechanistic studies of the reprogramming process.
- Published
- 2016
- Full Text
- View/download PDF
12. Cell Type of Origin Dictates the Route to Pluripotency
- Author
-
Fernando J. Rossello, Xiaodong Liu, Christian M. Nefzger, Joseph Chen, Owen J. L. Rackham, Sue Mei Lim, Jose M. Polo, Anja S Knaupp, Ryan Lister, Jaber Firas, Mirana Ramialison, Jacob M. Paynter, Sara Alaei, Sam Buckberry, Keshav Faye-Chauhan, Brenda Williams, Enrico Petretto, David R. Powel, Jahnvi Pflueger, and Susan K. Nilsson
- Subjects
Cell type ,Mesenchymal stem cell ,Cell type specific ,Reversion ,Cell generation ,Biology ,Induced pluripotent stem cell ,Reprogramming ,Cell identity ,Cell biology - Abstract
Our current understanding of induced pluripotent stem (iPS) cell generation has almost entirely been shaped by studies performed on reprogramming fibroblasts. However, whether or not the resulting model universally applies to the reprogramming process of other cell types is still largely unknown. By functionally and molecularly characterizing the reprogramming pathways of fibroblasts, neutrophils and keratinocytes, we unveil that while some aspects of the reprogramming process are conserved, the key events, including loss of original cell identity, mesenchymal to epithelial transition, the extent of developmental reversion and reactivation of the pluripotency network are to a large degree cell type specific. Thus we reveal universal and cell type specific features of the reprogramming process.
- Published
- 2018
13. Transcription factor‐mediated reprogramming: epigenetics and therapeutic potential
- Author
-
E Xiaodong Liu, Sue Mei Lim, Jose M. Polo, and Jaber Firas
- Subjects
Genetics ,Cell type ,Cell fusion ,Induced Pluripotent Stem Cells ,Immunology ,Cell Biology ,Biology ,Cellular Reprogramming ,Regenerative Medicine ,Regenerative medicine ,Epigenesis, Genetic ,Cell biology ,Biological Therapy ,Cell Transdifferentiation ,Animals ,Humans ,Immunology and Allergy ,Somatic cell nuclear transfer ,Epigenetics ,Transcription factor ,Reprogramming ,Transcription Factors ,Epigenesis - Abstract
Cellular reprogramming refers to the conversion of one cell type into another by altering its epigenetic marks. This can be achieved by three different methods: somatic cell nuclear transfer, cell fusion and transcription factor (TF)-mediated reprogramming. TF-mediated reprogramming can occur through several means, either reverting backwards to a pluripotent state before redifferentiating to a new cell type (otherwise known as induced pluripotency), by transdifferentiating directly into a new cell type (bypassing the intermediate pluripotent stage), or, by using the induced pluripotency pathway without reaching the pluripotent state. The possibility of reprogramming any cell type of interest not only sheds new insights on cellular plasticity, but also provides a novel use of this technology across several platforms, most notably in cellular replacement therapies, disease modelling and drug screening. This review will focus on the different ways of implementing TF-mediated reprogramming, their associated epigenetic changes and its therapeutic potential.
- Published
- 2015
14. Cell type of origin dictates the route to pluripotency
- Author
-
Sue Mei Lim, Keshav Faye-Chauhan, Christian M. Nefzger, Joseph Chen, Jaber Firas, Anja S Knaupp, Sam Buckberry, Jacob M. Paynter, Owen J. L. Rackham, Ryan Lister, Brenda Williams, Enrico Petretto, Xiaodong Liu, Jose M. Polo, Fernando J. Rossello, Sara Alaei, Mirana Ramialison, David R. Powell, Jahnvi Pflueger, and Susan K. Nilsson
- Subjects
0301 basic medicine ,EXPRESSION ,keratinocytes ,Cell type ,induced pluripotent stem cells ,Cell ,EGR1 ,Biology ,MOUSE ,Universal model ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,IPS CELLS ,MARKERS ,neutrophils ,fibroblasts ,medicine ,Animals ,Humans ,HEMATOPOIETIC STEM-CELLS ,Induced pluripotent stem cell ,lcsh:QH301-705.5 ,Early Growth Response Protein 1 ,transcriptional dynamics ,Science & Technology ,Mesenchymal stem cell ,reprogramming ,Cell Biology ,Cellular Reprogramming ,Flow Cytometry ,MASS CYTOMETRY ,Cell identity ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Biology (General) ,SOMATIC-CELLS ,Egr1 ,MOLECULAR ROADMAP ,Octamer Transcription Factor-3 ,Reprogramming ,Life Sciences & Biomedicine - Abstract
Summary: Our current understanding of induced pluripotent stem cell (iPSC) generation has almost entirely been shaped by studies performed on reprogramming fibroblasts. However, whether the resulting model universally applies to the reprogramming process of other cell types is still largely unknown. By characterizing and profiling the reprogramming pathways of fibroblasts, neutrophils, and keratinocytes, we unveil that key events of the process, including loss of original cell identity, mesenchymal to epithelial transition, the extent of developmental reversion, and reactivation of the pluripotency network, are to a large degree cell-type specific. Thus, we reveal limitations for the use of fibroblasts as a universal model for the study of the reprogramming process and provide crucial insights about iPSC generation from alternative cell sources. : Nefzger et al. find that the molecular reprogramming trajectories of fibroblasts, neutrophils, and keratinocytes have a cell-type-specific component that only fully converges in induced pluripotent stem cells. The authors also identify universal changes shared by all three cell types, including two transcriptional waves and a conserved transcriptional program involving Egr1 downregulation. Keywords: reprogramming, induced pluripotent stem cells, fibroblasts, neutrophils, keratinocytes, transcriptional dynamics, Egr1
- Published
- 2017
15. A Molecular Roadmap of Reprogramming Somatic Cells into iPS Cells
- Author
-
Ori Bar-Nur, Endre Anderssen, Effie Apostolou, Sue Mei Lim, Sridhar Ramaswamy, Jinfang Zhu, Ari Melnick, Sihem Cheloufi, Daisy A. Robinton, Konrad Hochedlinger, Sara Alaei, Christian M. Nefzger, Marti Borkent, Matthias Stadtfeld, Jose M. Polo, Sridaran Natesan, Maria E. Figueroa, Ryan M. Walsh, Benjamin A. Schwarz, and Jennifer Cloutier
- Subjects
Genetics ,0303 health sciences ,Biochemistry, Genetics and Molecular Biology(all) ,Somatic cell ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,03 medical and health sciences ,0302 clinical medicine ,SOX2 ,KLF4 ,DNA methylation ,Induced pluripotent stem cell ,Reprogramming ,Gene ,Transcription factor ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
SummaryFactor-induced reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) is inefficient, complicating mechanistic studies. Here, we examined defined intermediate cell populations poised to becoming iPSCs by genome-wide analyses. We show that induced pluripotency elicits two transcriptional waves, which are driven by c-Myc/Klf4 (first wave) and Oct4/Sox2/Klf4 (second wave). Cells that become refractory to reprogramming activate the first but fail to initiate the second transcriptional wave and can be rescued by elevated expression of all four factors. The establishment of bivalent domains occurs gradually after the first wave, whereas changes in DNA methylation take place after the second wave when cells acquire stable pluripotency. This integrative analysis allowed us to identify genes that act as roadblocks during reprogramming and surface markers that further enrich for cells prone to forming iPSCs. Collectively, our data offer new mechanistic insights into the nature and sequence of molecular events inherent to cellular reprogramming.
- Published
- 2012
16. Fine Tuning of Canonical Wnt Stimulation Enhances Differentiation of Pluripotent Stem Cells Independent of β-Catenin-Mediated T-Cell Factor Signaling
- Author
-
Yu Bo Yang Sun, Christian M. Nefzger, Tiziano Barberi, Xiaodong Liu, Suzan de Boer, Sue Mei Lim, Joseph Chen, Jose M. Polo, Anja S Knaupp, Jinhua Li, Kathryn C. Davidson, and Fernando J. Rossello
- Subjects
0301 basic medicine ,Pluripotent Stem Cells ,Cell signaling ,Wnt signaling pathway ,Cell Differentiation ,Cell Biology ,Biology ,Embryonic stem cell ,Cell biology ,03 medical and health sciences ,Mice ,030104 developmental biology ,Catenin ,Molecular Medicine ,Animals ,Humans ,Ectopic expression ,Stem cell ,Induced pluripotent stem cell ,Wnt Signaling Pathway ,Tissue homeostasis ,beta Catenin ,Developmental Biology ,Signal Transduction - Abstract
The canonical Wnt/β-catenin pathway is crucial for early embryonic patterning, tissue homeostasis, and regeneration. While canonical Wnt/β-catenin stimulation has been used extensively to modulate pluripotency and differentiation of pluripotent stem cells (PSCs), the mechanism of these two seemingly opposing roles has not been fully characterized and is currently largely attributed to activation of nuclear Wnt target genes. Here, we show that low levels of Wnt stimulation via ectopic expression of Wnt1 or administration of glycogen synthase kinase-3 inhibitor CHIR99021 significantly increases PSC differentiation into neurons, cardiomyocytes and early endodermal intermediates. Our data indicate that enhanced differentiation outcomes are not mediated through activation of traditional Wnt target genes but by β-catenin's secondary role as a binding partner of membrane bound cadherins ultimately leading to the activation of developmental genes. In summary, fine-tuning of Wnt signaling to subthreshold levels for detectable nuclear β-catenin function appears to act as a switch to enhance differentiation of PSCs into multiple lineages. Our observations highlight a mechanism by which Wnt/β-catenin signaling can achieve dosage dependent dual roles in regulating self-renewal and differentiation.
- Published
- 2016
17. Temporal Restriction of Pancreatic Branching Competence During Embryogenesis Is Mirrored In Differentiating Embryonic Stem Cells
- Author
-
Xueling Li, Andrew M. Holland, Edouard G. Stanley, Andrew G. Elefanty, Jacqueline V. Schiesser, Suzanne Jeanine Micallef, and Sue Mei Lim
- Subjects
Male ,Organogenesis ,Recombinant Fusion Proteins ,Embryonic Development ,Mice, Transgenic ,Embryoid body ,Biology ,Real-Time Polymerase Chain Reaction ,Green fluorescent protein ,Tissue Culture Techniques ,Mice ,medicine ,Animals ,Pancreas ,Cells, Cultured ,Embryoid Bodies ,Embryonic Stem Cells ,Homeodomain Proteins ,Matrigel ,Gene Expression Profiling ,Endoderm ,Embryogenesis ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Embryo ,Cell Biology ,Hematology ,Anatomy ,Embryonic stem cell ,Coculture Techniques ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Microscopy, Fluorescence ,Mouse Pancreas ,embryonic structures ,Trans-Activators ,Female ,Developmental Biology - Abstract
To develop methods for the generation of insulin-producing β-cells for the treatment of diabetes, we have used GFP-tagged embryonic stem cells (ESCs) to elucidate the process of pancreas development. Using the reporter Pdx1(GFP/w) ESC line, we have previously described a serum-free differentiation protocol in which Pdx1-GFP(+) cells formed GFP bright (GFP(br)) epithelial buds that resembled those present in the developing mouse pancreas. In this study we extend these findings to demonstrate that these cells can undergo a process of branching morphogenesis, similar to that seen during pancreatic development of the mid-gestation embryo. These partially disaggregated embryoid bodies containing GFP(br) buds initially form epithelial ring-like structures when cultured in Matrigel. After several days in culture, these rings undergo a process of proliferation and form a ramified network of epithelial branches. Comparative analysis of explanted dissociated pancreatic buds from E13.5 Pdx1(GFP/w) embryos and ESC-derived GFP(br) buds reveal a similar process of proliferation and branching, with both embryonic Pdx1(GFP/w) branching pancreatic epithelium and ESC-derived GFP(br) branching organoids expressing markers representing epithelial (EpCAM and E-Cadherin), ductal (Mucin1), exocrine (Amylase and Carboxypeptidase 1A), and endocrine cell types (Glucagon and Somatostatin). ESC-derived branching structures also expressed a suite of genes indicative of ongoing pancreatic differentiation, paralleling gene expression within similar structures derived from the E13.5 fetal pancreas. In summary, differentiating mouse ESCs can generate pancreatic material that has significant similarity to the fetal pancreatic anlagen, providing an in vitro platform for investigating the cellular and molecular mechanisms underpinning pancreatic development.
- Published
- 2012
18. Transient and Permanent Reconfiguration of Chromatin and Transcription Factor Occupancy Drive Reprogramming
- Author
-
Sam Buckberry, Sara Alaei, Susan J. Clark, Alex de Mendoza, Sue Mei Lim, Fernando J. Rossello, Christian M. Nefzger, Jaber Firas, Jahnvi Pflueger, Ryan Lister, Melissa L. Holmes, Anja S Knaupp, Ethan Ford, Michael R. Larcombe, Shalima S. Nair, and Jose M. Polo
- Subjects
0301 basic medicine ,Cell Biology ,Biology ,Embryonic stem cell ,Chromatin remodeling ,Chromatin ,Cell biology ,ChIP-sequencing ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,SOX2 ,Genetics ,Molecular Medicine ,sense organs ,Induced pluripotent stem cell ,Transcription factor ,Reprogramming ,030217 neurology & neurosurgery - Abstract
Somatic cell reprogramming into induced pluripotent stem cells (iPSCs) induces changes in genome architecture reflective of the embryonic stem cell (ESC) state. However, only a small minority of cells typically transition to pluripotency, which has limited our understanding of the process. Here, we characterize the DNA regulatory landscape during reprogramming by time-course profiling of isolated sub-populations of intermediates poised to become iPSCs. Widespread reconfiguration of chromatin states and transcription factor (TF) occupancy occurs early during reprogramming, and cells that fail to reprogram partially retain their original chromatin states. A second wave of reconfiguration occurs just prior to pluripotency acquisition, where a majority of early changes revert to the somatic cell state and many of the changes that define the pluripotent state become established. Our comprehensive characterization of reprogramming-associated molecular changes broadens our understanding of this process and sheds light on how TFs access and change the chromatin during cell-fate transitions.
- Published
- 2017
19. Enforced Expression of Mixl1 During Mouse ES Cell Differentiation Suppresses Hematopoietic Mesoderm and Promotes Endoderm Formation
- Author
-
Sue Mei Lim, Alan O Trounson, Lloyd Ashley Pereira, Benjamin Eugene Van Vranken, Michael S. Wong, Andrew G. Elefanty, Edouard G. Stanley, Claire E Hirst, and Marjorie Pick
- Subjects
Mesoderm ,BALB 3T3 Cells ,animal structures ,Blotting, Western ,Electrophoretic Mobility Shift Assay ,Germ layer ,Embryoid body ,Biology ,FGF and mesoderm formation ,Cell Line ,Mice ,Endoderm formation ,HMGB Proteins ,SOXF Transcription Factors ,medicine ,Animals ,Embryonic Stem Cells ,Homeodomain Proteins ,Endoderm ,Cell Differentiation ,Cell Biology ,Anatomy ,Cadherins ,Flow Cytometry ,Immunohistochemistry ,Cell biology ,medicine.anatomical_structure ,embryonic structures ,Hepatocyte Nuclear Factor 3-beta ,Molecular Medicine ,NODAL ,Developmental Biology ,Definitive endoderm - Abstract
The Mixl1 gene encodes a homeodomain transcription factor that is required for normal mesoderm and endoderm development in the mouse. We have examined the consequences of enforced Mixl1 expression during mouse embryonic stem cell (ESC) differentiation. We show that three independently derived ESC lines constitutively expressing Mixl1 (Mixl1C ESCs) differentiate into embryoid bodies (EBs) containing a higher proportion of E-cadherin (E-Cad)+ cells. Our analysis also shows that this differentiation occurs at the expense of hematopoietic mesoderm differentiation, with Mixl1C ESCs expressing only low levels of Flk1 and failing to develop hemoglobinized cells. Immunohistochemistry and immunofluorescence studies revealed that Mixl1C EBs have extensive areas containing cells with an epithelial morphology that express E-Cad, FoxA2, and Sox17, consistent with enhanced endoderm formation. Luciferase reporter transfection experiments indicate that Mixl1 can transactivate the Gsc, Sox17, and E-Cad promoters, supporting the hypothesis that Mixl1 has a direct role in definitive endoderm formation. Taken together, these studies suggest that high levels of Mixl1 preferentially allocate cells to the endoderm during ESC differentiation.
- Published
- 2009
20. A method for genetic modification of human embryonic stem cells using electroporation
- Author
-
Mirella Dottori, Pegah Jamshidi, Koula Sourris, Tanya Hatzistavrou, Steven Anthony Jackson, Sue Mei Lim, Lisa Azzola, Edouard G. Stanley, Martin F. Pera, Magdaline Costa, Andrew G. Elefanty, and Richard P. Davis
- Subjects
Electroporation ,Systems biology ,Genetic Vectors ,Gene targeting ,Computational biology ,Transfection ,Biology ,Embryonic stem cell ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Transduction (genetics) ,Gene Targeting ,embryonic structures ,Humans ,DNA microarray ,Genetic Engineering ,Developmental biology ,Embryonic Stem Cells - Abstract
The ability to genetically modify human embryonic stem cells (HESCs) will be critical for their widespread use as a tool for understanding fundamental aspects of human biology and pathology and for their development as a platform for pharmaceutical discovery. Here, we describe a method for the genetic modification of HESCs using electroporation, the preferred method for introduction of DNA into cells in which the desired outcome is gene targeting. This report provides methods for cell amplification, electroporation, colony selection and screening. The protocol we describe has been tested on four different HESC lines, and takes approximately 4 weeks from electroporation to PCR screening of G418-resistant clones.
- Published
- 2007
21. Productive Infection of Human Embryonic Stem Cell-Derived NKX2.1+ Respiratory Progenitors with Human Rhinovirus
- Author
-
Edouard G. Stanley, Tanya Labonne, Anthony Kicic, Elizabeth Ng, Andrew G. Elefanty, Suzanne Jeanine Micallef, Sue Mei Lim, Stephen M. Stick, Alan O Trounson, Claire E Hirst, Adam L Goulburn, Kak-Ming Ling, Antonietta Giudice, and Robert Alexander Jenny
- Subjects
Rhinovirus ,Cellular differentiation ,Thyroid Nuclear Factor 1 ,Mice, Nude ,Respiratory Mucosa ,Biology ,Cell Line ,Mice ,Directed differentiation ,medicine ,Animals ,Humans ,Progenitor cell ,Induced pluripotent stem cell ,Embryonic Stem Cells ,Epithelial cell differentiation ,Picornaviridae Infections ,Nuclear Proteins ,Cell Differentiation ,Cell Biology ,General Medicine ,respiratory system ,Cell-Based Drug Development, Screening, and Toxicology ,Embryonic stem cell ,Cell biology ,medicine.anatomical_structure ,Immunology ,Host-Pathogen Interactions ,Endoderm ,Developmental Biology ,Human embryonic stem cell line ,Transcription Factors - Abstract
Airway epithelial cells generated from pluripotent stem cells (PSCs) represent a resource for research into a variety of human respiratory conditions, including those resulting from infection with common human pathogens. Using an NKX2.1-GFP reporter human embryonic stem cell line, we developed a serum-free protocol for the generation of NKX2.1+ endoderm that, when transplanted into immunodeficient mice, matured into respiratory cell types identified by expression of CC10, MUC5AC, and surfactant proteins. Gene profiling experiments indicated that day 10 NKX2.1+ endoderm expressed markers indicative of early foregut but lacked genes associated with later stages of respiratory epithelial cell differentiation. Nevertheless, NKX2.1+ endoderm supported the infection and replication of the common respiratory pathogen human rhinovirus HRV1b. Moreover, NKX2.1+ endoderm upregulated expression of IL-6, IL-8, and IL-1B in response to infection, a characteristic of human airway epithelial cells. Our experiments provide proof of principle for the use of PSC-derived respiratory epithelial cells in the study of cell-virus interactions. Significance This report provides proof-of-principle experiments demonstrating, for the first time, that human respiratory progenitor cells derived from stem cells in the laboratory can be productively infected with human rhinovirus, the predominant cause of the common cold.
- Published
- 2014
22. Cell Type of Origin Dictates the Route to Pluripotency.
- Author
-
Nefzger, Christian M., Rossello, Fernando J., Joseph Chen, Xiaodong Liu, Knaupp, Anja S., Firas, Jaber, Paynter, Jacob M., Pflueger, Jahnvi, Buckberry, Sam, Sue Mei Lim, Williams, Brenda, Alaei, Sara, Faye-Chauhan, Keshav, Petretto, Enrico, Nilsson, Susan K., Lister, Ryan, Ramialison, Mirana, Powell, David R., Rackham, Owen J. L., and Polo, Jose M.
- Abstract
Our current understanding of induced pluripotent stem cell (iPSC) generation has almost entirely been shaped by studies performed on reprogramming fibroblasts. However, whether the resulting model universally applies to the reprogramming process of other cell types is still largely unknown. By characterizing and profiling the reprogramming pathways of fibroblasts, neutrophils, and keratinocytes, we unveil that key events of the process, including loss of original cell identity, mesenchymal to epithelial transition, the extent of developmental reversion, and reactivation of the pluripotency network, are to a large degree cell-type specific. Thus, we reveal limitations for the use of fibroblasts as a universal model for the study of the reprogramming process and provide crucial insights about iPSC generation from alternative cell sources. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
23. Pdgfrα and Flk1 are direct target genes of Mixl1 in differentiating embryonic stem cells
- Author
-
Claire Elizabeth Hirst, Sue Mei Lim, John E. Pimanda, Anna K Mossman, Kathy Knezevic, Koula Sourris, Lloyd Pereira, Edouard G. Stanley, Michael S. Wong, Mary E. Janes, and Andrew G. Elefanty
- Subjects
Transcriptional Activation ,Receptor, Platelet-Derived Growth Factor alpha ,Molecular Sequence Data ,Embryoid body ,Biology ,Green fluorescent protein ,Cell Line ,Mesoderm ,Mice ,Gene expression ,Animals ,Amino Acid Sequence ,RNA, Messenger ,Promoter Regions, Genetic ,Embryonic Stem Cells ,Oligonucleotide Array Sequence Analysis ,Medicine(all) ,Homeodomain Proteins ,Binding Sites ,Base Sequence ,Gene Expression Profiling ,Endoderm ,Gene Expression Regulation, Developmental ,Promoter ,Cell Differentiation ,Cell Biology ,General Medicine ,Flow Cytometry ,Embryonic stem cell ,Molecular biology ,Vascular Endothelial Growth Factor Receptor-2 ,Homeobox ,MIXL1 ,Chromatin immunoprecipitation ,Developmental Biology ,Protein Binding - Abstract
The Mixl1 homeodomain protein plays a key role in mesendoderm patterning during embryogenesis, but its target genes remain to be identified. We compared gene expression in differentiating heterozygous Mixl1 GFP/w and homozygous null Mixl1 GFP/Hygro mouse embryonic stem cells to identify potential downstream transcriptional targets of Mixl1. Candidate Mixl1 regulated genes whose expression was reduced in GFP + cells isolated from differentiating Mixl1 GFP/Hygro embryoid bodies included Pdgfrα and Flk1 . Mixl1 bound to ATTA sequences located in the Pdgfrα and Flk1 promoters and chromatin immunoprecipitation assays confirmed Mixl1 occupancy of these promoters in vivo. Furthermore, Mixl1 transactivated the Pdgfrα and Flk1 promoters through ATTA sequences in a DNA binding dependent manner. These data support the hypothesis that Mixl1 directly regulates Pdgfrα and Flk1 gene expression and strengthens the position of Mixl1 as a key regulator of mesendoderm development during mammalian gastrulation.
- Published
- 2011
24. A targeted NKX2.1 human embryonic stem cell reporter line enables identification of human basal forebrain derivatives
- Author
-
Antonietta Giudice, Colin W. Pouton, Stewart A. Anderson, Tanya Hatzistavrou, Edouard G. Stanley, Richard P. Davis, Andrew G. Elefanty, Richard J Lang, Elizabeth Ng, Justin L Bourke, Chew-Li Soh, Qing Cissy Yu, Suzanne J. Micallef, Darym Alden, Sue Mei Lim, Bradley Watmuff, David A. Elliott, John M. Haynes, Adam L Goulburn, and Alan O Trounson
- Subjects
Cell type ,Neurogenesis ,Thyroid Nuclear Factor 1 ,Retinoic acid ,Mice, Transgenic ,Biology ,Green fluorescent protein ,OLIG2 ,chemistry.chemical_compound ,Mice ,Prosencephalon ,Genes, Reporter ,Animals ,Humans ,Cell Lineage ,Molecular Targeted Therapy ,Progenitor cell ,Cells, Cultured ,Embryonic Stem Cells ,Genetics ,Basal forebrain ,Nuclear Proteins ,Cell Differentiation ,Cell Biology ,Flow Cytometry ,Embryonic stem cell ,Cell biology ,chemistry ,Animals, Newborn ,Cell Tracking ,embryonic structures ,Forebrain ,Molecular Medicine ,Developmental Biology ,Transcription Factors - Abstract
We have used homologous recombination in human embryonic stem cells (hESCs) to insert sequences encoding green fluorescent protein (GFP) into the NKX2.1 locus, a gene required for normal development of the basal forebrain. Generation of NKX2.1-GFP+ cells was dependent on the concentration, timing, and duration of retinoic acid treatment during differentiation. NKX2.1-GFP+ progenitors expressed genes characteristic of the basal forebrain, including SHH, DLX1, LHX6, and OLIG2. Time course analysis revealed that NKX2.1-GFP+ cells could upregulate FOXG1 expression, implying the existence of a novel pathway for the generation of telencephalic neural derivatives. Further maturation of NKX2.1-GFP+ cells gave rise to γ-aminobutyric acid-, tyrosine hydroxylase-, and somatostatin-expressing neurons as well as to platelet-derived growth factor receptor α-positive oligodendrocyte precursors. These studies highlight the diversity of cell types that can be generated from human NKX2.1+ progenitors and demonstrate the utility of NKX2.1GFP/w hESCs for investigating human forebrain development and neuronal differentiation.
- Published
- 2011
25. NKX2-5 eGFP/w hESCs for isolation of human cardiac progenitors and cardiomyocytes
- Author
-
Andrew G. Elefanty, Christine Biben, Stefan R. Braam, Owen W.J. Prall, Dorien Ward-van Oostwaard, David A. Elliott, Xueling Li, Christine L. Mummery, Claire E Hirst, Richard P. Davis, Katerina Koutsis, Elizabeth Ng, Rhys J.P. Skelton, Colin W. Pouton, Sue Mei Lim, Qing C Yu, Adam L Goulburn, John M. Haynes, Susan Melanie Hawes, Ebba Louise Lagerqvist, Edouard G. Stanley, Robert Alexander Jenny, Tanya Hatzistavrou, David M. Kaye, Ouda Khammy, and Robert Passier
- Subjects
Cellular differentiation ,Green Fluorescent Proteins ,Vascular Cell Adhesion Molecule-1 ,Cell Separation ,Biology ,Polymerase Chain Reaction ,Biochemistry ,Green fluorescent protein ,Gene expression ,Humans ,Myocyte ,Myocytes, Cardiac ,Receptors, Immunologic ,Molecular Biology ,Transcription factor ,Embryonic Stem Cells ,reproductive and urinary physiology ,Homeodomain Proteins ,Gene Expression Profiling ,Cell Differentiation ,Cell Biology ,equipment and supplies ,Antigens, Differentiation ,Molecular biology ,Embryonic stem cell ,Gene expression profiling ,embryonic structures ,Homeobox Protein Nkx-2.5 ,cardiovascular system ,biological phenomena, cell phenomena, and immunity ,Stem cell ,Biomarkers ,Myoblasts, Cardiac ,Transcription Factors ,Biotechnology - Abstract
NKX2-5 is expressed in the heart throughout life. We targeted eGFP sequences to the NKX2-5 locus of human embryonic stem cells (hESCs); NKX2-5(eGFP/w) hESCs facilitate quantification of cardiac differentiation, purification of hESC-derived committed cardiac progenitor cells (hESC-CPCs) and cardiomyocytes (hESC-CMs) and the standardization of differentiation protocols. We used NKX2-5 eGFP(+) cells to identify VCAM1 and SIRPA as cell-surface markers expressed in cardiac lineages.
- Published
- 2011
26. Expression from a betageo gene trap in the Slain1 gene locus is predominantly associated with the developing nervous system
- Author
-
Lloyd Pereira, Claire E Hirst, Sue Mei Lim, Robyn Mayberry, Edouard G. Stanley, and Andrew G. Elefanty
- Subjects
Nervous system ,Male ,Embryology ,Time Factors ,Limb Buds ,Recombinant Fusion Proteins ,Molecular Sequence Data ,Biology ,Nervous System ,Mice ,Embryonic Structure ,Sequence Homology, Nucleic Acid ,medicine ,Animals ,Humans ,Regulation of gene expression ,Genetics ,Reporter gene ,Base Sequence ,Histocytochemistry ,Reverse Transcriptase Polymerase Chain Reaction ,Gene Expression Profiling ,Gene Expression Regulation, Developmental ,Proteins ,Embryo, Mammalian ,beta-Galactosidase ,Embryonic stem cell ,Cell biology ,Gene expression profiling ,Gastrointestinal Tract ,medicine.anatomical_structure ,Epiblast ,embryonic structures ,Developmental biology ,Developmental Biology - Abstract
Slain1 was originally identified as a novel stem cell-associated gene in transcriptional profiling experiments comparing mouse and human embryonic stem cells (ESCs) and their immediate differentiated progeny. In order to obtain further insight into the potential function of Slain1, we examined the expression of beta-galactosidase in a gene-trap mouse line in which a beta-geo reporter gene was inserted into the second intron of Slain1. In early stage embryos (E7.5), the Slain1-betageo fusion protein was expressed within the entire epiblast, but by E9.5 became restricted to the developing nervous system and gastrointestinal tract. In later stage embryos (E11.5 - E13.5), expression was predominantly within the developing nervous system. Lower level expression was also observed in the developing limb buds, in the condensing mesenchyme, along the apical epidermal ridge and, at later stages, within the digital zones. These observations suggest that Slain1 may play a role in the development of the nervous system, as well as in the morphogenesis of several embryonic structures.
- Published
- 2010
27. Brachyury and Related Tbx Proteins Interact with the Mixl1 Homeodomain Protein and Negatively Regulate Mixl1 Transcriptional Activity
- Author
-
Edouard G. Stanley, Sue Mei Lim, Michael S. Wong, Lloyd Pereira, Andrew G. Elefanty, and Alexandra Sides
- Subjects
Proteomics ,Fetal Proteins ,Transcription, Genetic ,Cellular differentiation ,lcsh:Medicine ,Mesodermal Cells ,Biochemistry ,Mice ,Molecular Cell Biology ,Protein Interaction Mapping ,Fluorescent Antibody Technique, Indirect ,Promoter Regions, Genetic ,lcsh:Science ,Oligonucleotide Array Sequence Analysis ,Genetics ,Multidisciplinary ,Stem Cells ,Cell Differentiation ,medicine.anatomical_structure ,Cellular Types ,Endoderm ,Research Article ,Brachyury ,DNA transcription ,Biology ,DNA-binding protein ,Cell Line, Tumor ,DNA-binding proteins ,medicine ,Animals ,Humans ,Transcription factor ,Embryonic Stem Cells ,Cell Nucleus ,Homeodomain Proteins ,lcsh:R ,Proteins ,Protein interactions ,Promoter ,Fibroblasts ,HEK293 Cells ,Gene Expression Regulation ,Homeobox ,lcsh:Q ,MIXL1 ,Gene expression ,T-Box Domain Proteins ,Developmental Biology - Abstract
Mixl1 is a homeodomain transcription factor required for mesoderm and endoderm patterning during mammalian embryogenesis. Despite its crucial function in development, co-factors that modulate the activity of Mixl1 remain poorly defined. Here we report that Mixl1 interacts physically and functionally with the T-box protein Brachyury and related members of the T-box family of transcription factors. Transcriptional and protein analyses demonstrated overlapping expression of Mixl1 and Brachyury during embryonic stem cell differentiation. In vitro protein interaction studies showed that the Mixl1 with Brachyury associated via their DNA-binding domains and gel shift assays revealed that the Brachyury T-box domain bound to Mixl1-DNA complexes. Furthermore, luciferase reporter experiments indicated that association of Mixl1 with Brachyury and related T-box factors inhibited the transactivating potential of Mixl1 on the Gsc and Pdgfrα promoters. Our results indicate that the activity of Mixl1 can be modulated by protein-protein interactions and that T-box factors can function as negative regulators of Mixl1 activity.
- Published
- 2011
28. Productive Infection of Human Embryonic Stem Cell-Derived NKX2.1+ Respiratory Progenitors With Human Rhinovirus.
- Author
-
JENNY, ROBERT A., HIRST, CLAIRE, SUE MEI LIM, GOULBURN, ADAM L., MICALLEF, SUZANNE J., LABONNE, TANYA, KICIC, ANTHONY, KAK-MING LING, STICK, STEPHEN M., NG, ELIZABETH S., TROUNSON, ALAN, GIUDICE, ANTONIETTA, ELEFANTY, ANDREW G., and STANLEY, EDOUARD G.
- Published
- 2015
- Full Text
- View/download PDF
29. Expression from a βgeo gene trap in the Slain1 gene locus is predominantly associated with the developing nervous system.
- Author
-
HIRST, CLAIRE E., SUE-MEI LIM, PEREIRA, LLOYD A., MAYBERRY, ROBYN A., STANLEY, EDOUARD G., and ELEFANTY, ANDREW G.
- Subjects
GENE expression ,LOCUS (Genetics) ,NERVOUS system development ,SENSORY ganglia ,EMBRYONIC stem cells ,GENETIC transcription ,LABORATORY mice ,EMBRYOLOGY - Published
- 2010
- Full Text
- View/download PDF
30. Derivation of endothelial cells from human embryonic stem cells in fully defined medium enables identification of lysophosphatidic acid and platelet activating factor as regulators of eNOS localization
- Author
-
Sue Mei Lim, Qing C Yu, Kathy Koutsis, Edouard G. Stanley, Helena C. Parkington, Hong B. Liu, Claire E Hirst, Suzanne J. Micallef, Vanta J. Jokubaitis, Anthony E. Dear, Andrew G. Elefanty, Koula Sourris, and Magdaline Costa
- Subjects
CD31 ,Nitric Oxide Synthase Type III ,Antigens, CD34 ,Biology ,Cell Line ,chemistry.chemical_compound ,Enos ,Lysophosphatidic acid ,Human Umbilical Vein Endothelial Cells ,Humans ,Platelet Activating Factor ,Progenitor cell ,Induced pluripotent stem cell ,Embryonic Stem Cells ,Medicine(all) ,Tumor Necrosis Factor-alpha ,Gene Expression Profiling ,Endothelial Cells ,Cell Differentiation ,General Medicine ,Cell Biology ,biology.organism_classification ,Embryonic stem cell ,Culture Media ,Cell biology ,Vascular endothelial growth factor ,Drug Combinations ,chemistry ,Biochemistry ,Cell culture ,Proteoglycans ,Collagen ,Laminin ,Lysophospholipids ,Developmental Biology - Abstract
The limited availability of human vascular endothelial cells (ECs) hampers research into EC function whilst the lack of precisely defined culture conditions for this cell type presents problems for addressing basic questions surrounding EC physiology. We aimed to generate endothelial progenitors from human pluripotent stem cells to facilitate the study of human EC physiology, using a defined serum-free protocol. Human embryonic stem cells (hESC-ECs) differentiated under serum-free conditions generated CD34+KDR+ endothelial progenitor cells after 6days that could be further expanded in the presence of vascular endothelial growth factor (VEGF). The resultant EC population expressed CD31 and TIE2/TEK, took up acetylated low-density lipoprotein (LDL) and up-regulated expression of ICAM-1, PAI-1 and ET-1 following treatment with TNFα. Immunofluorescence studies indicated that a key mediator of vascular tone, endothelial nitric oxide synthase (eNOS), was localised to a perinuclear compartment of hESC-ECs, in contrast with the pan-cellular distribution of this enzyme within human umbilical vein ECs (HUVECs). Further investigation revealed that that the serum-associated lipids, lysophosphatidic acid (LPA) and platelet activating factor (PAF), were the key molecules that affected eNOS localisation in hESC-ECs cultures. These studies illustrate the feasibility of EC generation from hESCs and the utility of these cells for investigating environmental cues that impact on EC phenotype. We have demonstrated a hitherto unrecognized role for LPA and PAF in the regulation of eNOS subcellular localization.
- Full Text
- View/download PDF
31. INS GFP/w human embryonic stem cells facilitate isolation of in vitro derived insulin-producing cells
- Author
-
Farida Sarangi, Xueling Li, Maria Cristina Nostro, Leonard C. Harrison, Sue Mei Lim, Claire E Hirst, Gordon Keller, Edouard G. Stanley, Qing Cissy Yu, Jacqueline V. Schiesser, Andrew G. Elefanty, David A. Elliott, and Suzanne J. Micallef
- Subjects
PAX6 Transcription Factor ,Recombinant Fusion Proteins ,Cellular differentiation ,Endocrinology, Diabetes and Metabolism ,Green Fluorescent Proteins ,LIM-Homeodomain Proteins ,Embryoid body ,Biology ,Cell Line ,Green fluorescent protein ,Genes, Reporter ,Insulin-Secreting Cells ,Internal Medicine ,Humans ,Insulin ,Paired Box Transcription Factors ,RNA, Messenger ,Eye Proteins ,Embryoid Bodies ,Embryonic Stem Cells ,Oligonucleotide Array Sequence Analysis ,Homeodomain Proteins ,Recombination, Genetic ,Gene Expression Profiling ,Nuclear Proteins ,Gene targeting ,Cell Differentiation ,Zebrafish Proteins ,Embryonic stem cell ,Molecular biology ,In vitro ,Clone Cells ,Repressor Proteins ,Transplantation ,Diabetes Mellitus, Type 1 ,Homeobox Protein Nkx-2.2 ,Cell culture ,embryonic structures ,Transcription Factors - Abstract
We aimed to generate human embryonic stem cell (hESC) reporter lines that would facilitate the characterisation of insulin-producing (INS⁺) cells derived in vitro.Homologous recombination was used to insert sequences encoding green fluorescent protein (GFP) into the INS locus, to create reporter cell lines enabling the prospective isolation of viable INS⁺ cells.Differentiation of INS(GFP/w) hESCs using published protocols demonstrated that all GFP⁺ cells co-produced insulin, confirming the fidelity of the reporter gene. INS-GFP⁺ cells often co-produced glucagon and somatostatin, confirming conclusions from previous studies that early hESC-derived insulin-producing cells were polyhormonal. INS(GFP/w) hESCs were used to develop a 96-well format spin embryoid body (EB) differentiation protocol that used the recombinant protein-based, fully defined medium, APEL. Like INS-GFP⁺ cells generated with other methods, those derived using the spin EB protocol expressed a suite of pancreatic-related transcription factor genes including ISL1, PAX6 and NKX2.2. However, in contrast with previous methods, the spin EB protocol yielded INS-GFP⁺ cells that also co-expressed the beta cell transcription factor gene, NKX6.1, and comprised a substantial proportion of monohormonal INS⁺ cells.INS(GFP/w) hESCs are a valuable tool for investigating the nature of early INS⁺ progenitors in beta cell ontogeny and will facilitate the development of novel protocols for generating INS⁺ cells from differentiating hESCs.
- Full Text
- View/download PDF
32. Enforced expression of mixll during mouse ES cell differentiation suppresses hematopoietic mesoderm and promotes endoderm formation
- Author
-
Sue Mei Lim, Lloyd Ashley Pereira, Michael Sebastian Wong, Claire Hirst, Benjamin Eugene Van Vranken, Marjorie Pick, Alan Trounson, Andrew George Elefanty, and Edouard Stanley
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.