Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Chromatogr A ; 1395: 136-42, 2015 May 22.
Article in English | MEDLINE | ID: mdl-25882588

ABSTRACT

Exposure of three native IgG1 monoclonal antibodies to 100mM acetate, pH 3.5 had no significant effect on their hydrodynamic size (11.5±0.5nm), while elution from protein A with the same buffer created a conformation of 5.5±1.0nm. Formation of the reduced-size conformation was preceded by the known destabilization of the second constant domain of the heavy chain (Cγ2) by contact with protein A, then compounded by exposure to low pH, creating extended flexibility in the hinge-Cγ2 region and allowing the Fab region to fold over the Fc region. The reduced-size conformation was necessary for complete elution. It persisted unchanged for at least 7 days under elution conditions. Physiological conditions restored native size, and it was maintained on re-exposure to 100mM acetate, pH 3.5. Protein A-mediated destabilization and subsequent restoration of native size did not create aggregates, but the reduced-size conformation was more susceptible to aggregation by secondary stress than native antibody. Protein A-mediated formation of the reduced-size conformation is probably universal during purification of human IgG1 antibodies, and may occur with other subclasses and IgG from other species, as well as Fc-fusion proteins.


Subject(s)
Chromatography, Affinity , Immunoglobulin G/chemistry , Staphylococcal Protein A/metabolism , Acetates/chemistry , Antibodies, Monoclonal/metabolism , Humans , Hydrogen-Ion Concentration , Immunoglobulin G/isolation & purification , Particle Accelerators , Staphylococcal Protein A/chemistry
2.
Hum Mol Genet ; 23(18): 4970-84, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24821703

ABSTRACT

A map of human embryo development that combines imaging, molecular, genetic and epigenetic data for comparisons to other species and across pathologies would be greatly beneficial for basic science and clinical applications. Here, we compared mRNA and protein expression of key mediators of DNA methylation and histone modifications between mouse and human embryos, embryos from fertile/infertile couples, and following growth factor supplementation. We observed that individual mouse and human embryos are characterized by similarities and distinct differences in DNA methylation and histone modification patterns especially at the single-cell level. In particular, while mouse embryos first exhibited sub-compartmentalization of different histone modifications between blastomeres at the morula stage and cell sub-populations in blastocysts, differential histone modification expression was detected between blastomeres earlier in human embryos at the four- to eight-cell stage. Likewise, differences in epigenetic mediator expression were also observed between embryos from fertile and infertile couples, which were largely equalized in response to growth factor supplementation, suggesting that select growth factors might prevent alterations in epigenetic profiles during prolonged embryo culture. Finally, we determined that reduced expression via morpholino technologies of a single histone-modifying enzyme, Rps6ka4/Msk2, resulted in cleavage-stage arrest as assessed by time-lapse imaging and was associated with aneuploidy generation. Taken together, data document differences in epigenetic patterns between species with implications for fertility and suggest functional roles for individual epigenetic factors during pre-implantation development.


Subject(s)
Blastomeres/metabolism , DNA Methylation , Embryonic Development , Methyltransferases/genetics , Animals , Embryo, Mammalian/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Histones/metabolism , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Species Specificity
3.
Biol Open ; 2(2): 121-31, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23429299

ABSTRACT

During preimplantation development, the embryo must establish totipotency and enact the earliest differentiation choices, processes that involve extensive chromatin modification. To identify novel developmental regulators, we screened for genes that are preferentially transcribed in the pluripotent inner cell mass (ICM) of the mouse blastocyst. Genes that encode chromatin remodeling factors were prominently represented in the ICM, including Chd1l, a member of the Snf2 gene family. Chd1l is developmentally regulated and expressed in embryonic stem (ES) cells, but its role in development has not been investigated. Here we show that inhibiting Chd1l protein production by microinjection of antisense morpholinos causes arrest prior to the blastocyst stage. Despite this important function in vivo, Chd1l is non-essential for cultured ES cell survival, pluripotency, or differentiation, suggesting that Chd1l is vital for events in embryos that are distinct from events in ES cells. Our data reveal a novel role for the chromatin remodeling factor Chd1l in the earliest cell divisions of mammalian development.

4.
Mol Syst Biol ; 9: 632, 2013.
Article in English | MEDLINE | ID: mdl-23295861

ABSTRACT

Landmark events occur in a coordinated manner during pre-implantation development of the mammalian embryo, yet the regulatory network that orchestrates these events remains largely unknown. Here, we present the first systematic investigation of the network in pre-implantation mouse embryos using morpholino-mediated gene knockdowns of key embryonic stem cell (ESC) factors followed by detailed transcriptome analysis of pooled embryos, single embryos, and individual blastomeres. We delineated the regulons of Oct4, Sall4, and Nanog and identified a set of metabolism- and transport-related genes that were controlled by these transcription factors in embryos but not in ESCs. Strikingly, the knockdown embryos arrested at a range of developmental stages. We provided evidence that the DNA methyltransferase Dnmt3b has a role in determining the extent to which a knockdown embryo can develop. We further showed that the feed-forward loop comprising Dnmt3b, the pluripotency factors, and the miR-290-295 cluster exemplifies a network motif that buffers embryos against gene expression noise. Our findings indicate that Oct4, Sall4, and Nanog form a robust and integrated network to govern mammalian pre-implantation development.


Subject(s)
Blastocyst/physiology , DNA-Binding Proteins/genetics , Embryonic Stem Cells/physiology , Gene Regulatory Networks , Homeodomain Proteins/genetics , Octamer Transcription Factor-3/genetics , Transcription Factors/genetics , Animals , Blastocyst/metabolism , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA-Binding Proteins/metabolism , Embryo Culture Techniques , Embryo, Mammalian/metabolism , Embryonic Development , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Homeodomain Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , MicroRNAs/genetics , Nanog Homeobox Protein , Octamer Transcription Factor-3/metabolism , Oligonucleotide Array Sequence Analysis , Transcription Factors/metabolism , DNA Methyltransferase 3B
5.
Cell Stem Cell ; 9(2): 156-65, 2011 Aug 05.
Article in English | MEDLINE | ID: mdl-21816366

ABSTRACT

Somatic tissues in female eutherian mammals are mosaic due to random X inactivation. In contrast to mice, X chromosome reactivation does not occur during the reprogramming of human female somatic cells to induced pluripotent stem cells (iPSCs), although this view is contested. Using balanced populations of female Rett patient and control fibroblasts, we confirm that all cells in iPSC colonies contain an inactive X, and additionally find that all colonies made from the same donor fibroblasts contain the same inactive X chromosome. Notably, this extreme "skewing" toward a particular dominant, active X is also a general feature of primary female fibroblasts during proliferation, and the skewing seen in reprogramming and fibroblast culture can be alleviated by overexpression of telomerase. These results have important implications for in vitro modeling of X-linked diseases and the interpretation of long-term culture studies in cancer and senescence using primary female fibroblast cell lines.


Subject(s)
Cellular Reprogramming/genetics , Chromosomes, Human, X/metabolism , Telomerase/metabolism , Animals , Base Sequence , Cell Proliferation , Cells, Cultured , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Mice , Models, Biological , Molecular Sequence Data , X Chromosome Inactivation
6.
J Biol Chem ; 286(20): 17755-65, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21454472

ABSTRACT

We compare here the structural and functional properties of heparan sulfate (HS) chains from both male or female adult mouse liver through a combination of molecular sieving, enzymatic cleavage, and strong anion exchange-HPLC. The results demonstrated that male and female HS chains are significantly different by a number of parameters; size determination showed that HS chain lengths were ∼100 and ∼22 kDa, comprising 30-40 and 6-8 disaccharide repeats, respectively. Enzymatic depolymerization and disaccharide composition analyses also demonstrated significant differences in domain organization and fine structure. N-Unsubstituted glucosamine (ΔHexA-GlcNH(3)(+), ΔHexA-GlcNH(3)(+)(6S), ΔHexA(2S)-GlcNH(3)(+), and N-acetylglucosamine (ΔHexA-GlcNAc) are the predominant disaccharides in male mouse liver HS. However, N-sulfated glucosamine (ΔHexA-GlcNSO(3)) is the predominant disaccharide found in female liver. These structurally different male and female liver HS forms exert differential effects on human mesenchymal cell proliferation and subsequent osteogenic differentiation. The present study demonstrates the potential usefulness of gender-specific liver HS for the manipulation of human mesenchymal cell properties, including expansion, multipotentiality, and subsequent matrix mineralization. Our results suggest that HS chains show both tissue- and gender-specific differences in biochemical composition that directly reflect their biological activity.


Subject(s)
Cell Differentiation/drug effects , Cell Proliferation/drug effects , Heparitin Sulfate/pharmacology , Liver/chemistry , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Sex Characteristics , Adult , Animals , Female , Heparitin Sulfate/chemistry , Heparitin Sulfate/isolation & purification , Humans , Male , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred CBA
7.
J Clin Invest ; 121(3): 1217-21, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21317531

ABSTRACT

Human induced pluripotent stem cells (hiPSCs) and human embryonic stem cells (hESCs) are promising candidate cell sources for regenerative medicine. However, despite the common ability of hiPSCs and hESCs to differentiate into all 3 germ layers, their functional equivalence at the single cell level remains to be demonstrated. Moreover, single cell heterogeneity amongst stem cell populations may underlie important cell fate decisions. Here, we used single cell analysis to resolve the gene expression profiles of 362 hiPSCs and hESCs for an array of 42 genes that characterize the pluripotent and differentiated states. Comparison between single hESCs and single hiPSCs revealed markedly more heterogeneity in gene expression levels in the hiPSCs, suggesting that hiPSCs occupy an alternate, less stable pluripotent state. hiPSCs also displayed slower growth kinetics and impaired directed differentiation as compared with hESCs. Our results suggest that caution should be exercised before assuming that hiPSCs occupy a pluripotent state equivalent to that of hESCs, particularly when producing differentiated cells for regenerative medicine aims.


Subject(s)
Embryonic Stem Cells/cytology , Gene Expression Regulation , Induced Pluripotent Stem Cells/cytology , Transcription, Genetic , Algorithms , Animals , Cell Differentiation , Cell Lineage , Cell Separation , Collagen/chemistry , Drug Combinations , Flow Cytometry , Gene Expression Profiling , Genetic Techniques , Humans , Kinetics , Laminin/chemistry , Mice , Mice, SCID , Myocytes, Cardiac/cytology , Proteoglycans/chemistry , Teratoma/metabolism
8.
Dev Biol ; 344(1): 66-78, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20430022

ABSTRACT

Divisions of polarised blastomeres that allocate polar cells to outer and apolar cells to inner positions initiate the first cell fate decision in the mouse embryo. Subsequently, outer cells differentiate into trophectoderm while inner cells retain pluripotency to become inner cell mass (ICM) of the blastocyst. Elimination of zygotic expression of trophectoderm-specific transcription factor Cdx2 leads to defects in the maintenance of the blastocyst cavity, suggesting that it participates only in the late stage of trophectoderm formation. However, we now find that mouse embryos also have a maternally provided pool of Cdx2 mRNA. Moreover, depletion of both maternal and zygotic Cdx2 from immediately after fertilization by three independent approaches, dsRNAi, siRNAi and morpholino oligonucleotides, leads to developmental arrest at much earlier stages than expected from elimination of only zygotic Cdx2. This developmental arrest is associated with defects in cell polarisation, reflected by expression and localisation of cell polarity molecules such as Par3 and aPKC and cell compaction at the 8- and 16-cell stages. Cells deprived of Cdx2 show delayed development with increased cell cycle length, irregular cell division and increased incidence of apoptosis. Although some Cdx2-depleted embryos initiate cavitation, the cavity cannot be maintained. Furthermore, expression of trophectoderm-specific genes, Gata3 and Eomes, and also the trophectoderm-specific cytokeratin intermediate filament, recognised by Troma1, are greatly reduced or undetectable. Taken together, our results indicate that Cdx2 participates in two steps leading to trophectoderm specification: appropriate polarisation of blastomeres at the 8- and 16-cell stage and then the maintenance of trophectoderm lineage-specific differentiation.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Transcription Factors/physiology , Animals , Apoptosis , CDX2 Transcription Factor , Cell Differentiation , Ectoderm/metabolism , Female , Immunohistochemistry/methods , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred C57BL , Models, Biological , Protein Kinase C/metabolism , RNA Interference
9.
Dev Dyn ; 238(4): 950-5, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19301395

ABSTRACT

The high attrition rate of in vitro human embryo culture presents a major obstacle in the treatment of clinical infertility by in vitro fertilization (IVF). Physical and genetic requirements are not well understood for human or mouse preimplantation embryo development. Group culture is an established requirement for optimal embryo development in the mouse model. However, conventional microdrop culture limitations hinder investigations of the effects of physical parameters on in vitro embryo development. We report a microfluidics platform that enables embryo culture in precisely defined, sub-microliter volumes (5-500 nl) which cannot be investigated using conventional methods. Groups of two embryos per microfluidic well successfully developed to the blastocyst stage, at a rate of over 80%, which is comparable to those cultured in 20-microl microdrops. This system can be used to dissect physical requirements of in vitro single or group embryo culture, and be made highly parallel to increase experimental throughput.


Subject(s)
Embryo Culture Techniques/methods , Microfluidic Analytical Techniques/instrumentation , Microfluidic Analytical Techniques/methods , Animals , Female , Mice
10.
PLoS One ; 3(12): e4109, 2008.
Article in English | MEDLINE | ID: mdl-19129941

ABSTRACT

BACKGROUND: Compared to the emerging embryonic stem cell (ESC) gene network, little is known about the dynamic gene network that directs reprogramming in the early embryo. We hypothesized that Oct4, an ESC pluripotency regulator that is also highly expressed at the 1- to 2-cell stages in embryos, may be a critical regulator of the earliest gene network in the embryo. METHODOLOGY/PRINCIPAL FINDINGS: Using antisense morpholino oligonucleotide (MO)-mediated gene knockdown, we show that Oct4 is required for development prior to the blastocyst stage. Specifically, Oct4 has a novel and critical role in regulating genes that encode transcriptional and post-transcriptional regulators as early as the 2-cell stage. Our data suggest that the key function of Oct4 may be to switch the developmental program from one that is predominantly regulated by post-transcriptional control to one that depends on the transcriptional network. Further, we propose to rank candidate genes quantitatively based on the inter-embryo variation in their differential expression in response to Oct4 knockdown. Of over 30 genes analyzed according to this proposed paradigm, Rest and Mta2, both of which have established pluripotency functions in ESCs, were found to be the most tightly regulated by Oct4 at the 2-cell stage. CONCLUSIONS/SIGNIFICANCE: We show that the Oct4-regulated gene set at the 1- to 2-cell stages of early embryo development is large and distinct from its established network in ESCs. Further, our experimental approach can be applied to dissect the gene regulatory network of Oct4 and other pluripotency regulators to deconstruct the dynamic developmental program in the early embryo.


Subject(s)
Embryonic Development/genetics , Gene Expression Regulation, Developmental , Gene Regulatory Networks/physiology , Octamer Transcription Factor-3/genetics , Animals , Blastocyst , Embryonic Stem Cells , Female , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Mice , Mothers , Octamer Transcription Factor-3/metabolism , Pregnancy , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
11.
Cell Cycle ; 6(18): 2276-83, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17881898

ABSTRACT

High content cell-based genetic and small molecule library screens are powerful strategies in drug discovery and investigations of disease mechanisms. We report that primary cells derived from a transgenic mouse model expressing a fluorescence mitosis biosensor provide unambiguous phenotype readouts without the need for transfection or immunocytochemistry. Phenotype profiles of cell cycle disruption and of apoptosis are easily detectable at a single time point selected from time-lapse live fluorescence microscopy. Most importantly, this transgenic mouse model may be crossed with cancer mouse models to derive biosensor-expressing primary cancer cells for use in high content screening strategies targeting discovery of tumor-specific chemotherapeutic compounds.


Subject(s)
Cell Cycle/genetics , Fibroblasts/physiology , Genetic Testing/methods , Models, Animal , Phenotype , Animals , Female , Fibroblasts/cytology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Pregnancy , Species Specificity
12.
Stem Cells ; 25(11): 2845-54, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17702986

ABSTRACT

Cell surface heparan sulfate (HS) and chondroitin sulfate (CS) proteoglycans have been implicated in a multitude of biological processes, including embryonic implantation, tissue morphogenesis, wound repair, and neovascularization through their ability to regulate growth factor activity and morphogenic gradients. However, the direct role of the glycosaminoglycan (GAG) sugar-side chains in the control of human mesenchymal stem cell (hMSC) differentiation into the osteoblast lineage is poorly understood. Here, we show that the abundant cell surface GAGs, HS and CS, are secreted in proteoglycan complexes that directly regulate the bone morphogenetic protein (BMP)-mediated differentiation of hMSCs into osteoblasts. Enzymatic depletion of the HS and CS chains by heparinase and chondroitinase treatment decreased HS and CS expression but did not alter the expression of the HS core proteins perlecan and syndecan. When digested separately, depletion of HS and CS chains did not effect hMSC proliferation but rather increased BMP bioactivity through SMAD1/5/8 intracellular signaling at the same time as increasing canonical Wnt signaling through LEF1 activation. Long-term culturing of cells in HS- and CS-degrading enzymes also increased bone nodule formation, calcium accumulation, and the expression of such osteoblast markers as alkaline phosphatase, RUNX2, and osteocalcin. Thus, the enzymatic disruption of HS and CS chains on cell surface proteoglycans alters BMP and Wnt activity so as to enhance the lineage commitment and osteogenic differentiation of hMSCs.


Subject(s)
Bone Morphogenetic Proteins/physiology , Cell Differentiation/physiology , Chondroitin Sulfates/physiology , Heparitin Sulfate/physiology , Mesenchymal Stem Cells/physiology , Osteogenesis/physiology , Cell Differentiation/drug effects , Chondroitin Sulfates/antagonists & inhibitors , Chondroitinases and Chondroitin Lyases/pharmacology , Heparin Lyase/pharmacology , Heparitin Sulfate/antagonists & inhibitors , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Osteogenesis/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...