128 results on '"Ren-He Xu"'
Search Results
52. Universal Corneal Epithelial-Like Cells Derived from Human Embryonic Stem Cells for Cellularization of a Corneal Scaffold
- Author
-
Juan Yang, Dejin Zheng, Jung Woo Park, and Ren-He Xu
- Subjects
0301 basic medicine ,Cell ,Biomedical Engineering ,immunogenicity ,03 medical and health sciences ,Cornea ,medicine ,Decellularization ,Tight junction ,Chemistry ,decellularized cornea ,Articles ,differentiation ,human embryonic stem cells ,Embryonic stem cell ,Epithelium ,B2M knockout ,Cell biology ,Transplantation ,Ophthalmology ,Chemically defined medium ,030104 developmental biology ,medicine.anatomical_structure ,corneal epithelial cells ,embryonic structures ,cardiovascular system - Abstract
Purpose We generated universal corneal epithelial cells (CEC) from human embryonic stem cells (hESC) by genetically removing human leukocyte antigens (HLA) class I from the cell surface. Methods The serum-free, growth factor-free, and defined medium E6 was used to differentiate hESC to CEC. Decellularized murine corneas were recellularized with hESC-derived CEC. Using CRISPR/Cas9, β-2-microglobulin (B2M) was deleted in hESC to block the assembly of HLA class-I antigens on the cell surface to generate B2M -/- CEC. Results E6 alone was sufficient to allow hESC differentiation to CEC. A time-course analysis of the global gene expression of the differentiating cells indicates that the differentiation closely resembles the corneal development in vivo. The hESC-CEC were highly proliferative, and could form multilayer epithelium in decellularized murine cornea, retain its transparency, and form intact tight junctions on its surface. As reported before, B2M knockout led to the absence of HLA class-I on the cell surface of hESC and subsequently derived CEC following stimulation with inflammatory factors. Moreover, B2M -/- CEC, following transplantation into mouse eyes, caused less T-cell infiltration in the limbal region of the eye than the wild-type control. Conclusions CEC can be derived from hESC via a novel and simple protocol free of any proteins, hESC-CEC seeded on decellularized animal cornea form tight junctions and allow light transmittance, and B2M -/- CEC are hypoimmunogenic both in vitro and in vivo. Translational relevance B2M -/- hESC-CEC can be an unlimited and universal therapy for corneal repair in patients of any HLA type.
- Published
- 2018
- Full Text
- View/download PDF
53. Progresses and Challenges in Optimization of Human Pluripotent Stem Cell Culture
- Author
-
Ren-He Xu and Ge Lin
- Subjects
Pluripotent Stem Cells ,Cell type ,Receptors, Notch ,Nodal Protein ,Somatic cell ,Cell Culture Techniques ,Medicine (miscellaneous) ,General Medicine ,Computational biology ,Biology ,Embryonic stem cell ,Genomic Instability ,Activins ,Wnt Proteins ,Transforming Growth Factor beta ,Bone Morphogenetic Proteins ,Animals ,Humans ,Fibroblast Growth Factor 2 ,Human Induced Pluripotent Stem Cells ,Induced pluripotent stem cell ,Reprogramming ,Cells, Cultured ,Embryonic Stem Cells - Abstract
The pressing demand to elucidate the biology of human embryonic stem (ES) cells and to realize their therapeutic potential has greatly promoted the progresses in the optimization of the culture systems used for this highly promising cell type. These progresses include the characterization of exogenous regulators of pluripotency and differentiation, the development of animal-free, defined, and scalable culture systems, and some pioneering efforts to establish good manufactory practice facilities to derive and expand clinical-grade human ES cells and their derivatives. All of these advancements appear to be also applicable to the derivation and culture of human induced pluripotent stem cells, an ES cell-like cell type derived from somatic cells via reprogramming. This review attempts to summarize these progresses and discuss some of the remaining challenges.
- Published
- 2010
- Full Text
- View/download PDF
54. Vaccination with Human Pluripotent Stem Cells Generates a Broad Spectrum of Immunological and Clinical Responses Against Colon Cancer
- Author
-
Hui Zeng, Yi Li, Zihai Li, Ren-He Xu, and Bei Liu
- Subjects
Pluripotent Stem Cells ,Cellular differentiation ,Autoimmunity ,Adenocarcinoma ,Biology ,Cell Line ,Mice ,Immune system ,Cancer stem cell ,Animals ,Humans ,Induced pluripotent stem cell ,Mice, Inbred BALB C ,Vaccination ,Cell Differentiation ,Cell Biology ,Xenograft Model Antitumor Assays ,Embryonic stem cell ,Colonic Neoplasms ,Immunology ,Cancer cell ,Molecular Medicine ,Female ,Immunotherapy ,Stem cell ,Oncofetal antigen ,Spleen ,Developmental Biology - Abstract
The history of immunizing with embryonic materials to generate an antitumor immune response dates back to a century ago. The premise is that cancer cells share the expression of oncofetal antigens with embryonic materials and that the immune response against these antigens in the embryonic tissues is cross-protective against cancer. However, such a practice has never advanced beyond experimental animal settings, because of lack of uniformed source tissues and ethical challenges. With the availability of well-characterized human pluripotent stem cells, it is now possible to ask whether tumor protective immunity could indeed be elicited with stem cells. Herein, we investigated whether vaccination with defined human embryonic stem cells (hESCs) or induced pluripotent stem (iPS) cells was effective against a colon carcinoma. We discovered that vaccination of mice with hESC line H9 generated consistent cellular and humoral immune responses against CT26 colon carcinoma. Protection correlated strongly with the expansion of tumor-responsive and interferon-γ-producing cells and the profound loss of CD11b+Gr-1+ myeloid-derived suppressor cells in the spleen. No evidence of autoimmunity was observed. We also compared the immunogenicity against colon cancer between a hESC line CT2 and an iPS cell line TZ1 that were generated in the same stem cell facility. We found that the iPS cell line was inferior to the hESC line in conferring tumor protection, suggesting that there is heterogeneity of expression of oncofetal antigens by hESCs and iPS cells. We conclude that the hESC-based vaccine is a promising modality for immunotherapy of cancer. Disclosure of potential conflicts of interest is found at the end of this article.
- Published
- 2009
- Full Text
- View/download PDF
55. Lack of ABCG2 Expression and Side Population Properties in Human Pluripotent Stem Cells
- Author
-
Xue-Jun Li, Jung Woo Park, Leann Crandall, Ge Lin, Xiaofang Wang, Hui Zeng, Ren-He Xu, Min Guo, Tiwanna Compton, and Fangping Chen
- Subjects
Pluripotent Stem Cells ,KOSR ,animal structures ,Cell Survival ,Cell Culture Techniques ,Drug Resistance ,Antineoplastic Agents ,Embryoid body ,Biology ,Mice ,Species Specificity ,Cancer stem cell ,ATP Binding Cassette Transporter, Subfamily G, Member 2 ,Animals ,Humans ,Induced pluripotent stem cell ,reproductive and urinary physiology ,Cell Differentiation ,Cell Biology ,Molecular biology ,Embryonic stem cell ,Culture Media ,Neoplasm Proteins ,Trophoblasts ,Endothelial stem cell ,embryonic structures ,Molecular Medicine ,ATP-Binding Cassette Transporters ,Benzimidazoles ,Fibroblast Growth Factor 2 ,sense organs ,Mitoxantrone ,biological phenomena, cell phenomena, and immunity ,Stem cell ,DNA Damage ,Developmental Biology ,Adult stem cell - Abstract
The multidrug transporter ABCG2 in cell membranes enables various stem cells and cancer cells to efflux chemicals, including the fluorescent dye Hoechst 33342. The Hoechst− cells can be sorted out as a side population with stem cell properties. Abcg2 expression in mouse embryonic stem cells (ESCs) reduces accumulation of DNA-damaging metabolites in the cells, which helps prevent cell differentiation. Surprisingly, we found that human ESCs do not express ABCG2 and cannot efflux Hoechst. In contrast, trophoblasts and neural epithelial cells derived from human ESCs are ABCG2+ and Hoechst−. Human ESCs ectopically expressing ABCG2 become Hoechst−, more tolerant of toxicity of mitoxantrone, a substrate of ABCG2, and more capable of self-renewal in basic fibroblast growth factor (bFGF)-free condition than control cells. However, Hoechstlow cells sorted as a small subpopulation from human ESCs express lower levels of pluripotency markers than the Hoechsthigh cells. Similar results were observed with human induced pluripotent stem cells. Conversely, mouse ESCs are Abcg2+ and mouse trophoblasts, Abcg2−. Thus, absence of ABCG2 is a novel feature of human pluripotent stem cells, which distinguishes them from many other stem cells including mouse ESCs, and may be a reason why they are sensitive to suboptimal culture conditions.
- Published
- 2009
- Full Text
- View/download PDF
56. Expression Profiling of Nuclear Receptors in Human and Mouse Embryonic Stem Cells
- Author
-
Yangsik Jeong, Bong Hyun Kim, Angie L. Bookout, Jifeng Zhang, Tingwan Sun, David J. Mangelsdorf, Y. Eugene Chen, Sierra Root, Minerva T. Garcia-Barrio, Yang Xie, Chang Qing Xie, Ren-He Xu, and Mingui Fu
- Subjects
Cellular differentiation ,Receptors, Cytoplasmic and Nuclear ,Embryoid body ,Biology ,Polymerase Chain Reaction ,Mice ,Endocrinology ,Gene expression ,Animals ,Humans ,Research Resource ,Receptor ,Molecular Biology ,Cells, Cultured ,Embryonic Stem Cells ,Regulation of gene expression ,Gene Expression Profiling ,Computational Biology ,Gene Expression Regulation, Developmental ,Cell Differentiation ,General Medicine ,Embryonic stem cell ,humanities ,Cell biology ,Gene expression profiling ,Nuclear receptor ,human activities - Abstract
Nuclear receptors (NRs) regulate gene expression in essential biological processes including differentiation and development. Here we report the systematic profiling of NRs in human and mouse embryonic stem cell (ESC) lines and during their early differentiation into embryoid bodies. Expression of the 48 human and mouse NRs was assessed by quantitative real-time PCR. In general, expression of NRs between the two human cell lines was highly concordant, whereas in contrast, expression of NRs between human and mouse ESCs differed significantly. In particular, a number of NRs that have been implicated previously as crucial regulators of mouse ESC biology, including ERRbeta, DAX-1, and LRH-1, exhibited diametric patterns of expression, suggesting they may have distinct species-specific functions. Taken together, these results highlight the complexity of the transcriptional hierarchy that exists between species and governs early development. These data should provide a unique resource for further exploration of the species-specific roles of NRs in ESC self-renewal and differentiation.
- Published
- 2009
- Full Text
- View/download PDF
57. Dissecting Signaling Pathways That Govern Self-renewal of Rabbit Embryonic Stem Cells
- Author
-
Shufen Wang, Kai Chen, Xiyun Yan, Qi Zhou, Xiaohua Yuan, Jian Li, Yongchang Chen, Ren-He Xu, Weizhi Ji, Xiangyu Guo, Yi Shen, and Yuyu Niu
- Subjects
Pluripotent Stem Cells ,MAPK/ERK pathway ,MAP Kinase Signaling System ,Smad2 Protein ,Biology ,Fibroblast growth factor ,Biochemistry ,Cell Line ,Phosphatidylinositol 3-Kinases ,Transforming Growth Factor beta ,Animals ,Smad3 Protein ,Phosphorylation ,Molecular Biology ,Protein kinase B ,Embryonic Stem Cells ,beta Catenin ,PI3K/AKT/mTOR pathway ,Cell Proliferation ,Mitogen-Activated Protein Kinase 1 ,Mitogen-Activated Protein Kinase 3 ,Epidermal Growth Factor ,Wnt signaling pathway ,Cell Differentiation ,Cell Biology ,Embryonic stem cell ,Cell biology ,Wnt Proteins ,Rabbits ,Stem cell ,Carrier Proteins ,Proto-Oncogene Proteins c-akt ,Transforming growth factor - Abstract
The pluripotency and self-renewal of embryonic stem cells (ESC) are regulated by a variety of cytokines/growth factors with some species differences. We reported previously that rabbit ESC (rESC) are more similar to primate ESC than to mouse ESC. However, the signaling pathways that regulate rESC self-renewal had not been identified. Here we show that inhibition of the transforming growth factor beta (TGFbeta), fibroblast growth factor (FGF), and canonical Wnt/beta-catenin (Wnt) pathways results in enhanced differentiation of rESC accompanied by down-regulation of Smad2/3 phosphorylation and beta-catenin expression and up-regulation of phosphorylation of Smad1 and beta-catenin. These results imply that the TGFbeta, FGF, and Wnt pathways are required for rESC self-renewal. Inhibition of the MAPK/ERK and PI3K/AKT pathways, which lie downstream of the FGF pathway, led to differentiation of rESC accompanied by down-regulation of phosphorylation of ERK1/2 or AKT, respectively. Long-term self-renewal of rESC could be achieved by adding a mixture of TGFbeta ligands (activin A, Nodal, or TGFbeta1) plus basic FGF (bFGF) and Noggin in the absence of serum and feeder cells. Our findings also suggest that there is a regulatory network consisting of the FGF, Wnt, and TGFbeta pathways that controls rESC pluripotency and self-renewal. We conclude that bFGF controls the stem cell properties of rESC both directly and indirectly through TGFbeta or other pathways, whereas the effect of Wnt on rESC might be mediated by the TGFbeta pathway.
- Published
- 2008
- Full Text
- View/download PDF
58. X-inactivation in female human embryonic stem cells is in a nonrandom pattern and prone to epigenetic alterations
- Author
-
Ren-He Xu, Youko Matsuno, Nagesh Rao, Yin Shen, Shaun D. Fouse, Arthur D. Riggs, Matteo Pellegrini, Sierra H. Root, and Guoping Fan
- Subjects
Genetic Markers ,DNA, Complementary ,RNA, Untranslated ,Genotype ,Somatic cell ,Biology ,Polymorphism, Single Nucleotide ,X-inactivation ,Epigenesis, Genetic ,Genes, X-Linked ,X Chromosome Inactivation ,Dosage Compensation, Genetic ,Humans ,Epigenetics ,Promoter Regions, Genetic ,Cells, Cultured ,Embryonic Stem Cells ,X chromosome ,Genetics ,Regulation of gene expression ,Multidisciplinary ,Dosage compensation ,Reverse Transcriptase Polymerase Chain Reaction ,DNA Methylation ,Biological Sciences ,DNA methylation ,CpG Islands ,Female ,RNA, Long Noncoding ,XIST - Abstract
X chromosome inactivation (XCI) is an essential mechanism for dosage compensation of X-linked genes in female cells. We report that subcultures from lines of female human embryonic stem cells (hESCs) exhibit variation (0–100%) for XCI markers, including XIST RNA expression and enrichment of histone H3 lysine 27 trimethylation (H3K27me3) on the inactive X chromosome (Xi). Surprisingly, regardless of the presence or absence of XCI markers in different cultures, all female hESCs we examined (H7, H9, and HSF6 cells) exhibit a monoallelic expression pattern for a majority of X-linked genes. Our results suggest that these established female hESCs have already completed XCI during the process of derivation and/or propagation, and the XCI pattern of lines we investigated is already not random. Moreover, XIST gene expression in subsets of cultured female hESCs is unstable and subject to stable epigenetic silencing by DNA methylation. In the absence of XIST expression, ≈12% of X-linked promoter CpG islands become hypomethylated and a portion of X-linked alleles on the Xi are reactivated. Because alterations in dosage compensation of X-linked genes could impair somatic cell function, we propose that XCI status should be routinely checked in subcultures of female hESCs, with cultures displaying XCI markers better suited for use in regenerative medicine.
- Published
- 2008
- Full Text
- View/download PDF
59. Concise Review: One Stone for Multiple Birds: Generating Universally Compatible Human Embryonic Stem Cells
- Author
-
Dejin Zheng, Xiaofang Wang, and Ren-He Xu
- Subjects
0301 basic medicine ,Immunosuppression Therapy ,Carcinogenesis ,Human Embryonic Stem Cells ,Cell Culture Techniques ,Cell Biology ,Human leukocyte antigen ,Biology ,Major histocompatibility complex ,Embryonic stem cell ,Regenerative medicine ,Umbilical Cord ,03 medical and health sciences ,030104 developmental biology ,biology.protein ,Cancer research ,Molecular Medicine ,Gene silencing ,Humans ,Antibody ,Stem cell ,Induced pluripotent stem cell ,Developmental Biology - Abstract
With ongoing clinical trials, human embryonic stem cells (hESCs) have shown substantial potential for regenerative medicine. However, due to the mismatch of human leukocyte antigens (HLAs) between hESC-derived allografts and recipients, immunosuppressant regimens must be used to prevent immune rejection of the grafts. Considerable efforts have been devoted to overcoming this hurdle via the derivation and banking of human nuclear transfer ESCs, parthenogenetic ESCs, and induced pluripotent stem cells. However, ethical and safety concerns remain, hindering the application of these types of pluripotent cells. Other approaches have recently been explored to generate universally compatible hESCs through the silencing or deletion of HLAs or genes essential for HLA expression, including β-2-microglobulin and class-II MHC transactivator, as well as the induction of immunosuppression via the ectopic expression of non-classical HLAs (e.g., HLA-E and -G), cytotoxic T lymphocyte antigen 4 fused with immunoglobulin, and programmed death ligand-1. In this review, we introduce developments in this line of research and discuss strategies to reduce the tumorigenic concerns regarding hESCs, especially after they acquire the capability to escape immune surveillance. Stem Cells 2016;34:2269–2275
- Published
- 2016
60. CRISPR/Cas9-AAV Mediated Knock-in at NRL Locus in Human Embryonic Stem Cells
- Author
-
Fayu Yang, Junzhao Zhao, Yanghua Fu, Jia Qu, Ge Lin, Haitao Xi, Ding Chen, Feng Gu, Ren-He Xu, Xiao Zhi, and Xianglian Ge
- Subjects
0301 basic medicine ,Genetics ,Trans-activating crRNA ,next generation sequencing ,CRISPR interference ,Cas9 ,DNA repair ,lcsh:RM1-950 ,Gene targeting ,AAV ,homologous recombination ,Computational biology ,Biology ,human embryonic stem cells ,Genome engineering ,03 medical and health sciences ,030104 developmental biology ,lcsh:Therapeutics. Pharmacology ,Genome editing ,Drug Discovery ,Molecular Medicine ,CRISPR ,Original Article ,NRL ,CRISPR/Cas9 - Abstract
Clustered interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9)-mediated genome engineering technologies are sparking a new revolution in biological research. This technology efficiently induces DNA double strand breaks at the targeted genomic sequence and results in indel mutations by the error-prone process of nonhomologous end joining DNA repair or homologous recombination with a DNA repair template. The efficiency of genome editing with CRISPR/Cas9 alone in human embryonic stem cells is still low. Gene targeting with adeno-associated virus (AAV) vectors has been demonstrated in multiple human cell types with maximal targeting frequencies without engineered nucleases. However, whether CRISPR/Cas9-mediated double strand breaks and AAV based donor DNA mediated homologous recombination approaches could be combined to create a novel CRISPR/Cas9-AAV genetic tool for highly specific gene editing is not clear. Here we demonstrate that using CRISPR/Cas9-AAV, we could successfully knock-in a DsRed reporter gene at the basic motifleucine zipper transcription factor (NRL) locus in human embryonic stem cells. For the first time, this study provides the proof of principle that these two technologies can be used together. CRISPR/Cas9-AAV, a new genome editing tool, offers a platform for the manipulation of human genome.
- Published
- 2016
61. Derivation and Chondrogenic Commitment of Human Embryonic Stem Cell-Derived Mesenchymal Progenitors
- Author
-
Hicham, Drissi, Jason D, Gibson, Rosa M, Guzzo, and Ren-He, Xu
- Subjects
Tissue Engineering ,Tissue Scaffolds ,Bone Morphogenetic Protein 2 ,Feeder Cells ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Mesenchymal Stem Cells ,Cell Separation ,Flow Cytometry ,Mesenchymal Stem Cell Transplantation ,Regenerative Medicine ,Coculture Techniques ,Mice ,Chondrocytes ,Phenotype ,Animals ,Humans ,Cell Lineage ,Chondrogenesis ,Biomarkers ,Embryonic Stem Cells - Abstract
The induction of human embryonic stem cells to a mesenchymal-like progenitor population constitutes a developmentally relevant approach for efficient directed differentiation of human embryonic stem (hES) cells to the chondrogenic lineage. The initial enrichment of a hemangioblast intermediate has been shown to yield a replenishable population of highly purified progenitor cells that exhibit the typical mesenchymal stem cell (MSC) surface markers as well as the capacity for multilineage differentiation to bone, fat, and cartilage. Herein, we provide detailed methodologies for the derivation and characterization of potent mesenchymal-like progenitors from hES cells and describe in vitro assays for bone morphogenetic protein (BMP)-2-mediated differentiation to the chondrogenic lineage.
- Published
- 2015
62. Efficient differentiation of cardiomyocytes from human pluripotent stem cells with growth factors
- Author
-
Ren-He Xu, Chunhui Xu, and Rajneesh Jha
- Subjects
Pluripotent Stem Cells ,medicine.diagnostic_test ,Drug discovery ,Insulin ,medicine.medical_treatment ,Immunocytochemistry ,Cell replacement ,Cell Differentiation ,Biology ,In Vitro Techniques ,Flow Cytometry ,Real-Time Polymerase Chain Reaction ,Molecular biology ,Immunohistochemistry ,Article ,Cell biology ,Flow cytometry ,Real-time polymerase chain reaction ,medicine ,Myocyte ,Humans ,Intercellular Signaling Peptides and Proteins ,Myocytes, Cardiac ,Induced pluripotent stem cell - Abstract
Human pluripotent stem cells have tremendous replicative capacity and demonstrated potential to generate functional cardiomyocytes. These cardiomyocytes represent a promising source for cell replacement therapy to treat heart disease and may serve as a useful tool for drug discovery and disease modeling. Efficient cardiomyocyte differentiation, a prerequisite for the application of stem cell-derived cardiomyocytes, can be achieved with a growth factor-guided method. Undifferentiated cells are sequentially treated with activin A and BMP4 in a serum-free and insulin-free medium and then maintained in a serum-free medium with insulin. This method yields as much as >75% cardiomyocytes in the differentiation culture within 2 weeks, and the beating cardiomyocytes have expected molecular, cellular, and electrophysiological characteristics. In this chapter, we describe in detail the differentiation protocol and follow-up characterization focusing on immunocytochemistry, quantitative RT-PCR, and flow cytometry analysis.
- Published
- 2015
63. Cold-inducible RNA binding protein is required for the expression of adhesion molecules and embryonic cell movement in Xenopus laevis
- Author
-
Julian A. Tanner, Hsiang-Fu Kung, Ren-He Xu, Ying Peng, Pai Hao Yang, Henry F. Lee, Jian-Dong Huang, Ming Li, and Marie C.M. Lin
- Subjects
Embryo, Nonmammalian ,Morpholino ,Biophysics ,Xenopus ,Nerve Tissue Proteins ,RNA-binding protein ,Xenopus Proteins ,Biology ,Biochemistry ,Xenopus laevis ,Cell Movement ,Sense (molecular biology) ,Animals ,Cell Lineage ,RNA, Antisense ,Molecular Biology ,Messenger RNA ,Cell adhesion molecule ,Gene Expression Regulation, Developmental ,RNA-Binding Proteins ,RNA ,Cell migration ,Cell Biology ,biology.organism_classification ,Cell biology ,Cell Adhesion Molecules - Abstract
We have previously shown that the Xenopus homologue of cold-inducible RNA binding protein, XCIRP-1, is required for the morphogenetic migration of the pronephros during embryonic development. However, the underlying molecular mechanisms remain elusive. Here, we report that XCIRP is essential for embryonic cell movement, as suppression of XCIRP by microinjection of anti-sense mRNA and morpholino antisense oligonucleotides (MOs) significantly reduced protein expression, inhibited the cell migration rate, and inhibited eFGF and activin-induced animal cap elongation. By immunoprecipitation and RT-PCR, we further showed that the mRNA of a panel of adhesion molecules, including alphaE- and beta-catenin, C- and E-cadherin, and paraxial proto-cadherin, are the targets of XCIRP. Consistently, in animal cap explant studies, suppression of XCIRP by MOs inhibited the expression of these adhesion molecules, while over-expression of sense XCIRP-1 mRNA fully rescued this inhibition. Taken together, these results suggest for the first time that XCIRP is required to maintain the expression of adhesion molecules and cell movement during embryonic development.
- Published
- 2006
- Full Text
- View/download PDF
64. Basic Fibroblast Growth Factor Support of Human Embryonic Stem Cell Self-Renewal
- Author
-
Mark E. Levenstein, James A. Thomson, Ren-He Xu, Tenneille Ludwig, Rachel A. Llanas, Daisy Manning, and Kaitlyn VanDenHeuvel‐Kramer
- Subjects
KOSR ,Population ,Basic fibroblast growth factor ,Cell Culture Techniques ,Biology ,Fibroblast growth factor ,Article ,Cell Line ,chemistry.chemical_compound ,Humans ,education ,Cell Proliferation ,education.field_of_study ,integumentary system ,Dose-Response Relationship, Drug ,Stem Cells ,Cell Biology ,Fibroblasts ,Embryo, Mammalian ,Embryonic stem cell ,Cell biology ,chemistry ,Cell culture ,Culture Media, Conditioned ,Immunology ,Molecular Medicine ,Fibroblast Growth Factor 2 ,Stem cell ,Developmental Biology ,Adult stem cell - Abstract
Human embryonic stem (ES) cells have most commonly been cultured in the presence of basic fibroblast growth factor (FGF2) either on fibroblast feeder layers or in fibroblast-conditioned medium. It has recently been reported that elevated concentrations of FGF2 permit the culture of human ES cells in the absence of fibroblasts or fibroblast-conditioned medium. Herein we compare the ability of unconditioned medium (UM) supplemented with 4, 24, 40, 80, 100, and 250 ng/ml FGF2 to sustain low-density human ES cell cultures through multiple passages. In these stringent culture conditions, 4, 24, and 40 ng/ml FGF2 failed to sustain human ES cells through three passages, but 100 ng/ml sustained human ES cells with an effectiveness comparable to conditioned medium (CM). Two human ES cell lines (H1 and H9) were maintained for up to 164 population doublings (7 and 4 months) in UM supplemented with 100 ng/ml FGF2. After prolonged culture, the cells formed teratomas when injected into severe combined immunodeficient beige mice and expressed markers characteristic of undifferentiated human ES cells. We also demonstrate that FGF2 is degraded more rapidly in UM than in CM, partly explaining the need for higher concentrations of FGF2 in UM. These results further facilitate the large-scale, routine culture of human ES cells and suggest that fibroblasts and fibro-blast-conditioned medium sustain human ES cells in part by stabilizing FGF signaling above a critical threshold.
- Published
- 2005
- Full Text
- View/download PDF
65. Basic FGF and suppression of BMP signaling sustain undifferentiated proliferation of human ES cells
- Author
-
Xuezhu Feng, Dong S. Li, James A. Thomson, Tenneille Ludwig, Ruthann M. Peck, and Ren-He Xu
- Subjects
animal structures ,Basic fibroblast growth factor ,Cell Culture Techniques ,Biology ,Bone morphogenetic protein ,Biochemistry ,Cell Line ,chemistry.chemical_compound ,medicine ,Humans ,Noggin ,Fibroblast ,Molecular Biology ,reproductive and urinary physiology ,Cell Proliferation ,Regulation of gene expression ,Dose-Response Relationship, Drug ,Tissue Engineering ,Stem Cells ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Cell Biology ,equipment and supplies ,Embryonic stem cell ,Cell biology ,Drug Combinations ,medicine.anatomical_structure ,chemistry ,Cell culture ,Bone Morphogenetic Proteins ,embryonic structures ,Immunology ,Fibroblast Growth Factor 2 ,Carrier Proteins ,Leukemia inhibitory factor ,Signal Transduction ,Biotechnology - Abstract
Human embryonic stem cells (hESCs) are routinely cultured on fibroblast feeder layers or in fibroblast-conditioned medium (CM). Bone morphogenetic proteins (BMPs) have previously been shown to induce hESC differentiation, in apparent contrast to mouse embryonic stem (ES) cells, in which BMP4 synergizes with leukemia inhibitory factor (LIF) to maintain self-renewal. Here we demonstrate that hESCs cultured in unconditioned medium (UM) are subjected to high levels of BMP signaling activity, which is reduced in CM. The BMP antagonist noggin synergizes with basic fibroblast growth factor (bFGF) to repress BMP signaling and sustain undifferentiated proliferation of hESCs in the absence of fibroblasts or CM. These findings suggest a basic difference in the self-renewal mechanism between mouse and human ES cells and simplify the culture of hESCs.
- Published
- 2005
- Full Text
- View/download PDF
66. Derivation and Chondrogenic Commitment of Human Embryonic Stem Cell-Derived Mesenchymal Progenitors
- Author
-
Rosa M. Guzzo, Ren-He Xu, Jason D. Gibson, and Hicham Drissi
- Subjects
Directed differentiation ,Cellular differentiation ,Mesenchymal stem cell ,Hemangioblast ,Anatomy ,Biology ,Progenitor cell ,Stem cell ,Embryonic stem cell ,Stem cell transplantation for articular cartilage repair ,Cell biology - Abstract
The induction of human embryonic stem cells to a mesenchymal-like progenitor population constitutes a developmentally relevant approach for efficient directed differentiation of human embryonic stem (hES) cells to the chondrogenic lineage. The initial enrichment of a hemangioblast intermediate has been shown to yield a replenishable population of highly purified progenitor cells that exhibit the typical mesenchymal stem cell (MSC) surface markers as well as the capacity for multilineage differentiation to bone, fat, and cartilage. Herein, we provide detailed methodologies for the derivation and characterization of potent mesenchymal-like progenitors from hES cells and describe in vitro assays for bone morphogenetic protein (BMP)-2-mediated differentiation to the chondrogenic lineage.
- Published
- 2015
- Full Text
- View/download PDF
67. BMP4 initiates human embryonic stem cell differentiation to trophoblast
- Author
-
Xin Chen, Ren-He Xu, Thomas P. Zwaka, Dong S. Li, James A. Thomson, Gregory C. Addicks, Clay Glennon, and Rui Li
- Subjects
Cellular differentiation ,Biomedical Engineering ,Trophoblast ,Bioengineering ,Biology ,Bone morphogenetic protein ,Applied Microbiology and Biotechnology ,Embryonic stem cell ,Cell biology ,Transplantation ,medicine.anatomical_structure ,Bone morphogenetic protein 4 ,Cell culture ,embryonic structures ,Immunology ,medicine ,Molecular Medicine ,Stem cell ,Biotechnology - Abstract
The excitement and controversy surrounding the potential role of human embryonic stem (ES) cells in transplantation therapy have often overshadowed their potentially more important use as a basic research tool for understanding the development and function of human tissues. Human ES cells can proliferate without a known limit and can form advanced derivatives of all three embryonic germ layers. What is less widely appreciated is that human ES cells can also form the extra-embryonic tissues that differentiate from the embryo before gastrulation. The use of human ES cells to derive early human trophoblast is particularly valuable, because it is difficult to obtain from other sources and is significantly different from mouse trophoblast. Here we show that bone morphogenetic protein 4 (BMP4), a member of the transforming growth factor-beta (TGF-beta) superfamily, induces the differentiation of human ES cells to trophoblast. DNA microarray, RT-PCR, and immunoassay analyses demonstrate that the differentiated cells express a range of trophoblast markers and secrete placental hormones. When plated at low density, the BMP4-treated cells form syncytia that express chorionic gonadotrophin (CG). These results underscore fundamental differences between human and mouse ES cells, which differentiate poorly, if at all, to trophoblast. Human ES cells thus provide a tool for studying the differentiation and function of early human trophoblast and could provide a new understanding of some of the earliest differentiation events of human postimplantation development.
- Published
- 2002
- Full Text
- View/download PDF
68. Neural inhibition by c-Jun as a synergizing factor in bone morphogenetic protein 4 signaling
- Author
-
D Yan, James M. Phang, Jay M. Mei, Ying Peng, Ren-He Xu, Hsiang-Fu Kung, and X.-P Li
- Subjects
Embryo, Nonmammalian ,Proto-Oncogene Proteins c-jun ,Bone morphogenetic protein 8A ,Receptors, Cell Surface ,Bone Morphogenetic Protein 4 ,Xenopus Proteins ,Biology ,Bone morphogenetic protein 2 ,Xenopus laevis ,HMGB Proteins ,Animals ,Receptors, Growth Factor ,Bone morphogenetic protein receptor ,RNA, Messenger ,Neural Cell Adhesion Molecules ,Embryonic Induction ,Neurons ,SOXB1 Transcription Factors ,Stem Cells ,General Neuroscience ,Gene Expression Regulation, Developmental ,Nuclear Proteins ,Bone morphogenetic protein 10 ,Cell Differentiation ,Neural Inhibition ,Bone Morphogenetic Protein Receptors ,DNA-Binding Proteins ,Transcription Factor AP-1 ,Bone morphogenetic protein 7 ,Bone morphogenetic protein 6 ,Bone morphogenetic protein 5 ,Epidermal Cells ,GDF6 ,Bone Morphogenetic Proteins ,Cancer research ,Female ,Epidermis ,Signal Transduction ,Transcription Factors - Abstract
The transcription factor, activator protein 1 (AP-1) complexes (c-Jun and c-Fos heterodimers) has been shown to interact with transforming growth factor beta signaling in mammalian cells and Drosophila embryo. Here we show that c-Jun alone is involved in the anti-neuralizing activity of bone morphogenetic protein 4, a transforming growth factor beta superfamily member, in Xenopus neurogenesis. Co-injection of mRNAs encoding c-jun and a dominant negative bone morphogenetic protein receptor completely inhibits dominant negative bone morphogenetic protein receptor-induced neuralization and reverses the epidermal fate in the animal cap. Surprisingly, a dominant negative c-Jun does not induce neural tissue in the animal cap, but it synergizes with dominant negative bone morphogenetic protein receptor for neural induction. Temporal analysis using a dexamethasone-inducible c-Jun shows that exogenous c-Jun activity must be turned on before or at stage 11 to fulfill the anti-neuralizing effect. Neural inhibition by c-Jun does not occur until stage 13 suggesting that c-Jun probably acts by suppressing neural maintenance rather than neural initiation. This is also supported by the fact that c-Jun does not inhibit expression of the neural-initializing gene Zic-r1 but the neural cofactor Sox2, and that ectopic expression of Sox2 attenuates the anti-neuralizing effect of c-Jun. Finally, we display that the c-Jun effect is enhanced by an auto-regulatory loop between c-Jun and bone morphogenetic protein. These studies suggest that c-Jun/AP-1 is a converging point in both the fibroblast growth factor and transforming growth factor beta signaling pathways. Based on our findings, we propose that c-Jun synergizes with bone morphogenetic protein 4 signaling to inhibit neural development in Xenopus ectoderm.
- Published
- 2002
- Full Text
- View/download PDF
69. Establishment of human cell type-specific iPS cells with enhanced chondrogenic potential
- Author
-
Archana Sanjay, Vanessa Scanlon, Ren-He Xu, Rosa M. Guzzo, and Hicham Drissi
- Subjects
Cancer Research ,Cell type ,Induced Pluripotent Stem Cells ,Cell Differentiation ,Cell Biology ,Biology ,Fibroblasts ,Fetal Blood ,Cell biology ,Cell Line ,Kruppel-Like Factor 4 ,Cartilage ,Chondrocytes ,SOX2 ,Cell culture ,KLF4 ,Immunology ,Leukocytes, Mononuclear ,Humans ,Stem cell ,Induced pluripotent stem cell ,Reprogramming ,Cell potency ,Chondrogenesis - Abstract
The propensity of induced pluripotent stem (iPS) cells to differentiate into specific lineages may be influenced by a number of factors, including the selection of the somatic cell type used for reprogramming. Herein we report the generation of new iPS cells, which we derived from human articular chondrocytes and from cord blood mononucleocytes via lentiviral-mediated delivery of Oct4, Klf4, Sox2, and cMyc. Molecular, cytochemical, and cytogenic analyses confirmed the acquisition of hallmark features of pluripotency, as well as the retention of normal karyotypes following reprogramming of both the human articular chondrocytes (AC) and the cord blood (CB) cells. In vitro and in vivo functional analyses formally established the pluripotent differentiation capacity of all cell lines. Chondrogenic differentiation assays comparing iPS cells derived from AC, CB, and a well established dermal fibroblast cell line (HDFa-Yk26) identified enhanced proteoglycan-rich matrix formation and cartilage-associated gene expression from AC-derived iPS cells. These findings suggest that the tissue of origin may impact the fate potential of iPS cells for differentiating into specialized cell types, such as chondrocytes. Thus, we generated new cellular tools for the identification of inherent features driving high chondrogenic potential of reprogrammed cells.
- Published
- 2014
70. Histone acetylation is a checkpoint in FGF-stimulated mesoderm induction
- Author
-
D. Sredni, Keiko Ozato, Satoshi Yamagoe, Ren-He Xu, Gerald L. Princler, Jing Fan, Hsiang-Fu Kung, Donghong Yan, and Ying Peng
- Subjects
Histone deacetylase 5 ,Trichostatin A ,HDAC11 ,Histone deacetylase 2 ,Histone H2A ,medicine ,Histone deacetylase ,SAP30 ,Biology ,HDAC4 ,Molecular biology ,Developmental Biology ,medicine.drug - Abstract
We have previously demonstrated that the transcription factor, AP-1 (c-jun/c-fos heterodimer), mediates fibroblast growth factor (FGF) signaling during mesoderm induction in Xenopus embryo. In the present studies, we show that histone acetylation is involved in FGF-mediated signaling leading to mesoderm induction. Histone acetylation is a dynamic process regulated by the activities of two histone-modifying enzymes, the histone acetyltransferase(s) and histone deacetylase(s) (HDACs). We found that basal and FGF-regulated activator protein 1 (AP-1) activity in Xenopus embryo is markedly reduced by treatment of trichostatin A (TSA), a specific inhibitor of HDAC. However, activity of another transcription factor, NFκB, is enhanced by TSA treatment. AP-1-mediated mesoderm induction in the animal caps is dramatically suppressed by TSA at a dose-dependent manner. This suppression can be rescued by ectopic expression of HDAC3 at early stage. Finally, we found that histone acetylation in animal caps is inhibited by FGF whereas enhanced by TSA (as a control). Therefore, we propose that histone acetylation is a checkpoint for transduction of the FGF/AP-1 signals to induce mesoderm. Published 2000 Wiley-Liss, Inc.
- Published
- 2000
- Full Text
- View/download PDF
71. Loss of cell adhesion in Xenopus laevis embryos mediated by the cytoplasmic domain of XLerk, an erythropoietin-producing hepatocellular ligand
- Author
-
Ren-He Xu, Jaebong Kim, Lisa D. Chong, Teri L Jones, Hsiang-Fu Kung, and Ira O. Daar
- Subjects
Mesoderm ,Basic fibroblast growth factor ,Xenopus ,Xenopus Proteins ,Ligands ,Fibroblast growth factor ,Xenopus laevis ,chemistry.chemical_compound ,Cell Adhesion ,medicine ,Animals ,Receptor ,Cell adhesion ,Erythropoietin ,Membrane Glycoproteins ,Multidisciplinary ,biology ,Erythropoietin - physiology ,Erythropoietin-producing hepatocellular (Eph) receptor ,Xenopus laevis - embryology - physiology ,Biological Sciences ,biology.organism_classification ,Cell Adhesion - physiology ,Molecular biology ,Cell biology ,medicine.anatomical_structure ,Membrane Glycoproteins - physiology ,chemistry ,Ectopic expression - Abstract
The erythropoietin-producing hepatocellular (Eph) family of ligands and receptors has been implicated in the control of axon guidance and the segmental restriction of cells during embryonic development. In this report, we show that ectopic expression of XLerk, a Xenopus homologue of the murine Lerk-2 (ephrin-B1) transmembrane ligand, causes dissociation of Xenopus embryonic blastomeres by the mid-blastula transition. Moreover, a mutant that lacks the extracellular receptor binding domain can induce this phenotype. The carboxyl-terminal 19 amino acids of the cytoplasmic domain of XLerk are necessary but not sufficient to induce cellular dissociation. Basic fibroblast growth factor, but not activin, can rescue both the loss of cell adhesion and mesoderm induction in ectodermal explants expressing XLerk. Collectively, these results show that the cytoplasmic domain of XLerk has a signaling function that is important for cell adhesion, and fibroblast growth factor signaling modulates this function., published_or_final_version
- Published
- 1998
- Full Text
- View/download PDF
72. Epigenetic regulation of miR-302 by JMJD1C inhibits neural differentiation of human embryonic stem cells
- Author
-
Xiaofang Wang, Jianle Wang, Ren-He Xu, Jung Woo Park, and Hicham Drissi
- Subjects
Jumonji Domain-Containing Histone Demethylases ,Cellular differentiation ,Basic fibroblast growth factor ,Bone morphogenetic protein ,Biochemistry ,Cell Line ,Epigenesis, Genetic ,chemistry.chemical_compound ,Transforming Growth Factor beta ,Histone methylation ,Humans ,Promoter Regions, Genetic ,Molecular Biology ,reproductive and urinary physiology ,Embryonic Stem Cells ,Neurons ,Gene knockdown ,COUP Transcription Factor I ,biology ,urogenital system ,Cell Differentiation ,Oxidoreductases, N-Demethylating ,Cell Biology ,Transforming growth factor beta ,Embryonic stem cell ,Molecular biology ,Cell biology ,MicroRNAs ,chemistry ,embryonic structures ,Bone Morphogenetic Proteins ,biology.protein ,Fibroblast Growth Factor 2 ,Signal transduction ,biological phenomena, cell phenomena, and immunity ,Signal Transduction ,Developmental Biology - Abstract
It has been recently reported that the regulatory circuitry formed by OCT4, miR-302, and NR2F2 controls both pluripotency and neural differentiation of human embryonic stem cells (hESCs). We show here that JMJD1C, a histone 3 lysine 9 (H3K9) demethylase expressed in hESCs, directly interacts with this circuitry. hESCs with stable knockdown of JMJD1C remain pluripotent while having reduced miR-302 expression, decreased BMP signaling, and enhanced TGFβ signaling. JMJD1C binds to the miR-302 promoter and reduces H3K9 methylation. Withdrawal of basic fibroblast growth factor (bFGF) from the culture induces neural differentiation of the knockdown, but not the control, cells within 3 days, accompanied by elevated NR2F2 expression. This can be attenuated with miR-302 mimics or an H3K9 methytransferase inhibitor. Together, our findings suggest that JMJD1C represses neural differentiation of hESCs at least partially by epigenetically sustaining miR-302 expression and that JMJD1C knockdown is sufficient to trigger neural differentiation upon withdrawal of exogenous bFGF.
- Published
- 2013
73. ◾ Stem Cells and Tissue Regeneration
- Author
-
Kristen Martins-Taylor, Ren-He Xu, Xiaofang Wang, and Xue-Jun Li
- Subjects
Stem cell ,Biology ,Cell biology - Published
- 2013
- Full Text
- View/download PDF
74. The role of BMP-4 and GATA-2 in the induction and differentiation of hematopoietic mesoderm in Xenopus laevis
- Author
-
Hsiang-Fu Kung, Atsushi Suzuki, Mitsugu Maéno, Naoto Ueno, Leonard I. Zon, Paul E. Mead, Clair Kelley, and Ren-He Xu
- Subjects
Mesoderm ,medicine.medical_specialty ,animal structures ,Immunology ,Cell Biology ,Hematology ,Germ layer ,Biology ,Biochemistry ,FGF and mesoderm formation ,Cell biology ,GDF1 ,medicine.anatomical_structure ,Endocrinology ,Epiblast ,Internal medicine ,embryonic structures ,medicine ,Paraxial mesoderm ,Aorta-gonad-mesonephros ,NODAL - Abstract
Vertebrate embryonic blood formation is regulated by factors that participate in dorsal-ventral patterning and mesoderm induction. The GATA-binding transcription factors are required for normal hematopoiesis and are expressed during gastrulation when ventral mesoderm (VM) is induced to form blood. Based on the recent demonstration that bone morphogenetic protein (BMP-4) is a potent ventralizing factor and inducer of hematopoietic tissue, we hypothesized that GATA-2 could be induced or activated by BMP-4. Here we demonstrate that BMP-4 can stimulate GATA-2 expression, and that expression of a dominant negative BMP-4 receptor can suppress GATA-2 induction by BMP-4 in ventral mesoderm. Over-expression of GATA-2 in ventral mesoderm leads to increased globin production and forced expression of GATA-2 in primitive ectoderm adjacent to ventral mesoderm also stimulates globin expression. Our results suggest that BMP-4 and GATA-2 can function in two adjacent germ layers, mesoderm and ectoderm, to participate in blood cell formation during embryogenesis.
- Published
- 1996
- Full Text
- View/download PDF
75. Involvement of Ras/Raf/AP-1 in BMP-4 signaling during Xenopus embryonic development
- Author
-
Mitsugu Maéno, Nancy H. Colburn, Naoto Ueno, Ren-He Xu, Hsiang-Fu Kung, Zigang Dong, Atsushi Suzuki, D. Sredni, and Jaebong Kim
- Subjects
Mesoderm ,animal structures ,Proto-Oncogene Proteins c-jun ,Xenopus ,Mutant ,Protein Serine-Threonine Kinases ,Models, Biological ,Proto-Oncogene Proteins p21(ras) ,Proto-Oncogene Proteins ,medicine ,Animals ,Erythropoiesis ,Luciferase ,Embryonic Induction ,Multidisciplinary ,biology ,Embryogenesis ,Proteins ,Embryo ,biology.organism_classification ,Molecular biology ,Proto-Oncogene Proteins c-raf ,Transcription Factor AP-1 ,medicine.anatomical_structure ,Bone morphogenetic protein 4 ,Bone Morphogenetic Proteins ,Mutation ,Tissue Transplantation ,embryonic structures ,Signal transduction ,Signal Transduction ,Research Article - Abstract
Previously, we elucidated the role of bone morphogenetic protein 4 (BMP-4) in the dorsal-ventral patterning of the Xenopus embryo by using a dominant negative mutant of the BMP-4 receptor (DN-BR). The present paper describes the involvement of Ras, Raf, and activator protein 1 (AP-1) in BMP-4 signaling during Xenopus embryonic development. The AP-1 activity was determined by injecting an AP-1-dependent luciferase reporter gene into two-cell-stage Xenopus embryos and measuring the luciferase activity at various developmental stages. We found that injection of BMP-4 mRNA increased AP-1 activity, whereas injection of DN-BR mRNA inhibited AP-1 activity. Similar inhibitory effects were seen with injection of mRNAs encoding dominant negative mutants of c-Ha-Ras, c-Raf, or c-Jun. These results suggest that the endogenous AP-1 activity is regulated by BMP-4/Ras/Raf/Jun signals. We next investigated the effects of Ras/Raf/AP-1 signals on the biological functions of BMP-4. DN-BR-induced dorsalization of the embryo, revealed by the formation of a secondary body axis or dorsalization of the ventral mesoderm explant analyzed by histological and molecular criteria, was significantly reversed by coinjection of [Val12]Ha-Ras, c-Raf, or c-Jun mRNA. Furthermore, the BMP-4-stimulated erythroid differentiation in the ventral mesoderm was substantially inhibited by coinjection with the dominant negative c-Ha-Ras, c-Raf, or c-Jun mutant. Our results suggest the involvement of Ras/Raf/AP-1 in the BMP-4 signaling pathway.
- Published
- 1996
- Full Text
- View/download PDF
76. The protective role of the immunomodulator AS101 against chemotherapy-induced alopecia studies on human and animal models
- Author
-
Adi Shani, Benjamin Sredni, Rivka Gal, Raphael Catane, Israel Bruderman, Karl L Mettinger, Jeremy Shapira, Ren-He Xu, Bella Kaufman, Yona Kalechman, Uzi Gafter, Michael Albeck, John Whisnant, and Thomas Tichler
- Subjects
Cancer Research ,Chemotherapy ,business.industry ,medicine.medical_treatment ,Alpha (ethology) ,Cancer ,Pharmacology ,medicine.disease ,Carboplatin ,Clinical trial ,chemistry.chemical_compound ,Oncology ,chemistry ,Toxicity ,Immunology ,Chemoprotective ,medicine ,business ,Etoposide ,medicine.drug - Abstract
The immunomodulator AS101 has been demonstrated to exhibit radioprotective and chemoprotective effects in mice. Following phase-I studies, preliminary results from phase-II clinical trials on non-small-cell-lung-cancer patients showed a reduction in the severity of alopecia in patients treated with AS101 in combination with chemotherapy. To further substantiate these findings, the present study was extended to include 58 patients treated either with the optimal dose of 3 mg/m2 AS101 combined with carboplatin and VP-16, or with chemotherapy alone. As compared with patients treated with chemotherapy alone, there was a significant decrease in the level of alopecia in patients receiving the combined therapy. The newly developed rat model was used to elucidate the protective mechanism involved in this effect. We show that significant prevention of chemotherapy-induced alopecia is obtained in rats treated with Ara-C combined with AS101, administered i.p. or s.c. or applied topically to the dorsal skin. We show that this protection by AS101 is mediated by macrophage-derived factors induced by AS101. Protection by AS101 can be ascribed, at least in part, to IL-1, since treatment of rats with IL-1 RA largely abrogated the protective effect of AS101. Moreover, we demonstrate that in humans there is an inverse correlation between the grade of alopecia and the increase in IL-1 alpha. In addition, protection by AS101 could be related to PGE2 secretion, since injection of indomethacin before treatment with AS101 and Ara-C partly abrogated the protective effect of AS101. To assess the ability of AS101 to protect against chemotherapy-induced alopecia, phase-II clinical trials have been initiated with cancer patients suffering from various malignancies.
- Published
- 1996
- Full Text
- View/download PDF
77. Stem cell-based bone repair
- Author
-
Yurong, Fei, Ren-He, Xu, and Marja M, Hurley
- Subjects
Review Article - Abstract
To accelerate bone repair, one strategy is to deliver the cells that make bone. The current review focuses on stem cell-based bone repair. Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) can self-renew unlimitedly and differentiate into the bone forming cells – osteoblasts. Scientists have been actively investigating culture conditions to stably and efficiently induce differentiation of these stem cells into osteoblasts. However, ESCs have the issues of ethnics, immune response and both ESCs and iPSCs have tumorigenic potential. In contrast, bone marrow stromal/stem cells (BMSCs) hold great potential to enhance bone formation. Use of BMSCs can avoid the ethical issues and can obviate the immune response problem. However, BMSCs are a rare population with limited self-renewal ability and their differentiation ability decreases in elderly individuals. Considering the unlimited self-renewal ability, it is promising to develop protocols to differentiate ESCs into osteoblasts faithfully and efficiently. It is important to eliminate undifferentiated ESCs or iPSCs because of their tumorigenic potential. Therefore, future studies need to identify BMSCs specific cell surface markers since the cell surface markers utilized currently are not specific to BMSCs. Future studies also need to enhance the osteogenic potential without using viral vectors for transgene delivery to eliminate the risk of tumor generation.
- Published
- 2012
78. Concise review: Genomic stability of human induced pluripotent stem cells
- Author
-
Kristen Martins-Taylor and Ren-He Xu
- Subjects
Genetics ,DNA Copy Number Variations ,Induced Pluripotent Stem Cells ,Cell Biology ,Biology ,Embryonic stem cell ,Polymorphism, Single Nucleotide ,Genomic Instability ,Cell Line ,Karyotyping ,Molecular Medicine ,Humans ,Copy-number variation ,DNA microarray ,Induced pluripotent stem cell ,Exome ,Virtual karyotype ,Embryonic Stem Cells ,In Situ Hybridization, Fluorescence ,Developmental Biology ,Epigenomics ,Comparative genomic hybridization - Abstract
The usefulness of human induced pluripotent stem cells (hiPSCs) in research and therapeutic applications highly relies on their genomic integrity and stability. Many laboratories including ours have addressed this concern by comparing genomic (at both karyotypic and subkaryotypic levels) and epigenomic abnormalities of hiPSC lines (derived via either DNA- or non-DNA-based methods), as well as human embryonic stem cell lines during long-term culture. A variety of methods have been used for this purpose, such as karyotyping and fluorescent in situ hybridization to detect karyotypic abnormalities, array-based comparative genomic hybridization to detect copy number variations (CNVs), single-nucleotide polymorphism-based microarrays to detect both CNVs and loss of heterozygosity, analysis of integration sites in the genome, and whole genome sequencing for protein-coding exome and DNA methylome profiling. Here, we summarize the progresses in this dynamically evolving field and also discuss how the findings apply to the study and application of hiPSCs.
- Published
- 2011
79. Inhibition of B16 melanoma metastasis by the immunomodulator AS101
- Author
-
Ren-He Xu, H Kung, Yona Kalechman, Benjamin Sredni, and Michael Albeck
- Subjects
Cancer Research ,Tumor suppressor gene ,Oncogene ,Cell growth ,Cell ,Cell cycle ,Biology ,In vitro ,medicine.anatomical_structure ,Oncology ,Apoptosis ,medicine ,Cancer research ,Tumor necrosis factor alpha - Abstract
The immunomodulator ammonium trichloro (dioxyethylene-O-O')tellurate (AS101) has previously been found to induce secretion of various cytokines in mouse and human, which include interleukin-l, interleukin-2, colony-stimulating factor, interferon-gamma, tumor necrosis factor, etc. It also protects mice from lethal and sublethal effects of chemotherapy and irradiation. The present studies were designed to evaluate its effect on pulmonary metastasis following intravenous (i.v.) injection of mouse B16 melanoma cells on day 0 of the experiment. AS101, given 10 mu g/mouse intraperitoneally in 7 daily injections starting the day before B16 cell infusion (day -1) led to a significant inhibition by 60%. When B16 cells were pretreated with AS101 for 24 h before injection, the lung metastases were further reduced by subsequent AS101 treatment of the tumor-loaded mice. In mice that had been depleted of natural killer (NK) cells using anti-asialo-GM1 antisera, AS101 was deprived of its stimulatory effect on the NK activity. The inhibition by AS101 on the B16 lung metastasis was also profoundly reduced by the antisera. Moreover, in vitro treatment of B16 cells with AS101 resulted in suppression of the cell growth in a semisolid culture. This was accompanied by an inhibition of the DNA synthesis and a dephosphorylation/activation of the retinoblastoma susceptibility protein (RE), a tumor suppressor gene product, in the B16 cells. Taken together, these data suggest that AS101 possesses an anti-metastatic activity, which probably involves two mechanisms: the stimulation of the host NK cell activity and the inhibition of the tumor cell proliferation.
- Published
- 2011
80. Specification of region-specific neurons including forebrain glutamatergic neurons from human induced pluripotent stem cells
- Author
-
Ren-He Xu, Alexander C. Lichtler, Shuning Zhan, Lixia Yue, Min Guo, Xue-Jun Li, Mark S. Kronenberg, Zheng Zhang, Jung Woo Park, Kristen Martins-Taylor, Xiaofang Wang, Hui Zeng, Hui Xia Liu, and Fang Ping Chen
- Subjects
Cellular differentiation ,Induced Pluripotent Stem Cells ,Glutamic Acid ,lcsh:Medicine ,Biology ,03 medical and health sciences ,Glutamatergic ,Prosencephalon ,0302 clinical medicine ,Directed differentiation ,Humans ,Progenitor cell ,Induced pluripotent stem cell ,lcsh:Science ,030304 developmental biology ,Neurons ,0303 health sciences ,Multidisciplinary ,Reverse Transcriptase Polymerase Chain Reaction ,lcsh:R ,Cell Differentiation ,Anatomy ,Neuroscience/Neurodevelopment ,Flow Cytometry ,Immunohistochemistry ,Embryonic stem cell ,Developmental Biology/Stem Cells ,Developmental Biology/Neurodevelopment ,Electrophysiology ,Neuroepithelial cell ,Forebrain ,lcsh:Q ,Neuroscience ,030217 neurology & neurosurgery ,Research Article - Abstract
Background Directed differentiation of human induced pluripotent stem cells (hiPSC) into functional, region-specific neural cells is a key step to realizing their therapeutic promise to treat various neural disorders, which awaits detailed elucidation. Methodology/Principal Findings We analyzed neural differentiation from various hiPSC lines generated by others and ourselves. Although heterogeneity in efficiency of neuroepithelial (NE) cell differentiation was observed among different hiPSC lines, the NE differentiation process resembles that from human embryonic stem cells (hESC) in morphology, timing, transcriptional profile, and requirement for FGF signaling. NE cells differentiated from hiPSC, like those from hESC, can also form rostral phenotypes by default, and form the midbrain or spinal progenitors upon caudalization by morphogens. The rostrocaudal neural progenitors can further mature to develop forebrain glutamatergic projection neurons, midbrain dopaminergic neurons, and spinal motor neurons, respectively. Typical ion channels and action potentials were recorded in the hiPSC-derived neurons. Conclusions/Significance Our results demonstrate that hiPSC, regardless of how they were derived, can differentiate into a spectrum of rostrocaudal neurons with functionality, which supports the considerable value of hiPSC for study and treatment of patient-specific neural disorders.
- Published
- 2010
81. Human embryonic stem cell derivation, maintenance, and differentiation to trophoblast
- Author
-
Ge, Lin, Kristen, Martins-Taylor, and Ren-He, Xu
- Subjects
Male ,Gene Expression Profiling ,Cell Culture Techniques ,Cell Differentiation ,Cell Separation ,Embryo, Mammalian ,Immunohistochemistry ,Trophoblasts ,Drug Combinations ,Mice ,Blastocyst ,Pregnancy ,Culture Media, Conditioned ,Animals ,Humans ,Female ,Proteoglycans ,Collagen ,Laminin ,Biomarkers ,Cells, Cultured ,Embryonic Stem Cells - Abstract
Since the first report of derivation of human embryonic stem cell (hESC) lines in 1998, many progresses have been achieved to reliably and efficiently derive, maintain, and differentiate this therapeutically promising cell type. This chapter introduces some basic and widely recognized methods that we use in our hESC core laboratory. Specifically, it includes methods for (1) deriving hESC lines without using enzyme and antibody to isolate the inner cell mass; (2) sustaining hESC self-renewal under feeder-dependent, feeder-conditioned, and defined conditions as well as pluripotency validation and quality control assays; and (3) inducing hESC differentiation to trophoblast with BMP4.
- Published
- 2010
82. Human Embryonic Stem Cell Derivation, Maintenance, and Differentiation to Trophoblast
- Author
-
Ge Lin, Kristen Martins-Taylor, and Ren-He Xu
- Subjects
Cell type ,biology ,Trophoblast ,Embryo ,equipment and supplies ,Embryonic stem cell ,Cell biology ,Gene expression profiling ,medicine.anatomical_structure ,Laminin ,embryonic structures ,biology.protein ,medicine ,Inner cell mass ,biological phenomena, cell phenomena, and immunity ,Core laboratory ,reproductive and urinary physiology - Abstract
Since the first report of derivation of human embryonic stem cell (hESC) lines in 1998, many progresses have been achieved to reliably and efficiently derive, maintain, and differentiate this therapeutically promising cell type. This chapter introduces some basic and widely recognized methods that we use in our hESC core laboratory. Specifically, it includes methods for (1) deriving hESC lines without using enzyme and antibody to isolate the inner cell mass; (2) sustaining hESC self-renewal under feeder-dependent, feeder-conditioned, and defined conditions as well as pluripotency validation and quality control assays; and (3) inducing hESC differentiation to trophoblast with BMP4.
- Published
- 2010
- Full Text
- View/download PDF
83. Inhibition of Caspase-mediated Anoikis Is Critical for Basic Fibroblast Growth Factor-sustained Culture of Human Pluripotent Stem Cells*
- Author
-
Ge Lin, Hui Zeng, Ren-He Xu, Xiaofang Wang, and Kristen Martins-Taylor
- Subjects
Pluripotent Stem Cells ,Programmed cell death ,Cell Survival ,medicine.medical_treatment ,Basic fibroblast growth factor ,Biology ,Fibroblast growth factor ,Biochemistry ,Models, Biological ,Cell Line ,chemistry.chemical_compound ,Mice ,Molecular Basis of Cell and Developmental Biology ,Insulin-Like Growth Factor II ,medicine ,Animals ,Humans ,Anoikis ,Induced pluripotent stem cell ,Molecular Biology ,Cells, Cultured ,Cell Proliferation ,Growth factor ,Cell Cycle ,Cell Biology ,Embryonic stem cell ,Cell biology ,Enzyme Activation ,chemistry ,Cell culture ,Caspases ,Fibroblast Growth Factor 2 - Abstract
Apoptosis and proliferation are two dynamically and tightly regulated processes that together maintain the homeostasis of renewable tissues. Anoikis is a subtype of apoptosis induced by detachment of adherent cells from the extracellular matrix. By using the defined mTeSR1 medium and collecting freshly detached cells, we found here that human pluripotent stem (PS) cells including embryonic stem (ES) cells and induced pluripotent stem cells are subject to constant anoikis in culture, which is escalated in the absence of basic fibroblast growth factor (bFGF). Withdrawal of bFGF also promotes apoptosis and differentiation of the remaining adherent cells without affecting their cell cycle progression. Insulin-like growth factor 2 (IGF2) has previously been reported to act downstream of FGF signaling to support self-renewal of human ES cells. However, we found that IGF2 cannot substitute bFGF in the TeSR1-supported culture, although endogenous IGF signaling is required to sustain self-renewal of human ES cells. On the other hand, all of the bFGF withdrawal effects observed here can be markedly prevented by the caspase inhibitor z-VAD-FMK. We further demonstrated that the bFGF-repressed anoikis is dependent on activation of ERK and AKT and associated with inhibition of Bcl-2-interacting mediator of cell death and the caspase-ROCK1-myosin signaling. Anoikis is independent of pre-detachment apoptosis and differentiation of the cells. Because previous studies of human PS cells have been focused on attached cells, our findings revealed a neglected role of bFGF in sustaining self-renewal of human PS cells: preventing them from anoikis via inhibition of caspase activation.
- Published
- 2009
84. NANOG is a Direct Target of TGFβ/Activin Mediated SMAD Signaling in Human ES Cells
- Author
-
Sierra H. Root, Tori L. Sampsell-Barron, Feng Gu, James A. Thomson, Jessica Antosiewicz-Bourget, Junying Yu, Ren-He Xu, Ron Stewart, Ruthann M. Peck, Guangjin Pan, and Shulan Tian
- Subjects
Transcriptional Activation ,Homeobox protein NANOG ,animal structures ,Cellular differentiation ,Rex1 ,Dioxoles ,Smad2 Protein ,SMAD ,Biology ,Article ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,SOX2 ,Genetics ,Humans ,SMAD binding ,Smad3 Protein ,Promoter Regions, Genetic ,Lymphotoxin-alpha ,Embryonic Stem Cells ,reproductive and urinary physiology ,Cell Proliferation ,030304 developmental biology ,Homeodomain Proteins ,0303 health sciences ,Nanog Homeobox Protein ,Cell Differentiation ,Cell Biology ,STEMCELL ,Molecular biology ,Recombinant Proteins ,Cell biology ,Fibroblast Growth Factors ,Benzamides ,Bone Morphogenetic Proteins ,embryonic structures ,Molecular Medicine ,biological phenomena, cell phenomena, and immunity ,030217 neurology & neurosurgery ,Protein Binding ,Signal Transduction - Abstract
Self-renewal of human embryonic stem cells (ESCs) is promoted by FGF and TGFbeta/Activin signaling, and differentiation is promoted by BMP signaling, but how these signals regulate genes critical to the maintenance of pluripotency has been unclear. Using a defined medium, we show here that both TGFbeta and FGF signals synergize to inhibit BMP signaling; sustain expression of pluripotency-associated genes such as NANOG, OCT4, and SOX2; and promote long-term undifferentiated proliferation of human ESCs. We also show that both TGFbeta- and BMP-responsive SMADs can bind with the NANOG proximal promoter. NANOG promoter activity is enhanced by TGFbeta/Activin and FGF signaling and is decreased by BMP signaling. Mutation of putative SMAD binding elements reduces NANOG promoter activity to basal levels and makes NANOG unresponsive to BMP and TGFbeta signaling. These results suggest that direct binding of TGFbeta/Activin-responsive SMADs to the NANOG promoter plays an essential role in sustaining human ESC self-renewal.
- Published
- 2008
85. In vitro induction of trophoblast from human embryonic stem cells
- Author
-
Ren-He, Xu
- Subjects
Immunoassay ,Stem Cells ,Cell Culture Techniques ,Cell Differentiation ,Fibroblasts ,Flow Cytometry ,Trophoblasts ,Mice ,Culture Media, Conditioned ,Bone Morphogenetic Proteins ,Animals ,Humans ,Placental Hormones ,Biomarkers ,Cells, Cultured ,Oligonucleotide Array Sequence Analysis - Abstract
Human embryonic stem (ES) cells can proliferate without a known limit and can form advanced derivatives of all three embryonic germ layers. What is less widely appreciated is that human ES cells can also form the extra-embryonic tissues that differentiate from the embryo before gastrulation. The use of human ES cells to derive early human trophoblast is particularly valuable, because it is difficult to obtain from other sources and is significantly different from mouse trophoblast. Here we describe a method by using bone morphogenetic protein (BMP)4, a member of the transforming growth factor (TGF)-beta superfamily, to induce the differentiation of human ES cells to trophoblast. Immunoassays (as well as DNA microarray and reverse-transcription polymerase chain reaction analyses--data not shown) demonstrate that the differentiated cells express a range of trophoblast markers and secrete placental hormones. When plated at low density, the BMP4-treated cells form syncytia that express chorionic gonadotrophin (CG). This technique underscores fundamental differences between human and mouse ES cells, which differentiate poorly, if at all, to trophoblast. Human ES cells thus provide a tool for studying the differentiation and function of early human trophoblast and could provide a new understanding of some of the earliest differentiation events of human postimplantation development.
- Published
- 2005
86. In Vitro Induction of Trophoblast from Human Embryonic Stem Cells
- Author
-
Ren-He Xu
- Subjects
Homeobox protein NANOG ,Syncytium ,medicine.anatomical_structure ,Cellular differentiation ,embryonic structures ,medicine ,Trophoblast ,Stem cell ,Biology ,Bone morphogenetic protein ,Embryonic stem cell ,Cell biology ,Adult stem cell - Abstract
Human embryonic stem (ES) cells can proliferate without a known limit and can form advanced derivatives of all three embryonic germ layers. What is less widely appreciated is that human ES cells can also form the extra-embryonic tissues that differentiate from the embryo before gastrulation. The use of human ES cells to derive early human trophoblast is particularly valuable, because it is difficult to obtain from other sources and is significantly different from mouse trophoblast. Here we describe a method by using bone morphogenetic protein (BMP)4, a member of the transforming growth factor (TGF)-beta superfamily, to induce the differentiation of human ES cells to trophoblast. Immunoassays (as well as DNA microarray and reverse-transcription polymerase chain reaction analyses--data not shown) demonstrate that the differentiated cells express a range of trophoblast markers and secrete placental hormones. When plated at low density, the BMP4-treated cells form syncytia that express chorionic gonadotrophin (CG). This technique underscores fundamental differences between human and mouse ES cells, which differentiate poorly, if at all, to trophoblast. Human ES cells thus provide a tool for studying the differentiation and function of early human trophoblast and could provide a new understanding of some of the earliest differentiation events of human postimplantation development.
- Published
- 2005
- Full Text
- View/download PDF
87. Trophoblast differentiation from embryonic stem cells
- Author
-
Thaddeus G. Golos and Ren-He Xu
- Published
- 2004
- Full Text
- View/download PDF
88. Maternal cold inducible RNA binding protein is required for embryonic kidney formation in Xenopus laevis
- Author
-
Ren-He Xu, Ying Peng, K.H.H. Kwok, Hsiang-Fu Kung, Kin-Hang Kok, Peter C. W. Fung, David Tay, and Marie C.M. Lin
- Subjects
Embryo, Nonmammalian ,Cell division ,Transcription, Genetic ,Biophysics ,Retinoic acid ,Xenopus ,Biology ,Xenopus Proteins ,Kidney ,Biochemistry ,Cold inducible RNA binding protein ,Pronephros ,Xbra ,Evolution, Molecular ,chemistry.chemical_compound ,Xenopus laevis ,Suppression, Genetic ,Structural Biology ,Sense (molecular biology) ,Genetics ,Morphogenesis ,Animals ,Protein Isoforms ,Abnormalities, Multiple ,Cloning, Molecular ,Morphogenetic movement ,Molecular Biology ,Phylogeny ,Reverse Transcriptase Polymerase Chain Reaction ,Embryogenesis ,RNA-Binding Proteins ,Cell Biology ,biology.organism_classification ,Embryonic stem cell ,Molecular biology ,Phenotype ,chemistry ,cDNA cloning ,Female ,Cell Division - Abstract
We cloned a major isoform of Xenopus homologue of cold inducible RNA binding protein (CIRP), XCIRP-1. XCIRP-1 was neither cold inducible nor essential for cell division during early embryonic development. Suppression of XCIRP-1 dose dependently produced tailbuds with deformations of the brain and internal organs. The defects were XCIRP-1 specific as they could be rescued by sense transcript. Suppression of XCIRP-1 also disrupted the morphogenetic migration of the C3 blastomeres (lineaged to become the embryonic kidney, the pronephros). In animal cap explants, depletion of XCIRP-1 inhibited activin/retinoic acid induced expressions of pronephros related Xlim-1 and WT1 genes. These results suggest that XCIRP-1 is required for the specification and morphogenetic lineage migration of the pronephros.
- Published
- 2000
89. Characterization of the functionally related sites in the neural inducing gene noggin
- Author
-
Ke Lan, Lu Xie, Caiping Ren, Weidong Liu, Jianling Shi, Hsiang-Fu Kung, Jing Fan, Kaitai Yao, Ren-He Xu, Ying Peng, Zhiwei He, Xiangling Feng, and Liangguo Xu
- Subjects
Mesoderm ,animal structures ,Xenopus ,Biophysics ,Ectoderm ,Bone Morphogenetic Protein 4 ,Biology ,Xenopus Proteins ,Bone morphogenetic protein ,Biochemistry ,Nervous System ,medicine ,Animals ,Noggin ,Molecular Biology ,Body Patterning ,Genetics ,Embryonic Induction ,integumentary system ,Proteins ,Cell Biology ,biology.organism_classification ,Recombinant Proteins ,Cell biology ,Bone morphogenetic protein 5 ,medicine.anatomical_structure ,GDF6 ,embryonic structures ,Bone Morphogenetic Proteins ,Mutagenesis, Site-Directed ,Neural cell adhesion molecule ,Carrier Proteins - Abstract
Previously we have shown that blocking bone morphogenetic protein (BMP) receptor signaling by a dominant negative BMP receptor causes neurogenesis in Xenopus animal caps (ACs), whereas the physiological neural inducer noggin acts as a homodimer physically binding to BMP-4 and disrupting its signaling at the ligand level. The present study attempted to elucidate the relationship between the structure and function of noggin. By replacing some cysteine residues with serine residues through a site-directed mutagenesis strategy, we generated three noggin mutants, C145S, C205S, and C(218, 220, 222)S (3CS). Although mRNAs encoded by these mutants were translated as efficiently as wild-type (WT) noggin mRNA, they behaved differently when expressed in vivo. Expression of WT noggin or C205S in Xenopus ACs converted the explants (prospective ectoderm) into neural tissue, indicated by the neural-like morphology and expression of the pan neural marker NCAM in the ACs. In contrast, ACs expressing C145S or 3CS sustained an epidermal fate like the control caps. Similar results were observed in the mesoderm where C205S (but not C145S and 3CS) displayed dorsalizing activity as well as WT noggin. Altogether, our results suggest that Cys145 alone or Cys(218, 220, 222) as a whole in noggin protein is required for the biological activities of noggin, probably participating in the dimerization of noggin with BMP-4 or itself.
- Published
- 2000
90. Opposite effects of FGF and BMP-4 on embryonic blood formation: roles of PV.1 and GATA-2
- Author
-
Mae-Ja Park, Ying Peng, Yoo-Seok Hwang, Katherine Tidman Ault, Ren-He Xu, D. Sredni, Jaebong Kim, and Hsiang-Fu Kung
- Subjects
Mesoderm ,medicine.medical_specialty ,Bone Morphogenetic Protein 4 ,Biology ,In Vitro Techniques ,Xenopus Proteins ,Fibroblast growth factor ,Models, Biological ,FGF and mesoderm formation ,03 medical and health sciences ,Xenopus laevis ,Internal medicine ,medicine ,Animals ,Cell Lineage ,Erythropoiesis ,Transcription factor ,Molecular Biology ,030304 developmental biology ,Homeodomain Proteins ,0303 health sciences ,Lateral plate mesoderm ,030302 biochemistry & molecular biology ,Muscle, Smooth ,Cell Biology ,Embryonic stem cell ,Receptors, Fibroblast Growth Factor ,Cell biology ,DNA-Binding Proteins ,Fibroblast Growth Factors ,GATA2 Transcription Factor ,Haematopoiesis ,medicine.anatomical_structure ,Endocrinology ,embryonic structures ,Bone Morphogenetic Proteins ,Developmental Biology ,Signal Transduction ,Transcription Factors - Abstract
In adult vertebrates, fibroblast growth factor (FGF) synergizes with many hematopoietic cytokines to stimulate the proliferation of hematopoietic progenitors. In vertebrate development, the FGF signaling pathway is important in the formation of some derivatives of ventroposterior mesoderm. However, the function of FGF in the specification of the embryonic erythropoietic lineage has remained unclear. Here we address the role of FGF in the specification of the erythropoietic lineage in the Xenopus embryo. We report that ventral injection of embryonic FGF (eFGF) mRNA at as little as 10 pg at the four-cell stage suppresses ventral blood island (VBI) formation, whereas expression of the dominant negative form of the FGF receptor in the lateral mesoderm, where physiologically no blood tissue is formed, results in a dramatic expansion of the VBI. Similar results were observed in isolated ventral marginal zones and animal caps. Bone morphogenetic protein-4 (BMP-4) is known to induce erythropoiesis in the Xenopus embryo. Therefore, we examined how the BMP-4 and FGF signaling pathways might interact in the decision of ventral mesoderm to form blood. We observed that eFGF inhibits BMP-4-induced erythropoiesis by differentially regulating expression of the BMP-4 downstream effectors GATA-2 and PV.1. GATA-2, which stimulates erythropoiesis, is suppressed by FGF. PV.1, which we demonstrate to inhibit blood development, is enhanced by FGF. Additionally, PV.1 and GATA-2 negatively regulate transcription of each other. Thus, BMP-4 induces two transcription factors which have opposing effects on blood development. The FGF and BMP-4 signaling pathways interact to regulate the specification of the erythropoietic lineage.
- Published
- 1999
91. Xenopus CRMP-2 is an early response gene to neural induction
- Author
-
Ren-He Xu, Marianne Subleski, Hsiang-Fu Kung, Tohru Kamata, Ira O. Daar, and Terry D. Copeland
- Subjects
Nervous system ,Embryo, Nonmammalian ,Transcription, Genetic ,Xenopus ,Molecular Sequence Data ,Embryonic Development ,Nerve Tissue Proteins ,Biology ,Nervous System ,Polymerase Chain Reaction ,Cellular and Molecular Neuroscience ,Gene expression ,Transcriptional regulation ,medicine ,Animals ,Amino Acid Sequence ,Noggin ,Molecular Biology ,In Situ Hybridization ,Sequence Homology, Amino Acid ,Gene Expression Regulation, Developmental ,Semaphorin-3A ,Gastrula ,biology.organism_classification ,Molecular biology ,medicine.anatomical_structure ,Bone Morphogenetic Proteins ,Intercellular Signaling Peptides and Proteins ,Chordin ,Neural plate ,Neural development ,Signal Transduction - Abstract
A neural specific protein, CRMP-2 (for Collapsin Response Mediator Protein-2), is considered to mediate collapsin-induced growth cone collapse during neural development. We have isolated the Xenopus homologue of the CRMP-2 (XCRMP-2) cDNA and studied the expression of XCRMP-2 mRNA and protein during neural induction. Induction of XCRMP-2 mRNA and protein expression, like N-CAM, occurred at the midgastrula stage and increased through early neural developmental stages. Whole mount in situ hybridization demonstrated that expression of XCRMP-2 mRNA was localized in neural tissues such as the neural plate and tube at early stages, while its expression in the brain, spinal cord, and eyes was observed at later stages. Immunostaining of Xenopus embryos with the antibody against CRMP-2 also showed that the protein was specifically expressed in the neural tissues at early stages. XCRMP-2 expression was induced by neural inducers such as noggin and chordin which antagonize a neural inhibitor, BMP4. A dominant negative BMP receptor also induced XCRMP-2 expression, suggesting that transcription of XCRMP-2 gene was negatively regulated by the BMP4 signaling. These results indicate that expression of XCRMP-2 is an early response marking neural commitment, and that transcriptional control of XCRMP-2 gene, is one of the targets of BMP4 signaling.
- Published
- 1998
92. The immunomodulator AS101 restores T(H1) type of response suppressed by Babesia rodhaini in BALB/c mice
- Author
-
Benjamin Sredni, Ji-Ping Da, F. Shalit, Yona Kalechman, Ren-He Xu, Gideon Strassmann, Michael Albeck, Ami Vonsover, Hanna Rosenblatt-Bin, Avraham Klein, and M. Huberman
- Subjects
Male ,Antiparasitic ,medicine.drug_class ,Immunology ,Biology ,BALB/c ,Interferon-gamma ,Mice ,Downregulation and upregulation ,Adjuvants, Immunologic ,Immunity ,Babesiosis ,medicine ,Parasite hosting ,Macrophage ,Animals ,Secretion ,RNA, Messenger ,Messenger RNA ,Mice, Inbred BALB C ,Ethylenes ,Th1 Cells ,biology.organism_classification ,Molecular biology ,Interleukin-12 ,Cytokines ,Interleukin-1 - Abstract
The immunomodulator AS101 has been previously shown to confer protection upon BALB/c mice infected with the intraerythrocytic parasite Babesia rodhaini (B. rodhaini). The present study focuses on the effect of AS101 administration on the acute phase of babesial infection where T helper cell subset patterns-TH1/TH2-were assessed in heavily infected mice. Secretion of cytokines of the TH1 subset (IL-2, IFN-gamma, IL-12) and of the TH2 subset (IL-10, IL-4) as well as TGF-beta was measured following the administration of AS101 2 weeks before parasite infection. Our results demonstrate that the parasites suppress IL-2 protein and IL-12 mRNA and that AS101 upregulates their secretion: IL-2, 8 u/ml vs 34 u/ml, respectively; IFN-gamma protein, 2370 pg/ml vs 4777 pg/ml, respectively. Conversely, babesial infection results in the upregulation of IL-10 and IL-4 proteins and TGF-beta transcripts, whereas AS101 downregulates their production: IL-10, 1800 pg/ml vs 360 pg/ml, respectively; IL-4, 58.3 pg/ml vs 24.5 pg/ml, respectively. A possible escape mechanism induced by B. rodhaini is suggested, starting with IL-10 inhibition of macrophage activities leading to a suppression of the TH1 response and of IL-2 in particular. It is therefore possible that AS101 may protect infected mice by activating cellular-mediated immunity and concurrently balancing the TH subset responses. It is suggested that AS101 may be effective as an antiparasitic drug.
- Published
- 1998
93. Mesoderm induction by heterodimeric AP-1 (c-Jun and c-Fos) and its involvement in mesoderm formation through the embryonic fibroblast growth factor/Xbra autocatalytic loop during the early development of Xenopus embryos
- Author
-
Jaebong Kim, Jih-Jing Lin, Ren-He Xu, and Hsiang-Fu Kung
- Subjects
Fetal Proteins ,Mesoderm ,animal structures ,Embryo, Nonmammalian ,Proto-Oncogene Proteins c-jun ,Xenopus ,Embryonic Development ,Fibroblast growth factor ,Biochemistry ,FGF and mesoderm formation ,Catalysis ,Xbra ,Transactivation ,Xenopus laevis ,Culture Techniques ,medicine ,Animals ,Molecular Biology ,biology ,NF-kappa B ,Cell Biology ,biology.organism_classification ,Molecular biology ,DNA-Binding Proteins ,Transcription Factor AP-1 ,medicine.anatomical_structure ,embryonic structures ,Mesoderm formation ,RNA ,Fibroblast Growth Factor 2 ,NODAL ,T-Box Domain Proteins ,Proto-Oncogene Proteins c-fos ,Transcription Factors - Abstract
We have previously demonstrated the involvement of AP-1/Jun in fibroblast growth factor (FGF) signaling by loss-of-function assay (Dong, Z., Xu, R.-H., Kim, J., Zhan, S.-N., Ma, W.-Y., Colburn, N. H., and Kung, H. (1996) J. Biol. Chem. 271, 9942-9946). Further investigations by gain-of-function are reported in this study. AP-1 transactivation activity was increased by the treatment of animal cap explants with FGF. Ectopic overexpression of two components of AP-1 (c-jun and c-fos together, but not alone) produced posteriorized embryos and induced mesoderm formation in animal cap explants, indicating that both AP-1 heterodimers are required for mesoderm induction. Since Ras/AP-1 functions downstream of FGF signaling, we then tested the involvement of Ras/AP-1 in mesoderm maintenance mediated by embryonic FGF/Xbra using dominant-negative mutants. Mesoderm maintenance mediated by embryonic FGF/Xbra was blocked by dominant-negative mutants of Ras/AP-1, and AP-1 enhanced the expression of Xbra. Further studies demonstrated the inhibition of Ras/AP-1-mediated mesoderm formation by dominant-negative mutants of the FGF receptor and Xbra. These results indicate that Ras/AP-1 and FGF/Xbra signals are involved in the mesoderm maintenance machinery and mesoderm formation through the synergistic action of the diversified signal pathways derived from the FGF/Xbra autocatalytic loop.
- Published
- 1998
94. Studies on the role of fibroblast growth factor signaling in neurogenesis using conjugated/aged animal caps and dorsal ectoderm-grafted embryos
- Author
-
Ren-He Xu, Masanori Taira, D. Sredni, Jaebong Kim, and Hsiang-Fu Kung
- Subjects
Central Nervous System ,medicine.medical_specialty ,animal structures ,Basic fibroblast growth factor ,Embryonic Development ,Ectoderm ,Receptors, Cell Surface ,Bone Morphogenetic Protein 4 ,Biology ,Xenopus Proteins ,Fibroblast growth factor ,chemistry.chemical_compound ,Xenopus laevis ,Internal medicine ,Notochord ,medicine ,Animals ,Receptors, Growth Factor ,Neuroectoderm ,General Neuroscience ,Neurogenesis ,Bone Morphogenetic Protein Receptors ,Articles ,Receptors, Fibroblast Growth Factor ,Cell biology ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Bone morphogenetic protein 4 ,Neural Crest ,embryonic structures ,Bone Morphogenetic Proteins ,Fibroblast Growth Factor 2 ,Neural development ,Signal Transduction - Abstract
Basic fibroblast growth factor (bFGF) has been shown to induce neural fate in dissociated animal cap (AC) cells or in AC explants cultured in low calcium and magnesium concentrations. However, long-term disclosure of the cap may cause diffusion of the secreted molecule bone morphogenetic protein 4 (BMP-4), a neural inhibitor present in the AC. This may contribute to the subsequent neurogenesis induced by bFGF. Here we used conjugated and aged blastula AC to avoid diffusion of endogenous molecules from the AC. Unlike noggin, bFGF failed to induce neural tissue in this system. However, it enhanced neuralization elicited by a dominant negative BMP receptor (DN-BR) that inhibits the BMP-4 signaling. Posterior neural markers were turned on by bFGF in AC expressing DN-BR or chordin. Blocking the endogenous FGF signal with a dominant negative FGF receptor (XFD) mainly inhibited development of posterior neural tissue in neuralized ACs. Thesein vitrostudies were confirmedin vivoin embryos grafted with XFD-expressing ACs in the place of neuroectoderm. Expression of some regional neural markers was inhibited, although markers for muscle and posterior notochord were still detectable in the grafted embryos, suggesting that XFD specifically affected neurogenesis but not the dorsal mesoderm. The use of thesein vitroandin vivomodel systems provides new evidence that FGF, although unable to initiate neurogenesis on its own, is required for neural induction as well as for posteriorization.
- Published
- 1997
95. AP-1/jun is required for early Xenopus development and mediates mesoderm induction by fibroblast growth factor but not by activin
- Author
-
Ren-He Xu, Nancy H. Colburn, Zigang Dong, Hsiang-Fu Kung, Shu Ning Zhan, Wei-Ya Ma, and Jaebong Kim
- Subjects
Mesoderm ,animal structures ,Fibroblast growth factor ,Biochemistry ,FGF and mesoderm formation ,Receptor tyrosine kinase ,GDF1 ,Xenopus laevis ,TGF beta signaling pathway ,medicine ,Animals ,Inhibins ,Molecular Biology ,Embryonic Induction ,biology ,Cell Biology ,Molecular biology ,Activins ,Fibroblast Growth Factors ,Transcription Factor AP-1 ,medicine.anatomical_structure ,embryonic structures ,Mesoderm formation ,biology.protein ,NODAL ,Signal Transduction - Abstract
In Xenopus, normal mesoderm formation depends on signaling through the fibroblast growth factor (FGF) tyrosine kinase receptor. An important signaling pathway from receptor tyrosine kinases involves Ras/Raf/MAP kinase. However, the downstream pathway that occurs in the nucleus to finally trigger gene expression for mesoderm formation remains unknown. We report here that a high level of activator protein-1 (AP-1)-dependent transcriptional activity is detected during the early development of Xenopus embryos. Injection of a dominant negative mutant jun (DNM-jun or TAM67) RNA into the two-cell stage embryos inhibited endogenous AP-1 activity and blocked normal embryonic development with severe posterior truncation in tadpoles. The inhibition of AP-1 activity and the phenotypic change induced by TAM67 was rescued by co-injection of wild-type c-jun RNA, but not by the control beta-galactosidase RNA. The FGF-stimulated mesoderm induction was markedly inhibited in animal cap explants from the embryos injected with TAM67. Activin induction of mesoderm, on the other hand, was normal in the embryos injected with TAM67 RNA. These findings suggest that AP-1 mediates FGF, but not activin, receptor signaling during mesoderm induction and the AP-1/Jun is a key signaling molecule in the development of posterior structure.
- Published
- 1996
96. A dominant negative bone morphogenetic protein 4 receptor causes neuralization in Xenopus ectoderm
- Author
-
Masanori Taira, Shuning Zhan, D. Sredni, Jaebong Kim, Ren-He Xu, and Hsiang-Fu Kung
- Subjects
medicine.medical_specialty ,animal structures ,Embryo, Nonmammalian ,Molecular Sequence Data ,Biophysics ,Xenopus ,Gene Expression ,Ectoderm ,Receptors, Cell Surface ,Biochemistry ,Nervous System ,Polymerase Chain Reaction ,Embryonic and Fetal Development ,Xenopus laevis ,Internal medicine ,medicine ,Animals ,Receptors, Growth Factor ,RNA, Messenger ,Noggin ,Growth Substances ,Molecular Biology ,DNA Primers ,biology ,Neuroectoderm ,Base Sequence ,Proteins ,Cell Differentiation ,Cell Biology ,Bone Morphogenetic Protein Receptors ,biology.organism_classification ,Cell biology ,Gastrulation ,Endocrinology ,medicine.anatomical_structure ,Bone morphogenetic protein 4 ,GDF6 ,embryonic structures ,Bone Morphogenetic Proteins ,biology.protein ,Follistatin - Abstract
Injection of DN-BR mRNA encoding a dominant negative type I receptor for bone morphogenetic protein 4 (BMP4) converted prospective ectoderm into neural tissue in Xenopus animal cap explants, in the absence of expression of mesodermal marker genes. The injected caps expressed a general neural marker NCAM and the forebrain marker opsin. Coinjection of wild-type BMP4 receptor mRNA completely reversed the neuralization by DN-BR. No expression of known neuralizing factors, i.e., noggin and follistatin, was detected in the DN-BR-injected animal caps. Furthermore, neuralization elicited by noggin or 3m, a LIM domain mutant of Xlim-1, was substantially inhibited by co-injection of BMP4 mRNA. Since BMP4 is expressed in the prospective ectoderm during gastrulation, our results suggest that the ventralizing factor BMP4 acts also as a physiological inhibitor of neuralization in the development of Xenopus ectoderm.
- Published
- 1995
97. FGF, BMP, and TGFβ signaling together control human embryonic stem cell fates
- Author
-
Ren-He Xu
- Subjects
R-SMAD ,Cell Biology ,Biology ,Fibroblast growth factor ,Molecular Biology ,Embryonic stem cell ,BMPR2 ,Cell biology - Published
- 2008
- Full Text
- View/download PDF
98. In Vitro Induction of Trophoblast from Human Embryonic Stem Cells.
- Author
-
Walker, John M., Soares, Michael J., Hunt, Joan S., and Ren-He Xu
- Abstract
Human embryonic stem (ES) cells can proliferate without a known limit and can form advanced derivatives of all three embryonic germ layers. What is less widely appreciated is that human ES cells can also form the extra-embryonic tissues that differentiate from the embryo before gastrulation. The use of human ES cells to derive early human trophoblast is particularly valuable, because it is difficult to obtain from other sources and is significantly different from mouse trophoblast. Here we describe a method by using bone morphogenetic protein (BMP)4 , a member of the transforming growth factor (TGF)-β superfamily, to induce the differentiation of human ES cells to trophoblast. Immunoassays (as well as DNA microarray and reverse-transcription polymerase chain reaction analyses—data not shown) demonstrate that the differentiated cells express a range of trophoblast markers and secrete placental hormones. When plated at low density, the BMP4-treated cells form syncytia that express chorionic gonadotrophin (CG). This technique underscores fundamental differences between human and mouse ES cells, which differentiate poorly, if at all, to trophoblast. Human ES cells thus provide a tool for studying the differentiation and function of early human trophoblast and could provide a new understanding of some of the earliest differentiation events of human postimplantation development. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
99. Role of DNMT3B in the regulation of early neural and neural crest specifiers.
- Author
-
Martins-Taylor, Kristen, Schroeder, Diane I., LaSalle, Janine M., Lalande, Marc, and Ren-He Xu
- Published
- 2012
- Full Text
- View/download PDF
100. Recurrent copy number variations in human induced pluripotent stem cells.
- Author
-
Martins-Taylor, Kristen, Nisler, Benjamin S., Taapken, Seth M., Compton, Tiwanna, Crandall, Leann, Montgomery, Karen Dyer, Lalande, Marc, and Ren-He Xu
- Subjects
LETTERS to the editor ,PLURIPOTENT stem cells - Abstract
A letter to the editor is presented about the recurrent copy number variations that can be observed in human induced pluripotent stem cells.
- Published
- 2011
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.