Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Cell ; 135(7): 1287-98, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-19109897

ABSTRACT

Embryonic stem (ES) cells have been available from inbred mice since 1981 but have not been validated for other rodents. Failure to establish ES cells from a range of mammals challenges the identity of cultivated stem cells and our understanding of the pluripotent state. Here we investigated derivation of ES cells from the rat. We applied molecularly defined conditions designed to shield the ground state of authentic pluripotency from inductive differentiation stimuli. Undifferentiated cell lines developed that exhibited diagnostic features of ES cells including colonization of multiple tissues in viable chimeras. Definitive ES cell status was established by transmission of the cell line genome to offspring. Derivation of germline-competent ES cells from the rat paves the way to targeted genetic manipulation in this valuable biomedical model species. Rat ES cells will also provide a refined test-bed for functional evaluation of pluripotent stem cell-derived tissue repair and regeneration.


Subject(s)
Blastocyst Inner Cell Mass/cytology , Embryonic Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Line , Chimera , Female , Fibroblast Growth Factors/antagonists & inhibitors , Glycogen Synthase Kinases/antagonists & inhibitors , Male , Mice , Mice, SCID , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Rats , Rats, Inbred F344 , Rats, Inbred Strains
2.
Cell ; 135(7): 1299-310, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-19109898

ABSTRACT

Rats have important advantages over mice as an experimental system for physiological and pharmacological investigations. The lack of rat embryonic stem (ES) cells has restricted the availability of transgenic technologies to create genetic models in this species. Here, we show that rat ES cells can be efficiently derived, propagated, and genetically manipulated in the presence of small molecules that specifically inhibit GSK3, MEK, and FGF receptor tyrosine kinases. These rat ES cells express pluripotency markers and retain the capacity to differentiate into derivatives of all three germ layers. Most importantly, they can produce high rates of chimerism when reintroduced into early stage embryos and can transmit through the germline. Establishment of authentic rat ES cells will make possible sophisticated genetic manipulation to create models for the study of human diseases.


Subject(s)
Blastocyst/cytology , Embryonic Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Chimera , Epigenesis, Genetic , Female , Fibroblast Growth Factors/antagonists & inhibitors , Glycogen Synthase Kinases/antagonists & inhibitors , Male , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Rats , Rats, Inbred Strains , Signal Transduction
3.
J Cell Sci ; 130(22): 3809-3817, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28982712

ABSTRACT

TFCP2L1 is a transcription factor that is crucial for self-renewal of mouse embryonic stem cells (mESCs). How TFCP2L1 maintains the pluripotent state of mESCs, however, remains unknown. Here, we show that knockdown of Tfcp2l1 in mESCs induces the expression of endoderm, mesoderm and trophectoderm markers. Functional analysis of mutant forms of TFCP2L1 revealed that TFCP2L1 depends on its N-terminus and CP2-like domain to maintain the undifferentiated state of mESCs. The N-terminus of TFCP2L1 is mainly associated with the suppression of mesoderm and trophectoderm differentiation, while the CP2-like domain is closely related to the suppression of endoderm commitment. Further studies showed that MTA1 directly interacts with TFCP2L1 and is indispensable for the TFCP2L1-mediated self-renewal-promoting effect and endoderm-inhibiting action. TFCP2L1-mediated suppression of mesoderm and trophectoderm differentiation, however, seems to be due to downregulation of Lef1 expression. Our study thus provides an expanded understanding of the function of TFCP2L1 and the pluripotency regulation network of ESCs.


Subject(s)
Lymphoid Enhancer-Binding Factor 1/metabolism , Mouse Embryonic Stem Cells/physiology , Repressor Proteins/physiology , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Self Renewal , Cells, Cultured , Ectoderm/cytology , Mesoderm/cytology , Mice , Trans-Activators
4.
Hepatology ; 67(3): 1041-1055, 2018 03.
Article in English | MEDLINE | ID: mdl-29024000

ABSTRACT

Notch signaling plays an emerging role in the regulation of immune cell development and function during inflammatory response. Activation of the ras homolog gene family member A/Rho-associated protein kinase (ROCK) pathway promotes leukocyte accumulation in tissue injury. However, it remains unknown whether Notch signaling regulates ras homolog gene family member A/ROCK-mediated immune responses in liver ischemia and reperfusion (IR) injury. This study investigated intracellular signaling pathways regulated by Notch receptors in the IR-stressed liver and in vitro. In a mouse model of IR-induced liver inflammatory injury, we found that mice with myeloid-specific Notch1 knockout showed aggravated hepatocellular damage, with increased serum alanine aminotransferase levels, hepatocellular apoptosis, macrophage/neutrophil trafficking, and proinflammatory mediators compared to Notch1-proficient controls. Unlike in the controls, myeloid Notch1 ablation diminished hairy and enhancer of split-1 (Hes1) and augmented c-Jun N-terminal kinase (JNK)/stress-activated protein kinase-associated protein 1 (JSAP1), JNK, ROCK1, and phosphatase and tensin homolog (PTEN) activation in ischemic livers. Disruption of JSAP1 in myeloid-specific Notch1 knockout livers improved hepatocellular function and reduced JNK, ROCK1, PTEN, and toll-like receptor 4 activation. Moreover, ROCK1 knockdown inhibited PTEN and promoted Akt, leading to depressed toll-like receptor 4. In parallel in vitro studies, transfection of lentivirus-expressing Notch1 intracellular domain promoted Hes1 and inhibited JSAP1 in lipopolysaccharide-stimulated bone marrow-derived macrophages. Hes1 deletion enhanced JSAP1/JNK activation, whereas clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9-mediated JSAP1 knockout diminished ROCK1/PTEN and toll-like receptor 4 signaling. CONCLUSION: Myeloid Notch1 deficiency activates the ras homolog gene family member A/ROCK pathway and exacerbates hepatocellular injury by inhibiting transcriptional repressor Hes1 and inducing scaffold protein JSAP1 in IR-triggered liver inflammation; our findings underscore the crucial role of the Notch-Hes1 axis as a novel regulator of innate immunity-mediated inflammation and imply the therapeutic potential for the management of organ IR injury in transplant recipients. (Hepatology 2018;67:1041-1055).


Subject(s)
Liver/pathology , Receptor, Notch1/genetics , Reperfusion Injury/metabolism , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Animals , Apoptosis/genetics , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Immunohistochemistry , Liver/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , Reactive Oxygen Species , Real-Time Polymerase Chain Reaction , Receptor, Notch1/metabolism , Signal Transduction , rhoA GTP-Binding Protein
5.
Dev Biol ; 431(2): 272-281, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28943339

ABSTRACT

ß-catenin-mediated signaling has been extensively studied in regard to its role in the regulation of human embryonic stem cells (hESCs). However, the results are controversial and the mechanism by which ß-catenin regulates the hESC fate remains unclear. Here, we report that ß-catenin and γ-catenin are functionally redundant in mediating hESC adhesion and are required for embryoid body formation, but both genes are dispensable for hESC maintenance, as the undifferentiated state of ß-catenin and γ-catenin double deficient hESCs can be maintained. Overexpression of ß-catenin induces rapid hESC differentiation. Functional assays revealed that TCF1 plays a crucial role in hESC differentiation mediated by ß-catenin. Forced expression of TCF1, but not other LEF1/TCF family members, resulted in hESC differentiation towards the definitive endoderm. Conversely, knockdown of TCF1 or inhibition of the interaction between TCF1 and ß-catenin delayed hESC exit from pluripotency. Furthermore, we demonstrated that GATA6 plays a predominant role in TCF1-mediated hESC differentiation. Knockdown of GATA6 completely eliminated the effect of TCF1, while forced expression of GATA6 induced hESC differentiation. Our data thus reveal more detailed mechanisms for ß-catenin in regulating hESC fate decisions and will expand our understanding of the self-renewal and differentiation circuitry in hESCs.


Subject(s)
Cell Lineage , GATA6 Transcription Factor/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Lymphoid Enhancer-Binding Factor 1/metabolism , Signal Transduction , beta Catenin/metabolism , Cell Adhesion , Cell Differentiation , Cell Self Renewal , Desmoplakins/metabolism , Endoderm/cytology , Humans , Transcription, Genetic , Up-Regulation , gamma Catenin
6.
J Biol Chem ; 292(41): 17121-17128, 2017 10 13.
Article in English | MEDLINE | ID: mdl-28848051

ABSTRACT

The transcription factor Gbx2 (gastrulation brain homeobox 2) is a direct target of the LIF/STAT3 signaling pathway, maintains mouse embryonic stem cell (mESC) self-renewal, and facilitates mouse epiblast stem cell (mEpiSC) reprogramming to naïve pluripotency. However, the mechanism by which Gbx2 mediates its effects on pluripotency remains unknown. Here, using an RNA-Seq approach, we identified Klf4 (Kruppel-like factor 4) as a direct target of Gbx2. Functional studies indicated that Klf4 mediates the self-renewal-promoting effects of Gbx2, because knockdown of Klf4 expression abrogated the ability of Gbx2 to maintain the undifferentiated state of mESCs. We also found that Gbx2 largely depends on Klf4 to reprogram mEpiSCs to a mESC-like state. In summary, our study has uncovered a mechanism by which Gbx2 maintains and induces naïve pluripotency. These findings expand our understanding of the pluripotency control network and may inform the development of culture conditions for improved ESC maintenance and differentiation.


Subject(s)
Cellular Reprogramming/physiology , Homeodomain Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Mouse Embryonic Stem Cells/metabolism , Animals , Cell Line , Gene Knockdown Techniques , Homeodomain Proteins/genetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Mice , Mouse Embryonic Stem Cells/cytology
7.
J Cell Sci ; 129(2): 269-76, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26598557

ABSTRACT

Activation of leukemia inhibitor factor (LIF)-Stat3 or Wnt/ß-catenin signaling promotes mouse embryonic stem cell (mESC) self-renewal. A myriad of downstream targets have been identified in the individual signal pathways, but their common targets remain largely elusive. In this study, we found that the LIF-Stat3 and Wnt/ß-catenin signaling pathways converge on Sp5 to promote mESC self-renewal. Forced Sp5 expression can reproduce partial effects of Wnt/ß-catenin signaling but mimics most features of LIF-Stat3 signaling to maintain undifferentiated mESCs. Moreover, Sp5 is able to convert mouse epiblast stem cells into a naïve pluripotent state. Thus, Sp5 is an important component of the regulatory network governing mESC naïve pluripotency.


Subject(s)
Leukemia Inhibitory Factor/metabolism , Mouse Embryonic Stem Cells/metabolism , STAT3 Transcription Factor/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation , Cell Self Renewal , Cells, Cultured , Gene Expression , Mice , Transcriptional Activation , Wnt Signaling Pathway
9.
EMBO J ; 32(19): 2548-60, 2013 Oct 02.
Article in English | MEDLINE | ID: mdl-23942238

ABSTRACT

Mouse embryonic stem cell (mESC) self-renewal can be maintained by activation of the leukaemia inhibitory factor (LIF)/signal transducer and activator of transcription 3 (Stat3) signalling pathway or dual inhibition (2i) of glycogen synthase kinase 3 (Gsk3) and mitogen-activated protein kinase kinase (MEK). Several downstream targets of the pathways involved have been identified that when individually overexpressed can partially support self-renewal. However, none of these targets is shared among the involved pathways. Here, we show that the CP2 family transcription factor Tfcp2l1 is a common target in LIF/Stat3- and 2i-mediated self-renewal, and forced expression of Tfcp2l1 can recapitulate the self-renewal-promoting effect of LIF or either of the 2i components. In addition, Tfcp2l1 can reprogram post-implantation epiblast stem cells to naïve pluripotent ESCs. Tfcp2l1 upregulates Nanog expression and promotes self-renewal in a Nanog-dependent manner. We conclude that Tfcp2l1 is at the intersection of LIF- and 2i-mediated self-renewal pathways and plays a critical role in maintaining ESC identity. Our study provides an expanded understanding of the current model of ground-state pluripotency.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Repressor Proteins/metabolism , Alkaline Phosphatase/metabolism , Animals , Benzamides/pharmacology , Cell Differentiation/physiology , Cells, Cultured , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Embryonic Stem Cells/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Leukemia Inhibitory Factor/metabolism , Mice , Mice, Inbred C57BL , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Repressor Proteins/genetics , STAT3 Transcription Factor/metabolism
10.
Hepatology ; 64(5): 1683-1698, 2016 11.
Article in English | MEDLINE | ID: mdl-27474884

ABSTRACT

Heat shock transcription factor 1 (HSF1) has been implicated in the differential regulation of cell stress and disease states. ß-catenin activation is essential for immune homeostasis. However, little is known about the role of macrophage HSF1-ß-catenin signaling in the regulation of NLRP3 inflammasome activation during ischemia/reperfusion (I/R) injury (IRI) in the liver. This study investigated the functions and molecular mechanisms by which HSF1-ß-catenin signaling influenced NLRP3-mediated innate immune response in vivo and in vitro. Using a mouse model of IR-induced liver inflammatory injury, we found that mice with a myeloid-specific HSF1 knockout (HSF1M-KO ) displayed exacerbated liver damage based on their increased serum alanine aminotransferase levels, intrahepatic macrophage/neutrophil trafficking, and proinflammatory interleukin (IL)-1ß levels compared to the HSF1-proficient (HSF1FL/FL ) controls. Disruption of myeloid HSF1 markedly increased transcription factor X-box-binding protein (XBP1), NLR family, pyrin domain-containing 3 (NLRP3), and cleaved caspase-1 expression, which was accompanied by reduced ß-catenin activity. Knockdown of XBP1 in HSF1-deficient livers using a XBP1 small interfering RNA ameliorated hepatocellular functions and reduced NLRP3/cleaved caspase-1 and IL-1ß protein levels. In parallel in vitro studies, HSF1 overexpression increased ß-catenin (Ser552) phosphorylation and decreased reactive oxygen species (ROS) production in bone-marrow-derived macrophages. However, myeloid HSF1 ablation inhibited ß-catenin, but promoted XBP1. Furthermore, myeloid ß-catenin deletion increased XBP1 messenger RNA splicing, whereas a CRISPR/CRISPR-associated protein 9-mediated XBP1 knockout diminished NLRP3/caspase-1. CONCLUSION: The myeloid HSF1-ß-catenin axis controlled NLRP3 activation by modulating the XBP1 signaling pathway. HSF1 activation promoted ß-catenin, which, in turn, inhibited XBP1, leading to NLRP3 inactivation and reduced I/R-induced liver injury. These findings demonstrated that HSF1/ß-catenin signaling is a novel regulator of innate immunity in liver inflammatory injury and implied the therapeutic potential for management of sterile liver inflammation in transplant recipients. (Hepatology 2016;64:1683-1698).


Subject(s)
DNA-Binding Proteins/physiology , Inflammasomes/physiology , Liver/blood supply , NLR Family, Pyrin Domain-Containing 3 Protein/physiology , Reperfusion Injury/etiology , Transcription Factors/physiology , beta Catenin/physiology , Animals , Heat Shock Transcription Factors , Mice , Signal Transduction
11.
Nature ; 467(7312): 211-3, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20703227

ABSTRACT

The use of homologous recombination to modify genes in embryonic stem (ES) cells provides a powerful means to elucidate gene function and create disease models. Application of this technology to engineer genes in rats has not previously been possible because of the absence of germline-competent ES cells in this species. We have recently established authentic rat ES cells. Here we report the generation of gene knockout rats using the ES-cell-based gene targeting technology. We designed a targeting vector to disrupt the tumour suppressor gene p53 (also known as Tp53) in rat ES cells by means of homologous recombination. p53 gene-targeted rat ES cells can be routinely generated. Furthermore, the p53 gene-targeted mutation in the rat ES-cell genome can transmit through the germ line via ES-cell rat chimaeras to create p53 gene knockout rats. The rat is the most widely used animal model in biological research. The establishment of gene targeting technology in rat ES cells, in combination with advances in genomics and the vast amount of research data on physiology and pharmacology in this species, now provide a powerful new platform for the study of human disease.


Subject(s)
Embryonic Stem Cells/cytology , Gene Knockout Techniques/methods , Genes, p53 , Rats/genetics , Animals , Base Sequence , Cell Culture Techniques , Embryo, Mammalian/cytology , Female , Germ-Line Mutation , Male , Mice , Molecular Sequence Data , Rats, Inbred F344 , Rats, Sprague-Dawley , Recombination, Genetic
12.
Cell Mol Life Sci ; 72(9): 1741-57, 2015 May.
Article in English | MEDLINE | ID: mdl-25595304

ABSTRACT

Embryonic stem cells (ESCs) can be maintained in culture indefinitely while retaining the capacity to generate any type of cell in the body, and therefore not only hold great promise for tissue repair and regeneration, but also provide a powerful tool for modeling human disease and understanding biological development. In order to fulfill the full potential of ESCs, it is critical to understand how ESC fate, whether to self-renew or to differentiate into specialized cells, is regulated. On the molecular level, ESC fate is controlled by the intracellular transcriptional regulatory networks that respond to various extrinsic signaling stimuli. In this review, we discuss and compare important signaling pathways in the self-renewal and differentiation of mouse, rat, and human ESCs with an emphasis on how these pathways integrate into ESC-specific transcription circuitries. This will be beneficial for understanding the common and conserved mechanisms that govern self-renewal, and for developing novel culture conditions that support ESC derivation and maintenance.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Regulatory Networks , Signal Transduction , Animals , Cell Differentiation , Cell Proliferation , Humans
13.
J Cell Sci ; 126(Pt 5): 1093-8, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23345404

ABSTRACT

Activation of signal transducer and activator of transcription 3 (Stat3) by leukemia inhibitory factor (LIF) maintains mouse embryonic stem cell (mESC) self-renewal and also facilitates reprogramming to ground state pluripotency. Exactly how LIF/Stat3 signaling exerts these effects, however, remains elusive. We identified gastrulation brain homeobox 2 (Gbx2) as a LIF/Stat3 downstream target that, when overexpressed, allows long-term expansion of undifferentiated mESCs in the absence of LIF/Stat3 signaling. Elevated Gbx2 expression also enhanced reprogramming of mouse embryonic fibroblasts to induced pluripotent stem cells. Moreover, overexpression of Gbx2 was sufficient to reprogram epiblast stem cells to ground state ESCs. Our results reveal a novel function of Gbx2 in mESC reprogramming and LIF/Stat3-mediated self-renewal.


Subject(s)
Cellular Reprogramming/physiology , Homeodomain Proteins/metabolism , Leukemia Inhibitory Factor/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factor 3/metabolism , Animals , Cell Line , Cellular Reprogramming/genetics , Homeodomain Proteins/genetics , Mice , Pluripotent Stem Cells/cytology , Reverse Transcriptase Polymerase Chain Reaction
14.
Stem Cells ; 32(5): 1149-60, 2014 May.
Article in English | MEDLINE | ID: mdl-24302476

ABSTRACT

STAT3 can be transcriptionally activated by phosphorylation of its tyrosine 705 or serine 727 residue. In mouse embryonic stem cells (mESCs), leukemia inhibitory factor (LIF) signaling maintains pluripotency by inducing JAK-mediated phosphorylation of STAT3 Y705 (pY705). However, the function of phosphorylated S727 (pS727) in mESCs remains unclear. In this study, we examined the roles of STAT3 pY705 and pS727 in regulating mESC identities, using a small molecule-based system to post-translationally modulate the quantity of transgenic STAT3 in STAT3(-/-) mESCs. We demonstrated that pY705 is absolutely required for STAT3-mediated mESC self-renewal, while pS727 is dispensable, serving only to promote proliferation and optimal pluripotency. S727 phosphorylation is regulated directly by fibroblast growth factor/Erk signaling and crucial in the transition of mESCs from pluripotency to neuronal commitment. Loss of S727 phosphorylation resulted in significantly reduced neuronal differentiation potential, which could be recovered by a S727 phosphorylation mimic. Moreover, loss of pS727 sufficed LIF to reprogram epiblast stem cells to naïve pluripotency, suggesting a dynamic equilibrium of STAT3 pY705 and pS727 in the control of mESC fate.


Subject(s)
Embryonic Stem Cells/metabolism , STAT3 Transcription Factor/metabolism , Serine/metabolism , Tyrosine/metabolism , Amino Acid Substitution , Animals , Benzamides/pharmacology , Blotting, Western , Cell Differentiation/genetics , Cell Proliferation , Cells, Cultured , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Embryonic Stem Cells/cytology , Fluorescent Antibody Technique , Gene Expression , Leukemia Inhibitory Factor/pharmacology , Mice, Knockout , Mice, Transgenic , Nestin/genetics , Nestin/metabolism , Neurons/cytology , Neurons/metabolism , Phosphorylation/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor/genetics , Serine/genetics , Tyrosine/genetics
15.
Nature ; 453(7194): 519-23, 2008 May 22.
Article in English | MEDLINE | ID: mdl-18497825

ABSTRACT

In the three decades since pluripotent mouse embryonic stem (ES) cells were first described they have been derived and maintained by using various empirical combinations of feeder cells, conditioned media, cytokines, growth factors, hormones, fetal calf serum, and serum extracts. Consequently ES-cell self-renewal is generally considered to be dependent on multifactorial stimulation of dedicated transcriptional circuitries, pre-eminent among which is the activation of STAT3 by cytokines (ref. 8). Here we show, however, that extrinsic stimuli are dispensable for the derivation, propagation and pluripotency of ES cells. Self-renewal is enabled by the elimination of differentiation-inducing signalling from mitogen-activated protein kinase. Additional inhibition of glycogen synthase kinase 3 consolidates biosynthetic capacity and suppresses residual differentiation. Complete bypass of cytokine signalling is confirmed by isolating ES cells genetically devoid of STAT3. These findings reveal that ES cells have an innate programme for self-replication that does not require extrinsic instruction. This property may account for their latent tumorigenicity. The delineation of minimal requirements for self-renewal now provides a defined platform for the precise description and dissection of the pluripotent state.


Subject(s)
Embryonic Stem Cells/cytology , Regeneration/physiology , Animals , Benzamides/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , Regeneration/drug effects , STAT3 Transcription Factor/deficiency , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
16.
J Hepatol ; 59(4): 762-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23714159

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) develops in response to chronic hepatic injury. Although induced cell death is regarded as the major component of p53 tumor-suppressive activity, we recently found that sustained p53 activation subsequent to DNA damage promotes inflammation-associated hepatocarcinogenesis. Here we aim at exploring the mechanism linking p53 activation and hepatic inflammation during hepatocarcinogenesis. METHODS: p53(-/-) hepatocytes expressing inducible p53 and primary wild type hepatocytes were treated to induce p53 expression. The supernatants were collected and analyzed for the presence of released inflammatory cytokines. Ethyl pyruvate was used in a rat model of carcinogen-induced hepatocarcinogenesis to examine its effect on p53-dependent chronic hepatic injury, inflammation, and tumorigenesis. RESULTS: Here we show that cytoplasmic translocation and circulating levels of potent inflammatory molecule high-mobility group protein 1 (HMGB1) were greater in wild type rats than in p53(+/-) rats following carcinogen administration. Restoration of p53 expression in p53-null hepatocytes or induction of endogenous p53 in wild type hepatocytes gives rise to the release of HMGB1. Administration of the HMGB1 release inhibitor ethyl pyruvate, which does not affect p53-mediated hepatic apoptosis, substantially prevented carcinogen-induced cirrhosis and tumorigenesis in rat livers. CONCLUSIONS: These results suggest that although p53 is usually regarded as a tumor suppressor, its constant activation can promote pro-tumorigenic inflammation, at least in part, via inducing HMGB1 release. Application of HMGB1 inhibitors when restoring p53 in cancer therapy might protect against pro-tumorigenic effects while leaving p53-mediated clearance of malignant cells intact.


Subject(s)
Genes, p53 , HMGB1 Protein/metabolism , Liver Neoplasms, Experimental/etiology , Animals , Cell Line , Diethylnitrosamine/toxicity , Gene Knockout Techniques , Hepatitis, Chronic/etiology , Hepatitis, Chronic/metabolism , Hepatitis, Chronic/pathology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Rats , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism
17.
Carcinogenesis ; 33(10): 2001-5, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22791818

ABSTRACT

The p53 tumor suppressor gene is highly mutated in human cancers. Individuals who inherit one p53 mutant allele are susceptible to a wide range of tumor types, including breast cancer and sarcoma. We recently generated p53 knockout rats through gene targeting in embryonic stem cells. Here we show that rats homozygous for the null allele are prone to early onset spontaneous sarcomas and lymphoma with high incidence of metastases. Heterozygous rats are also highly predisposed to cancer, but with a delayed onset and a wider spectrum of tumor types compared with homozygotes. Importantly, up to 20% of female heterozygotes developed breast cancer and about 70% of the tumors were positive for estrogen receptor. Exposing p53-deficient rats to a low dose of the carcinogen diethylnitrosamine dramatically decreased the latency for sarcoma development and survival time compared with equivalently treated wild-type rats. These unique features make this knockout line a valuable model for investigating human malignancy and in vivo carcinogenicity of chemicals and therapeutic compounds.


Subject(s)
Gene Knockout Techniques , Genes, p53 , Models, Animal , Alleles , Animals , Breast Neoplasms/genetics , Cell Transformation, Neoplastic/genetics , Female , Heterozygote , Homozygote , Lymphoma/genetics , Quinolines , Rats , Sarcoma/genetics
18.
Curr Opin Nephrol Hypertens ; 20(4): 391-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21546835

ABSTRACT

PURPOSE OF REVIEW: Several advances have been made to manipulate the rat genome in the last 2 years. This review aims to describe these advances in rat genetic manipulations, with an emphasis on their current status and their prospects and applications in the postgenomic era. RECENT FINDINGS: Authentic rat embryonic stem cells were derived in 2008 using the 2i/3i culture system. This led to the generation of the first gene knockout rats via embryonic stem cell-based gene targeting. The development of zinc-finger nucleases (ZFNs) provided an alternative approach that avoids the necessity of germline competent embryonic stem cells. Meanwhile, improvements have been made to the well established random mutagenesis mediated by transposons or N-ethyl-N-nitrosourea (ENU). The in-vitro rat spermatogonial stem cell (SSC) system has greatly optimized these phenotype-driven approaches for future applications. SUMMARY: The rat has long been a prime model organism in physiological, pharmacological and neurobehavioral studies. The recent advances of rat reverse genetic approaches, together with the classical ENU and transposon mutagenesis system, will contribute tremendously to the deciphering of gene functions and the creation of rat disease models.


Subject(s)
Genetic Techniques , Animals , Disease Models, Animal , Gene Targeting , Genotype , Humans , Mutation , Phenotype , Rats , Rats, Transgenic
19.
J Neural Transm (Vienna) ; 118(7): 997-1001, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21607742

ABSTRACT

Monoamine oxidase (MAO) A is the major metabolizing enzyme of serotonin (5-hydroxytryptamine, 5-HT) which regulates early brain development. In this study, wild-type (WT) and MAO A(neo) embryonic stem (ES) cell lines were established from the inner cell mass of murine blastocysts and their characteristics during ES and differentiating stages were studied. Our results show that the differentiation to neural cells in MAO A(neo) ES cells was reduced compared to WT, suggesting MAO A played a regulatory role in stem cells neural differentiation.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/enzymology , Monoamine Oxidase/genetics , Monoamine Oxidase/metabolism , Neural Stem Cells/enzymology , Neurogenesis/physiology , Neurons/enzymology , Animals , Cell Line , Embryonic Stem Cells/cytology , Female , Male , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neurons/cytology , Pregnancy
20.
J Cell Biochem ; 109(3): 606-14, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20039312

ABSTRACT

There is increasing evidence to suggest that embryonic stem cells (ESCs) are capable of differentiating into hepatocytes in vitro. In this study, we used a combination of cytokines and sodium butyrate in a novel three-step procedure to efficiently direct the differentiation of mouse ESCs into hepatocytes. Mouse ESCs were first differentiated into definitive endoderm cells by 3 days of treatment with Activin A. The definitive endoderm cells were then differentiated into hepatocytes by the addition of acidic fibroblast growth factor (aFGF) and sodium butyrate to the culture medium for 5 days. After 10 days of further in vitro maturation, the morphological and phenotypic markers of hepatocytes were characterized using immunohistochemistry, immunoblotting, and reverse transcription-polymerase chain reaction (RT-PCR). Furthermore, the cells were tested for functions associated with mature hepatocytes, including glycogen storage and indocyanine green uptake and release, and the ratio of hepatic differentiation was determined by counting the percentage of albumin-positive cells. In the presence of medium containing cytokines and sodium butyrate, numerous epithelial cells resembling hepatocytes were observed, and approximately 74% of the cells expressed the hepatic marker, albumin, after 18 days in culture. RT-PCR analysis and immunohistochemistry showed that these cells expressed adult liver cell markers, and had the abilities of glycogen storage and indocyanine green uptake and release. We have developed an efficient method for directing the differentiation of mouse ESCs into cells that exhibit the characteristics of mature hepatocytes. This technique will be useful for research into the molecular mechanisms underlying liver development, and could provide a source of hepatocytes for transplantation therapy and drug screening.


Subject(s)
Butyrates/pharmacology , Cell Differentiation , Cytokines/pharmacology , Embryonic Stem Cells/cytology , Hepatocytes/cytology , Animals , Butyrates/metabolism , Cell Lineage , Cells, Cultured , Embryonic Stem Cells/metabolism , Hepatocytes/metabolism , Mice
SELECTION OF CITATIONS
SEARCH DETAIL