Skip to main content
Springer logoLink to Springer
. 2011 May 3;8(1):1–15. doi: 10.1007/s12015-011-9261-7

LIF-Dependent Signaling: New Pieces in the Lego

Marie-Emmanuelle Mathieu 1,2, Claire Saucourt 1,2, Virginie Mournetas 1,2, Xavier Gauthereau 1,2, Nadine Thézé 1,2, Vincent Praloran 1,2, Pierre Thiébaud 1,2, Hélène Bœuf 1,2,
PMCID: PMC3285761  PMID: 21537995

Abstract

LIF, a member of the IL6 family of cytokine, displays pleiotropic effects on various cell types and organs. Its critical role in stem cell models (e.g.: murine ES, human mesenchymal cells) and its essential non redundant function during the implantation process of embryos, in eutherian mammals, put this cytokine at the core of many studies aiming to understand its mechanisms of action, which could benefit to medical applications. In addition, its conservation upon evolution raised the challenging question concerning the function of LIF in species in which there is no implantation. We present the recent knowledge about the established and potential functions of LIF in different stem cell models, (embryonic, hematopoietic, mesenchymal, muscle, neural stem cells and iPSC). We will also discuss EVO-DEVO aspects of this multifaceted cytokine.

Keywords: LIF cytokine, IL6, Pleiotropy, Stem cells, Signaling, Chromatin, miRNA, EVO-DEVO, Reprogrammation, p53

Generalities on LIF

LIF cytokine is a glycosylated protein (MW of 37–62 kDa depending on its degree of glycosylation) secreted by extraembryonic part of the embryo at the egg cylinder stage as well as by many cell types in adult organs (e.g.: endometrial cells, fibroblasts, hepatocytes, osteoblasts, monocytes, macrophages, T cells) [1, 2]. Three laboratories simultaneously discovered and cloned the LIF cytokine through its pleiotropic biological activities on i) the proliferation of adult human T cells (HILDA; [3]), ii) the maintenance of ES cells pluripotency (DIF [4]) and iii) the inhibition of leukemic cell differentiation (LIF; [5]). LIF was thus characterized as a pleiotropic cytokine with pro or anti-differentiation, pro or anti-survival effects depending upon cell maturity and cell types [6, 7].

LIF belongs to the “helical type 1” Interleukin 6 family, which includes IL11, IL27, CNTF, CT1, CLC, and OSM [812]. These cytokines interact with homo- or heteromeric receptors, all including the common gp130 subunit (IL6 signaling transducer, (IL6ST)) [13]. LIF receptor is composed of two subunits, gp130 and gp190 (LIF receptor beta). The gp130 common subunit is believed to explain the functional redundancies with several members of the IL-6 family [1]. The gp190 subunit is under ERK MAPK and axotrophin/March7 E3 ligase-dependent degradation pathways, respectively in liver cell models and in T cells [14, 15]. Glycosylated LIF can also hold mannose phosphate residues able to bind the Mannose 6 phosphate receptor to the core gp130/GP190 complex and allowing recycling of LIF ligand [16]. Recently, Sortilin, an intracellular sorting receptor, member of vacuolar protein sorting-10 (Vps10) domain-containing proteins, has been shown to facilitate the signaling of all helical type 1 cytokines which engage the gp130/LIFR beta complex [17].

Studies of LIF Knock-Out (KO) mice revealed that LIF is essential for the implantation process of blastocysts, for the maintenance of hematopoietic stem cell pools and for the not so well understood mechanisms leading to cachectic animals ([1820] and reviewed in [7]). In addition, in LIF rescued KO mice model, it was shown that LIF is essential for mammary gland involution after lactation [21]. More recent studies performed with LIF KO mice challenged for injury responses, demonstrated the importance of LIF at various stages of neurogenesis and for tissue regeneration after brain or spinal cord injury [2224]. Also, LIF is potentially involved in particular contexts of muscle stimulation and regeneration [2527] and analysis of LIF KO newborn mice revealed a 40% decrease in bone volume [28]. Double and triple KO model mice with other members of the IL6 family, as CT1 and CNTF, revealed also the importance of LIF and CNTF for motor neuron functions [29, 30]. The therapeutic potential of LIF in neurodegenerative and autoimmune diseases and in reproduction failure treatments has recently been reviewed stressing the importance to dissect LIF outcomes in the different cell contexts [31, 32]. Exemples of LIF functions in vitro and in muso are presented in Tables 1 and 2.

Table 1.

Pleiotropic effects of LIF: in vitro models

Model Effects of LIF Ref
Reproduction/fertility Healthy women volunteers and 8-week-old female mice. LIF induces oocytes and graafian follicle expansion. [176]
In vitro studies from germ and testis cell culture. LIF increases male germ cell maturation during spermatogenesis. [177]
Primary human endometrial epithelial cells and human endometrial epithelial cell line. LIF and IL11 synergize to regulate cell adhesion of endometrial epithelial cells with blastocyst during implantation process. [178]
Hormone regulation NCI-H295R adrenocortical cells LIF stimulates adrenal steroidogenesis. [179]
Hematopoiesis Bone marrow from 2 month old C3H/HeJ mice. LIF and multi-CSF synergize to stimulate murine megakaryocyte production in vitro. [180]
Human mesenchymal stem cells, Treg cells. LIF is involved in transplantation tolerance. [15, 181]
Muscle C2C12 myoblast cell line LIF induces the proliferation of myoblasts. [182]
Model of JNK1−/− mice There is a link between TNFalpha/LIF/JNK1 in myoblast proliferation/differentiation. [183]
Vessels IEM, murine endothelial cell line. LIF, in synergy with FGF, regulates signals controlling capillary outgrowth. [184]
Neurons Neural crest cultures from CBA mouse embryos at stage E9. LIF stimulates the generation of sensory neurons in culture of mouse neural crest. [185]
Human midbrain precursor cells. LIF favours neurogenic differentiation of long-term propagated human midbrain precursor cells. [186]
Eyes Human retinal cells. LIF allows CD133+ adult retinal cells to remain undifferentiated. [187]
Ears Corti1 cells. LIF is involved in auditory cell proliferation. [188]
Kidneys Culture of rat metanephric mesenchyme. LIF converts kidney mesenchyme to epithelia which then form nephrons. [189]
Liver Human hepatoma cell lines (HepG2). LIF, with oncostatin M, induces hepcidin production in hepatoma cell lines. Hepcidin is considered to be the main factor responsible for the development of anemia in inflammatory conditions. [190]
LIF in cancer Cell lines derived from thyroid tumor specimen in human patients. [191]
Normal and tumoral mammary derived cell lines. LIF inhibits (in non-tumorigenic cells) or induces (in tumorigenic cells) cell survival. [192]
ARMS and ERMS rhabdomyosarcoma cell lines. LIF is involved in rhabdomyosarcoma-derived metastasis. [193]
In vivo and in vitro studies. LIF is involved in melanoma tumor growth. [194]
In vivo and in vitro studies: BalbC/mice; SEKI cells; 8 human melanoma-derived cell lines. LIF has a potential role in melanoma-induced bone metastasis. [195]
In vivo and in vitro studies: human glioma cell lines: U373MG and A172 and human primary cell cultures. LIF and TGFbeta induce self renewal of glioma-initiating cells, promoting oncogenesis in vivo. [196]

Table 2.

Pleiotropic effects of LIF revealed by LIF−/− KO mice model analysis

Effects of LIF Ref
Implantation LIF is required for implantation of blastocysts. [19, 20]
Hematopoiesis LIF is involved in the maintenance of HSC pools. [20]
LIF is secreted by mesenchymal stromal cells to stimulate survival and proliferation of haematopoietic stem cells. [135]
LIF regulates the differentiation potential of MSC. [59]
Muscles LIF contributes to regenerate muscle. [197]
LIF is critical for the development of skeletal muscle hypertrophy in the functional overload model. [143]
Vessels LIF modulates oxygen-dependant VEGF expression and is essential for ensuring proper capillary density. [198]
Bones LIF regulates osteoclast size. [199]
Neurons LIF is a key regulator of neural injury. [200]
LIF is involved in glia phenotypes. [201]
LIF prevents oligodendrocytes destruction and improve remyelination of neurons in mice suffering from multiple sclerosis. [202]
LIF is required for normal development of hippocampal astrocytes, a process regulated by spontaneous neural impulse activity through the release of ATP. [203]
LIF is required for correct myelination for a short time window, during postnatal mouse optic nerve development. [152]
LIF may activate an endogenous rescue pathway that protects viable photoreceptor cells, leading to an increased photoreceptor survival in stressed retina. [204]
LIF signalling pathway is required for the initiation of the astrogliosis-like reaction of retinal Müller cells after optic nerve injury. [205]
LIF is necessary for injury-induced neurogenesis. [206]
Inflammation LIF is a major anti-inflammatory molecule produced in the CFA model (injection of complete Freund’s adjuvant which induces cutaneous inflammation). It is a key regulator of the cytokine cascade. [207]
Hormonal functions LIF regulates the production of pituitary ACTH and inhibits the production of prolactin and growth hormone. [208]

Recent studies have also demonstrated that LIF expression is under the control of the p53 pathway for the implantation process [3335]. This recent finding of a crosstalk between p53 (the so called “guardian of the genome” also recently involved in somatic cell reprogrammation) and LIF opens new perspectives for LIF studies in relation with the resetting of the pluripotent program, from committed or mature differentiated cells [3641].

LIF Signaling and Pleiotropy: As a Lego

We propose to view LIF signaling as a Lego built with different combinations of similar pieces, leading to various outcomes, which range from cell proliferation and survival to differentiation and apoptosis, depending on maturity and cell types [1, 2, 7, 42]. This Lego includes the “ptyr signaling toolkit” described recently by Lim and Pawson [43]. Indeed, major pieces, always present in the core of the Lego, are kinases (as JAKs, SRC members, ZAP70 cytosolic tyrosine kinases and MAPK family members), activated transcription factors (STAT, AP1 (e.g.:JunB/cfos), NFKb, MYC family members) and feedback loop components like SHP1 and SHP2 phosphatases, PIAS (Protein Inhibitor of the Activated STAT) family of proteins and SOCS (Suppressor of cytoking Signaling). In both mouse and human genomes, there are four JAKs (JAK1, 2, 3 TYK2), nine SRC [44], two ERK/MAPK (ERK1 and ERK2), eight STATs, two SHPs (i.e., SHP-1 and SHP-2), eight SOCS (SOCS1 to SOCS7 and CIS) and four PIAS (PIAS1, -3, -x, and -y) proteins [45]. Many combinations of these proteins will result in pleiotropic effects of this cytokine. For example, the LIF/JAK1/ERK1/2/PI3K/STAT3/JunB-cfos/KLF4/5/SOCS3 combination leads to maintenance of the pluripotency in the murine ES cell model, with ERK signaling pushing towards differentiation while STAT3 and its targets allow cells to remain undifferentiated by repressing endoderm and mesoderm differentiation programs [4648]. Epigenetic marks, not yet studied in details, represent an additional level of sophistication required to understand the mechanisms of LIF pleiotropy, in cell models in which LIF mediates opposite effects. Eed protein, a LIF-dependent STAT3 target of the repressive Polycomb complex, has been shown to silence differentiation-associated genes in self-renewing mES cells [49]. We have proposed that the level of expression and/or of activation of chromatin regulators could end up to opened or closed chromatin configuration, leading to accessibility (or not) of STAT3-dependent promoters, helping to explain opposite outcomes of LIF/STAT3 pathway [7]. It might be informative to perform parallel Chromatin Immunoprecipitation (ChIP)-seq analyses with anti-phosphoSTAT3 antibody in mES and in the M1 cell line (a leukemic myeloid cell line in which LIF triggers differentiation) to characterize LIF/STAT3—dependent promoters in two cell models in which LIF drives opposite effects [5054]. The importance of feedback loop control of LIF signaling, almost always including SOCS3, has recently been illustrated in mice engineered to express mutated forms of gp130 lacking the SOCS3-binding site. In those mice, which develop a variety of hematopoietic and immunological defects, STAT signaling is sustained highlighting the critical role of SOCS3 in limiting gp130 signaling [55]. In addition, differential kinetic of inactivation and desensitization of LIF-dependent pathways, which could be mediated by different inhibitory signaling components, as shown for the IL6 cytokine [56], could account for its various cell-dependent effects. The characterization of combinatorial LIF-dependent activated/repressed components (including proteins and also miRNAs, as demonstrated in mES and human mesenchymal cells [5759]) and the set up of tools allowing to understand the mechanisms of action of these proteins and/or miR complexes on cell physiology, is a future challenge in cytokine and stem cell fields.

LIF in Stem Cells

LIF in mES Cells: Highlights on New LIF Targets and of Connections with the Trio OCT4/NANOG/SOX2

Murine ES cell model, strictly depending on LIF for self renewal and maintenance of pluripotency, is a powerful model to study its effects on cells grown at different stages of maturity (from pluripotent to early differentiated cells) and to unravel the mechanisms of pluripotency and pleiotropy (Fig. 1). LIF, in synergy with BMP4 or Wnts protein members (Wnt3a and Wnt5, [60, 61]) induces PI3K and ERK signals which are contradictory signals leading respectively to maintenance of pluripotency (through NANOG) or to differentiation [46, 47, 6266]. ES cells are poised, able to respond shortly and efficiently to differentiation signals. However, a complex pluripotent program locks cells in self renewal and undifferentiated state, at least in cell culture. Years of studies of the mES cell model led to the characterization of master pieces of pluripotent program. This includes. along with the LIF signaling pathway, which represses expression of endoderm and mesoderm markers, the OCT4/SOX2 targets genes, which block differentiation towards trophectoderm lineage. Knowledge of these complementary and intricate pathways led also to the set up of defined medium complemented with specific chemicals which mimic the effects of essential components of stemness, as the 3i medium (a basic serum free medium supplemented with three chemical inhibitors, repressing MEKs, GSK3b and FGFR pathways) or the “pluripotin/LIF” medium [64, 6769] . Recently retinol, the alcohol form of Vitamin A, which is not metabolized in retinoic acid (RA) in ES cells, because of the absence of RALDH and ALDH enzymes, has been shown as a new powerful inducer of NANOG allowing maintenance of ES cell pluripotency in the absence of LIF [70, 71]. In addition, it has been demonstrated that mES cells express some markers in a salt/pepper way, as shown in Inner Cell Mass (ICM) of early blastocysts [72]. Indeed, genes as Rex1, Nanog, Zscan4 and many others have heterogeneous expression in morphologically homogeneous colonies of mES cells, probably allowing cells to respond quickly to differentiation signals [73, 74]. The importance of LIF-dependent components involved in the heterogeneity of mES cells is presently unknown.

Fig. 1.

Fig. 1

The mES system: the «three in one» cell model to study LIF mechanisms in pluripotent, committed and differentiated cells. Part of this figure has been taken from the Web site: http://en.wikipedia.org/wiki/Stem_cell, available under the «Creative Commons Attribution-ShareAlike 2.5 Generic License ». This model is based on references: 105, 112, 113 and 114

Genes Involved in Cell Pluripotency: Functional Involvement, One by One

There are at least a hundred of genes individually involved in the maintenance of mES cell pluripotency. Indeed, Knock Down (KD) or Knock Out (KO) of candidate genes, proves to be a powerful way to demonstrate their role in ES cell pluripotency. Proteins in each cell compartment, from the membrane to the nucleus have been shown to be critical for maintenance of pluripotency as shown for Gap junction proteins [7578], CrxOS [79], Yap [80], Pem [81], Zic3 [82, 83], Zfx [84], Pdcd2 [85], Cobra1 [86] and many other genes revealed by high throughput RNAi strategies [8789]. Also chromatin regulators (Jumonji members, [90] and Chd1, [91], which modulate respectively the level of histone methylation and the degree of chromatin compaction, and Ronin [92, 93], all able to regulate many genes together) and miRNAs (controlling cell cycle regulators via c-MYC [57, 94] or Master gene expression [95]) are also key players involved in the maintenance of mES cell pluripotency. The importance of miRNAs was established in previous studies showing that embryos or ES cells lacking proper miRNA synthesis (Dicer and Dgcr8 KO models) are not anymore pluripotent, stressing the importance of miRNA in ES cell plasticity [96101].

It is worth stressing that some of these genes behave as “rheostat” with various level of their expression leading to different (sometimes opposite) cell phenotypes, as first shown for Oct4 and then for Nanog and Sox2 [102104], illustrating the poised state of mES cells.

However, in seeking for the “Holy Grail” of the pluripotency markers, we might attempt to dissociate genes whose repression will lead to slow destruction of cells (which could go through a pseudo-differentiation stage before death, when touching Gap junctions, adhesion or cytoskeleton cell components, for example), to genes having specific effects on the pluripotent machinery, a not easy task.

Novel LIF Signatures

By extensive microarray studies performed in mES cells grown with or without LIF for 24 or 48 h and reinduced with LIF for 30 min, we have defined three types of LIF signatures: Pluri, Spe-Lifind and Pleio-Lifind [105]. Genes from both Pluri and Lifind clusters are essential for the proper self renewal and maintenance of pluripotency in mES cells.

Pluri genes, whose expression is restricted to undifferentiated mES cells, at least up to 10 days of differentiation triggered by LIF withdrawal, includes Esrrb, Gjb3, Krt42, Ak7, Ly6, Susd2, Irak3, Tcfcp2l1, Pim3, Ceacam1 and Mras. This extends the list of known stemness genes [89, 106, 107]. By analyzing the expression profiles of the Pluri genes in Microarray data obtained with the KD of Oct4 or Nanog [108] we concluded that while the expression of Esrrb and Ly6 is repressed in OCT4 or NANOG KD cell lines, expression of Susd2, Ak7, Krt42 and Irak3 is only under OCT4 control. In addition, expression of gap junction protein encoding genes is induced in OCT4 (Gjb3/Connexin 31 and Gjb5/connexin 31.1) or NANOG (Gja1/connexin43) Knock Down cell lines. Individual KD of Irak3, Susd2 and Ly6 leads to weak increase of differentiation markers suggesting their involvement in mES cell pluripotency. Efficient approaches allowing to disrupt the expression of many genes at once should be necessary for further characterization of gene clusters in mES cells [105].

In cells grown without LIF for 24 or 48 h and reinduced with LIF for 30 min, we have identified the Lifind (Lif induced) genes. Some of them are direct targets of STAT3 and a subset of these genes is also regulated by the PI3K/NANOG pathway [48, 109, 110]. Only part of the genes induced after 24 h of LIF withdrawal is also induced after a longer period of starvation. We have made a distinction between Spe-Lifind genes (only induced in restricted time windows of LIF withdrawal, 24 h) and Pleio-Lifind genes (induced by LIF after different periods of LIF starvation, 24 h, 48 h or 10 days after LIF withdrawal, this latter condition corresponding to differentiated cells which re-express LIF and its receptors [4, 7, 105, 111]). Since withdrawal of LIF for 24 h leads to reversible commitment [112114], we postulate that some of the Spe-Lifind genes could be involved in the reversible process.

Klf4 (a well known member of the “Yamanaka” cocktail, involved in reprogrammation of somatic cells to iPSC) and Klf5 are both Spe-Lifind genes shown to be critical actors of maintenance of pluripotency in mES [115120]. Both genes are also direct STAT3 targets and block endoderm (Klf4) or mesoderm (Klf5) differentiation [48]. Klf4 and 5 are also under the control of NANOG expression and a regulatory loop between KLF4 and NANOG has been suggested since KLF4 regulates the expression of NANOG by direct binding to its promoter [120]. Klf4 synergizes with OCT4, definitively connecting master genes (OCT4/SOX2/NANOG) with LIF/STAT3 pathway [48, 117, 120, 121]. Other Spe-Lifind genes, could be essential “ressetors” of pluripotent program during the reversible phase of commitment [7, 114].

LIF in Adult Stem Cells

Hematopoietic Stem Cells (HSC)

Several groups have described similar, different or even contradictory results about the in vitro effects of LIF on cultured normal adult hematopoietic stem cells and progenitors. In articles devoted to its functional effects on normal hematopoietic progenitors it was shown that LIF has a growth stimulating activity on human CFU-Eo and BFU-E [122] while it is ineffective on mouse CFU-GM [123]. LIF also induces the proliferation of very primitive multipotential progenitors (so called CFU-Blast) suggesting that this cytokine could stimulate the cell cycle entry of these mostly quiescent primitive progenitors [124]. Interestingly, LIF is active on CFU-Mix only in serum containing medium and its effect is mostly synergistic or additive to other multi-lineage cytokines (IL-3, IL-6 and GM-CSF) [125]. These data suggest that LIF mostly stimulates the in vitro proliferation of primitive hematopoietic progenitors. In vivo, LIF is active on hematopoietic progenitors and play a role in the regulation of the stem cell pool [20]. Permanently elevated levels of LIF in adult mice induced a polyvisceral and rapidly lethal pathology [126]. The most important hematological modifications are a reduction of bone marrow hematopoiesis related to myelosclerosis that contrasts with a splenomegaly due to an abnormal spleen hematopoiesis, increased levels of progenitors in the spleen and thrombocytosis [126, 127]. However the most informative results concerning the physiological role of LIF in the regulation of hematopoiesis were obtained from KO mouse models. LIF deficient mice are viable. However, when compared to wild type animals the pool of BFU-E and CFU-GM progenitors were reduced in bone marrow and even more in the spleen of LIF−/− mice [20]. More surprising is the fact that the null LIFR beta (gp190) spontaneous mutations in humans and induced by gene targeting in KO mice had no hematological abnormality despite the fact that other LIF target tissues were disturbed [128, 129]. Taken altogether, these in vitro and in vivo results suggest that LIF is not a mandatory cytokine for the physiological regulation of adult hematopoiesis. This is in agreement with its potential role on some basic functions such as effect on chromatin status, for example, for which a degree of redundancy is expected.

Mesenchymal Stem Cells (MSC)

Among their numerous biological functions, MSC display immune-modulatory properties. They suppress T cell proliferation induced by various stimuli in vitro and they show similar effect in vivo. Indeed several clinical trials evidenced that injection of MSC to Bone Marrow (BM) allografted patients reduced their graft versus host disease by improving the donor T cells immune tolerance [130]. Interestingly, LIF is produced by human BMMSC [131] and its overexpression and depletion in mouse models led to immune disturbances [132]. Recently, it was suggested that the MSC-induced immune tolerance is due to their production of LIF [133]. However, the cellular and molecular mechanisms (that also involve other identified MSC molecules) leading to this transplantation tolerance remain functionally ignored. Another established ex vivo effect of MSC is to improve the maintenance of HSC in liquid cocultures. Indeed, a positive effect of LIF on the in vitro maintenance of murine HSC was mediated by BM stromal cells in which it upregulates the expression of various hematopoietic cytokines [134]. By using an elegant model of young and old, wild type and LIF−/− mice for cross cocultures of MSC and Lin- Sca1+ hematopoietic progenitors and stem cells, it has been shown that LIF−/− mice had only slight disturbances of hematopoiesis when explored in culture [135] since LIF−/− and WT MSC were not significantly different in their capacity to maintain Lin− Sca1+ hematopoietic progenitors of WT and LIF−/− mice in liquid cocultures. Complementary experiments (including transplantation) should be performed if one wants to conclude about the real direct/indirect effects of LIF on HSC in vivo. However, a novel direct autocrine effect of LIF to maintain the multipotent program of MSC has recently been described. Indeed, down regulation of MSC LIF expression by specific miRNA favors their differentiation towards the osteoblast and adipogenic lineages [59]. In this context, it is worth stressing that in vivo silencing of the gp190 subunit of the LIF receptor leads to major disturbances of the bone and other mesenchymal tissues suggesting a major non redundant role of LIF in the maintenance and commitment of MSC [128, 129].

Muscle Satellite Cells

Muscle satellite cells are the major cell type responsible for post-natal skeletal muscle growth and regeneration. These quiescent cells are located under the basal lamina of muscle fibers, and become activated upon injury. They proliferate and differentiate into new muscle fibers and during regeneration, the satellite cell pool is also reconstituted [136138]. Since the first articles showing a role of LIF in stimulation of myoblast proliferation in culture [139, 140], it has been shown that LIF engages JAK1, STAT1, and STAT3 to promote cell proliferation and to repress myogenic differentiation, in primary myoblasts and in the C2C12 cell models [26, 141]. However, an opposite role for STAT3 in myoblast differentiation was shown by knockdown of endogenous STAT3 which dramatically blocked myogenic differentiation. It was then suggested that, as in the murine ES cell model, LIF/STAT3 might play distinct roles at different stages of muscle differentiation [142]. Further studies are needed to understand how STAT3 coordinates with myoD to control myogenic differentiation and how they cross-talk with other signaling pathways. It has also been demonstrated that LIF expression in skeletal muscle is critical for the development of skeletal muscle hypertrophy in the functional overload model [143]. LIF has also been suggested as being a novel myokine, secreted by muscle cells during exercise in link with activation of calcium-dependent pathway and changes in energy status [27]. However, in this study, the LIF secretion is around 3 pg/ml, which could be considered to be close to background level, based on known physical constant describing cytokine/receptor interactions [144].

Neural Stem Cells

Neural stem cells (NSCs) in the adult brain continuously provide new neurons to the hippocampal dentate gyrus (DG) and the olfactory bulb (OB). The progression from neural precursor cells (NPCs) to mature neurons is tightly controlled by coordination of cell-intrinsic programs and external signals within the neurogenic niche. Understanding signaling in adult neurogenesis is a key challenge to understand the physiological roles of neurogenesis, but also to provide knowledge required to use NSCs as potential therapy for treatment of brain diseases [145147].

Cytokines that signal through the LIFRbeta/gp130 receptor complex, including LIF and CNTF have been suggested to promote the self-renewal of embryonic and adult mouse or rat NPCs [22, 148]. However, with CNTF or LIF KO models, it was shown that while CNTF-induced STAT3 signaling is essential for the formation and/or maintenance of the neurogenic subgranular zone in the adult dentate gyrus, LIF was not required [149]. In addition, in a model of human neural stem cells, which can be expanded under EGF and FGF2, no effect of LIF was observed [150]. In contrast, in a rat model, LIF, and not CNTF, was crucial for the expansion of NPCs in the Sub Ventricular Zone (SVZ) after perinatal brain injury [151]. LIF was also shown to control neural differentiation and maintenance of stem cell-derived murine spiral ganglion neuron precursors, a finding which could be relevant in cell replacement studies with animal models featuring spiral ganglion neuron degeneration [23]. Another study shows that during normal development of mouse optic nerve, there is a defined developmental time window when LIF is required for correct myelination [152].

Understanding of LIF signaling in adult stem cells will still depend on detailed analysis performed in LIF KO model mice, challenged with particular stimuli aiming at increasing or decreasing the natural pool of stem cells in their relevant niche.

LIF in Reprogrammation: Antagonism Between LIF and p53 Pathways. What for?

Somatic cell reprogrammation which leads to the derivation of iPSC is mediated by specific set of genes (Oct4, Sox2, cMyc and Klf4 or Oct4, Sox2, Lin28 and Nanog), including LIF targets [119, 153, 154]. L-Myc has been shown recently to replace c-MYC in the reprogrammation cocktail with the potential advantage to reduce the tumorigenicity of the derived iPSC [155]. In addition, growing cells at low O2 concentration (3%–5% O2), [156], and blocking the p53 pathway [36, 37, 40, 41, 157], are also critical parameters which increase the efficiency of somatic cell reprogrammation.

The expression of the LIF cytokine is under the direct control of p53 (at least in mouse endometrium cells [35]), and of N-MYC (in the neuroblastoma cell model, [158]). Indeed, LIF promoter contains specific binding sites for these regulatory transcription factors. Whether N-MYC and p53 act together or not have not been established so far. LIF-induced genes and repression of p53 pathway seems to be paradoxical effectors for efficient cell reprogramming, unless both signals are not required simultaneously, a hypothesis which will deserve future attention.

LIF in EVO-DEVO

A large amount of data has been accumulated on the pleiotropic functions of LIF in mammals, but so far we have little insight on what could be its functions in non mammalian vertebrate with respect to early development and differentiation [159]. A limited number of signaling pathways is operating during metazoan development and most, if not all of them are conserved over a large evolutionary scale [160, 161]. Among them is the JAK/STAT pathway which conveys LIF signals. LIF belongs to class I-helical cytokines family which is considered to constitute a monophyletic group having evolved from a single ancestral gene through successive duplication events mainly in the vertebrate lineage [162]. Indeed, the core signaling elements of the JAK/STAT pathway has been found in invertebrate such as Drosophila with a unique cytokine-like peptide ligand capable of activating the pathway [163]. The knowledge of the conservation of IL6 family members, as LIF, between divergent vertebrate species and the functions of LIF during development can be very informative for understanding its pleiotropic functions. LIF ortholog has been identified through cDNA cloning and in silico analysis in several non mammalian vertebrates. Chicken cDNA LIF has been cloned and shown to be able to maintain blastodermal cells into an undifferentiated state [164]. cDNA encoding LIF ortholog has also been cloned in teleost species as zebrafish, carp and goldfish [165167]. The finding of LIF-like cytokine in fishes indicates that the cytokine already existed before the fish-tetrapod divergence that occurred aproximatively 450 million years ago. LIF ortholog is also present in amphibian and we have cloned, through in silico analysis, a Xenopus LIF cDNA (our unpublished data). Although the amino acid conservation of LIF sequences between mammalian and non mammalian vertebrate species is rather low (20%–40%), those sequences share a conserved tridimensional fold. LIF receptor (LIFR) has been identified in chick and zebrafish and functional analysis performed in both species. In chicken, LIFR is involved in the control of vasoactive intestinal peptide expression in sympathetic neurons [168]. Functional experiments using morpholino based mediated knock down in zebrafish have shown that, unlike LIF, whose knock down has no obvious effects on development, LIFR knock down impairs proper neural development [169]. Although there is no functional data about LIF and LIFR functions in Xenopus development yet, it has been found that STAT3 activation, through gp130 signaling, ventralizes embryo independently of BMP4 [170]. Whether this effect can be related to LIF signaling through its receptor awaits experiments. Since LIF is essential to murine ES cells pluripotency, it is striking to note that STAT3 is able to maintain Xenopus pluripotent neural crest cells in an undifferentiated state downstream of FGF signals [171].

According to the current data and the evolutionary conservation of the components of the LIF signaling pathway, we speculate that LIF can generate distinct responses at different times during development/differentiation in different species. Most of the genes, that we have defined as LIF signatures in the mES model, is conserved in non mammalian vertebrate genomes and their study in the amphibian Xenopus model should allow for a rapid and efficient screening of the functions of the genes that make up the LIF signatures. This should also allow a better understanding of the functions of the LIF pathway during development.

Conclusions

The challenge, when studying LIF, is to catch the proper time window of its effects. Indeed, while LIF is essential for maintenance of pluripotency of murine ES cells (mES), it is not required for human ES cells (hES). It has been elegantly demonstrated that this was due to the fact that murine and human ES cells are respectively derived from early and late epiblast, stressing the differential effects of LIF that might depend on the embryonic stage. In addition, LIF/STAT3 signaling reverts murine EpiSC cells (Epiblast-derived cells, which are similar to human ES cells) to mES cells probably by LIF-dependent expression of KLF4, as suggested by recent reports [172175].

Functions of LIF studied in KO model mice, along with functional tests performed at different time windows in the mES cells model, with new LIF targets, should increase our knowledge on this still fascinating cytokine in the near future.

Acknowledgements

This work was supported by CNRS, University Bordeaux Segalen and fundings under the sixth Research framework program of the European Union (FungenES, LSHG-CT-2003- 503494), the Association de la Recherche contre le Cancer, the Ligue Nationale contre le cancer (comité Charentes Aquitaine), the Association Française contre les Myopathies, IFR66/SFR Trans Biomed Bordeaux and the Etablissement Français du Sang, Aquitaine.

We thank JF Moreau and P Veschambre for critical reading of the manuscript.

We apologise for LIF-dedicated articles not cited in this review.

Conflicts of interest

The authors declare no potential conflict of interest.

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Abbreviations

AK7

Adenylate Kinase 7

ALDH

AcetaLdehyde DeHydrogenase

AP1

Activator Protein 1

BFU-E

Burst Forming Unit Erythroid

BMP

Bone Morphogenetic Protein

cDNA

complementary DeoxyriboNucleic Acid

Ceacam1

Carcinoembryonic antigen-related cell adhesion molecule 1

CFU-Blast

Colony-Forming Units Blast

CFU-E

Colony-Forming Units Erythroid

CFU-Eo

Colony-Forming Units Eosinophil

CFU-GM

Colony-Forming Units Granulocyte Macrophage

CFU-Mix

Colony-Forming Units Mix

Chd1

Chromodomain-helicase-DNA-binding protein 1

CHIP-seq

Chromatin ImmunoPrecipitation followed by sequencing

CIS

Cytokine-Inductible SH2

CNTF

Ciliary NeuroTrophic Factor

CLC

Cardiotrophin-Like Cytokine

CT1

CardioTrophin-1

Cobra1

cofactor of BRCA1 (breast cancer 1)

CrxOs

Crx Opposite strand

Dapp1

Dual adaptor for PY and PI3K

DG

Dentate Gyrus

EED

Polycomb protein Embryonic Ectoderm Development

EGF

Epidermal Growth Factor

EpiSC

epiblast stemcells

ERK

Extracellular signal-Regulated Kinases

ES

Embryonic Stem

ESRRB

EStrogen-Related Receptor beta

FGF

Fibroblast Growth Factor

FGFR

Fibroblast Growth Factor Receptor

Gja1

Gap junction membrane channel protein alpha 1

Gjb3

Gap junction membrane channel protein beta 3

GM-CSF

Granulocyte Macrophage Colony Stimulating Factor

GP

GlycoProtein

GSK3b

Glycogen Synthase Kinase 3 beta

hES

human Embryonic Stem

HSC

Hematopoietic Stem Cell

ICM

Inner Cell Mass

IL6/11/27

InterLeukin 6, 11, 27

IL6ST

InterLeukin 6 Signal Transducer

iPSC

induced Pluripotent Stem Cells

IRAK 3

Interleukin 1 Receptor-Associated Kinase 3

JAK

Janus Kinase

JNK

Stress-Jun-activated protein Kinase

JunB

Oncogene JUN-B

KD

Knock Down

kDa

kilo Dalton

KLF4/5

Kruppel-Like Factor 4/5

KO

Knock-Out

KRT42

Keratin 42

LIF

Leukemia Inhibitory Factor

Lifind

LIF-induced

LIFR

LIF Receptor

Lin

Lineage

L-Myc

Avian myelocytomatosis viral oncogene homolog 1, lung carcinoma-derived

Ly6

Ly6g6e lymphocyte antigen6 complex

MAPK

Mitogen Activated Protein Kinase

MARCH7

Membrane Associated Ring finger (C3HC4) 7

MEK

MAP Kinase Kinase

mES cells

mouse Embryonic Stem cells

miRNA

micro RNA

MRas

Muscle and Microspikes ras

MSC

Mesenchymal Stem Cells

MW

Molecular Weight

MYO D

MYOgenic Differentiation Antigen

NANOG

Homeobox transcription factor Nanog

NFKb

Nuclear Factor Kappa-B

N-Myc

v-myc avian myelocytomatosis viral-related oncogene, neuroblastoma-derived

NPC

Neural Precursor Cells

NSC

Neural Stem Cells

OCT4

OCTamer 3/4

OSM

Oncostatin M

Pem

Placenta and embryonic expression protein

PI3K

PhosphatidylInositol 3-Kinase

PIAS

Protein Inhibitor of the Activated STAT

Pim3

Proviral integration site 3

Pleio-Lifind

Pleiotropic LIF-Induced

TGFbeta

Transforming Growth Factor beta

TNFalpha

Tumor Necrosis Factor alpha

RALDH

RetinALdehyde Deshydrogenase

Rex1

Reduced expression protein 1 (or Zfp-42, Zinc finger protein 42)

Sca1

Stem Cell antigen 1

SH

Src Homology

SHP

SH2-containing Phosphatase

Spe-Lifind

Specific LIF-Induced

SOCS

Suppressor Of Cytoking Signaling

SOX2

Sex-determining region Y-box2

STAT

Signal Transducer and Activator of Transcription

Susd2

Sushi domain containing 2

Tcfcp2l1

Transcription factor CP2-like 1

TYK2

Tyrosine Kinase 2

WT

Wild Type

Yap

Yes associated protein

ZAP70

Zeta-chain-Associated Protein kinase 70

Zic3

Zinc finger protein of the cerebellum 3

Zfx

Zinc finger protein, X-linked

Zscan4

Zinc finger and SCAN domain containing 4

References

  • 1.Taupin JL, Pitard V, Dechanet J, Miossec V, Gualde N, Moreau JF. Leukemia inhibitory factor: part of a large ingathering family. International Reviews of Immunology. 1998;16(3–4):397–426. doi: 10.3109/08830189809043003. [DOI] [PubMed] [Google Scholar]
  • 2.Metcalf D. The unsolved enigmas of leukemia inhibitory factor. Stem Cells. 2003;21(1):5–14. doi: 10.1634/stemcells.21-1-5. [DOI] [PubMed] [Google Scholar]
  • 3.Moreau JF, Donaldson DD, Bennett F, Witek-Giannotti J, Clark SC, Wong GG. Leukaemia inhibitory factor is identical to the myeloid growth factor human interleukin for DA cells. Nature. 1988;336(6200):690–692. doi: 10.1038/336690a0. [DOI] [PubMed] [Google Scholar]
  • 4.Smith AG, Nichols J, Robertson M, Rathjen PD. Differentiation inhibiting activity (DIA/LIF) and mouse development. Developmental Biology. 1992;151(2):339–351. doi: 10.1016/0012-1606(92)90174-F. [DOI] [PubMed] [Google Scholar]
  • 5.Gough NM, Gearing DP, King JA, Willson TA, Hilton DJ, Nicola NA, Metcalf D. Molecular cloning and expression of the human homologue of the murine gene encoding myeloid leukemia-inhibitory factor. Proceedings of the National Academy of Sciences of the United States of America. 1988;85(8):2623–2627. doi: 10.1073/pnas.85.8.2623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Shellard J, Perreau J, Brûlet P. Role of leukemia inhibitory factor during mammalian development. European Cytokine Network. 1996;7(4):699–712. [PubMed] [Google Scholar]
  • 7.Trouillas M, Saucourt C, Guillotin B, Gauthereau X, Taupin J, Moreau J, Bœuf H. The LIF cytokine: towards adulthood. European Cytokine Network. 2009;20(2):51–62. doi: 10.1684/ecn.2009.0148. [DOI] [PubMed] [Google Scholar]
  • 8.Heinrich PC, Behrmann I, Haan S, Hermanns HM, Müller-Newen G, Schaper F. Principles of interleukin (IL)-6-type cytokine signalling and its regulation. The Biochemical Journal. 2003;374(Pt 1):1–20. doi: 10.1042/BJ20030407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Murakami M, Kamimura D, Hirano T. New IL-6 (gp130) family cytokine members, CLC/NNT1/BSF3 and IL-27. Growth Factors. 2004;22(2):75–77. doi: 10.1080/08977190410001715181. [DOI] [PubMed] [Google Scholar]
  • 10.Hermanns HM, Müller-Newen G, Heinrich PC, Haan S. Bow to your partner for signaling. Nature Structural & Molecular Biology. 2005;12(6):476–478. doi: 10.1038/nsmb0605-476. [DOI] [PubMed] [Google Scholar]
  • 11.Rousseau F, Chevalier S, Guillet C, Ravon E, Diveu C, Froger J, Barbier F, Grimaud L, Gascan H. Ciliary neurotrophic factor, cardiotrophin-like cytokine, and neuropoietin share a conserved binding site on the ciliary neurotrophic factor receptor alpha chain. The Journal of Biological Chemistry. 2008;283(44):30341–30350. doi: 10.1074/jbc.M803239200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Rousseau, F., Basset, L., Froger, J., Dinguirard, N., Chevalier, S., Gascan, H. (2010). IL-27 structural analysis demonstrates similarities with ciliary neurotrophic factor (CNTF) and leads to the identification of antagonistic variants. Proceedings of the National Academy of Sciences of the United States of America. [DOI] [PMC free article] [PubMed]
  • 13.Gearing DP. The leukemia inhibitory factor and its receptor. Advances in Immunology. 1993;53:31–58. doi: 10.1016/S0065-2776(08)60497-6. [DOI] [PubMed] [Google Scholar]
  • 14.Blanchard F, Duplomb L, Wang Y, Robledo O, Kinzie E, Pitard V, Godard A, Jacques Y, Baumann H. Stimulation of leukemia inhibitory factor receptor degradation by extracellular signal-regulated kinase. The Journal of Biological Chemistry. 2000;275(37):28793–28801. doi: 10.1074/jbc.M003986200. [DOI] [PubMed] [Google Scholar]
  • 15.Gao W, Thompson L, Zhou Q, Putheti P, Fahmy TM, Strom TB, Metcalfe SM. Treg versus Th17 lymphocyte lineages are cross-regulated by LIF versus IL-6. Cell Cycle. 2009;8(9):1444–1450. doi: 10.4161/cc.8.9.8348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Blanchard F, Duplomb L, Raher S, Vusio P, Hoflack B, Jacques Y, Godard A. Mannose 6-Phosphate/Insulin-like growth factor II receptor mediates internalization and degradation of leukemia inhibitory factor but not signal transduction. The Journal of Biological Chemistry. 1999;274(35):24685–24693. doi: 10.1074/jbc.274.35.24685. [DOI] [PubMed] [Google Scholar]
  • 17.Larsen JV, Hansen M, Møller B, Madsen P, Scheller J, Nielsen M, Petersen CM. Sortilin facilitates signaling of ciliary neurotrophic factor and related helical type 1 cytokines targeting the gp130/leukemia inhibitory factor receptor beta heterodimer. Molecular and Cellular Biology. 2010;30(17):4175–4187. doi: 10.1128/MCB.00274-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bhatt H, Brunet LJ, Stewart CL. Uterine expression of leukemia inhibitory factor coincides with the onset of blastocyst implantation. Proceedings of the National Academy of Sciences of the United States of America. 1991;88(24):11408–11412. doi: 10.1073/pnas.88.24.11408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Stewart CL, Kaspar P, Brunet LJ, Bhatt H, Gadi I, Köntgen F, Abbondanzo SJ. Blastocyst implantation depends on maternal expression of leukaemia inhibitory factor. Nature. 1992;359(6390):76–79. doi: 10.1038/359076a0. [DOI] [PubMed] [Google Scholar]
  • 20.Escary JL, Perreau J, Duménil D, Ezine S, Brûlet P. Leukaemia inhibitory factor is necessary for maintenance of haematopoietic stem cells and thymocyte stimulation. Nature. 1993;363(6427):361–364. doi: 10.1038/363361a0. [DOI] [PubMed] [Google Scholar]
  • 21.Kritikou EA, Sharkey A, Abell K, Came PJ, Anderson E, Clarkson RW, Watson CJ. A dual, non-redundant, role for LIF as a regulator of development and STAT3-mediated cell death in mammary gland. Development. 2003;130:3459–3468. doi: 10.1242/dev.00578. [DOI] [PubMed] [Google Scholar]
  • 22.Bauer S, Patterson PH. Leukemia inhibitory factor promotes neural stem cell self-renewal in the adult brain. The Journal of Neuroscience. 2006;26(46):12089–12099. doi: 10.1523/JNEUROSCI.3047-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Oshima K, Teo DTW, Senn P, Starlinger V, Heller S. LIF promotes neurogenesis and maintains neural precursors in cell populations derived from spiral ganglion stem cells. BMC Developmental Biology. 2007;7:112. doi: 10.1186/1471-213X-7-112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Simamura E, Shimada H, Higashi N, Uchishiba M, Otani H, Hatta T. Maternal leukemia inhibitory factor (LIF) promotes fetal neurogenesis via a LIF-ACTH-LIF signaling relay pathway. Endocrinology. 2010;151(4):1853–1862. doi: 10.1210/en.2009-0985. [DOI] [PubMed] [Google Scholar]
  • 25.Hunt LC, Tudor EM, White JD. Leukemia inhibitory factor-dependent increase in myoblast cell number is associated with phosphotidylinositol 3-kinase-mediated inhibition of apoptosis and not mitosis. Experimental Cell Research. 2010;316(6):1002–1009. doi: 10.1016/j.yexcr.2009.11.022. [DOI] [PubMed] [Google Scholar]
  • 26.Spangenburg EE, Booth FW. Multiple signaling pathways mediate LIF-induced skeletal muscle satellite cell proliferation. American Journal of Physiology. Cell Physiology. 2002;283(1):C204–C211. doi: 10.1152/ajpcell.00574.2001. [DOI] [PubMed] [Google Scholar]
  • 27.Broholm C, Pedersen BK. Leukaemia inhibitory factor—an exercise-induced myokine. Exercise Immunology Review. 2010;16:77–85. [PubMed] [Google Scholar]
  • 28.Bozec A, Bakiri L, Hoebertz A, Eferl R, Schilling AF, Komnenovic V, Scheuch H, et al. Osteoclast size is controlled by Fra-2 through LIF/LIF-receptor signalling and hypoxia. Nature. 2008;454(7201):221–225. doi: 10.1038/nature07019. [DOI] [PubMed] [Google Scholar]
  • 29.Sendtner M, Götz R, Holtmann B, Escary JL, Masu Y, Carroll P, Wolf E, Brem G, Brület P, Thoenen H. Cryptic physiological trophic support of motoneurons by LIF revealed by double gene targeting of CNTF and LIF. Current Biology. 1996;6(6):686–694. doi: 10.1016/S0960-9822(09)00450-3. [DOI] [PubMed] [Google Scholar]
  • 30.Holtmann B, Wiese S, Samsam M, Grohmann K, Pennica D, Martini R, Sendtner M. Triple knock-out of CNTF, LIF, and CT-1 defines cooperative and distinct roles of these neurotrophic factors for motoneuron maintenance and function. The Journal of Neuroscience. 2005;25(7):1778–1787. doi: 10.1523/JNEUROSCI.4249-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Aghajanova L. Update on the role of leukemia inhibitory factor in assisted reproduction. Current Opinion in Obstetrics & Gynecology. 2010;22(3):213–219. doi: 10.1097/GCO.0b013e32833848e5. [DOI] [PubMed] [Google Scholar]
  • 32.Slaets H, Hendriks JJA, Stinissen P, Kilpatrick TJ, Hellings N. Therapeutic potential of LIF in multiple sclerosis. Trends in Molecular Medicine. 2010;16(11):493–500. doi: 10.1016/j.molmed.2010.08.007. [DOI] [PubMed] [Google Scholar]
  • 33.Hu W, Feng Z, Teresky AK, Levine AJ. p53 regulates maternal reproduction through LIF. Nature. 2007;450(7170):721–724. doi: 10.1038/nature05993. [DOI] [PubMed] [Google Scholar]
  • 34.Hu W, Feng Z, Atwal GS, Levine AJ. p53: a new player in reproduction. Cell Cycle. 2008;7(7):848–852. doi: 10.4161/cc.7.7.5658. [DOI] [PubMed] [Google Scholar]
  • 35.Stewart CL. Reproduction: the unusual suspect. Nature. 2007;450(7170):619. doi: 10.1038/450619a. [DOI] [PubMed] [Google Scholar]
  • 36.Kawamura T, Suzuki J, Wang YV, Menendez S, Morera LB, Raya A, Wahl GM, Belmonte JCI. Linking the p53 tumour suppressor pathway to somatic cell reprogramming. Nature. 2009;460(7259):1140–1144. doi: 10.1038/nature08311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hong H, Takahashi K, Ichisaka T, Aoi T, Kanagawa O, Nakagawa M, Okita K, Yamanaka S. Suppression of induced pluripotent stem cell generation by the p53-p21 pathway. Nature. 2009;460(7259):1132–1135. doi: 10.1038/nature08235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Liu Y, Hoya-Arias R, Nimer SD. The role of p53 in limiting somatic cell reprogramming. Cell Research. 2009;19(11):1227–1228. doi: 10.1038/cr.2009.121. [DOI] [PubMed] [Google Scholar]
  • 39.Marión RM, Strati K, Li H, Murga M, Blanco R, Ortega S, Fernandez-Capetillo O, Serrano M, Blasco MA. A p53-mediated DNA damage response limits reprogramming to ensure iPS cell genomic integrity. Nature. 2009;460(7259):1149–1153. doi: 10.1038/nature08287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Menendez S, Camus S, Belmonte JCI. p53: guardian of reprogramming. Cell Cycle. 2010;9(19):3887–3891. doi: 10.4161/cc.9.19.13301. [DOI] [PubMed] [Google Scholar]
  • 41.Li H, Collado M, Villasante A, Strati K, Ortega S, Cañamero M, Blasco MA, Serrano M. The Ink4/Arf locus is a barrier for iPS cell reprogramming. Nature. 2009;460(7259):1136–1139. doi: 10.1038/nature08290. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Hilton DJ. LIF: lots of interesting functions. Trends in Biochemical Sciences. 1992;17(2):72–76. doi: 10.1016/0968-0004(92)90505-4. [DOI] [PubMed] [Google Scholar]
  • 43.Lim WA, Pawson T. Phosphotyrosine signaling: evolving a new cellular communication system. Cell. 2010;142(5):661–667. doi: 10.1016/j.cell.2010.08.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ingley E. Src family kinases: regulation of their activities, levels and identification of new pathways. Biochimica et Biophysica Acta. 2008;1784(1):56–65. doi: 10.1016/j.bbapap.2007.08.012. [DOI] [PubMed] [Google Scholar]
  • 45.Yagil Z, Nechushtan H, Kay G, Yang CM, Kemeny DM, Razin E. The enigma of the role of protein inhibitor of activated STAT3 (PIAS3) in the immune response. Trends in Immunology. 2010;31(5):199–204. doi: 10.1016/j.it.2010.01.005. [DOI] [PubMed] [Google Scholar]
  • 46.Smith AG. Embryo-derived stem cells: of mice and men. Annual Review of Cell and Developmental Biology. 2001;17:435–462. doi: 10.1146/annurev.cellbio.17.1.435. [DOI] [PubMed] [Google Scholar]
  • 47.Niwa H. How is pluripotency determined and maintained? Development. 2007;134(4):635–646. doi: 10.1242/dev.02787. [DOI] [PubMed] [Google Scholar]
  • 48.Bourillot P, Aksoy I, Schreiber V, Wianny F, Schulz H, Hummel O, Hubner N, Savatier P. Novel STAT3 target genes exert distinct roles in the inhibition of mesoderm and endoderm differentiation in cooperation with Nanog. Stem Cells. 2009;27(8):1760–1771. doi: 10.1002/stem.110. [DOI] [PubMed] [Google Scholar]
  • 49.Ura H, Usuda M, Kinoshita K, Sun C, Mori K, Akagi T, Matsuda T, Koide H, Yokota T. STAT3 and Oct-3/4 control histone modification through induction of Eed in embryonic stem cells. The Journal of Biological Chemistry. 2008;283(15):9713–9723. doi: 10.1074/jbc.M707275200. [DOI] [PubMed] [Google Scholar]
  • 50.Boeuf H, Hauss C, Graeve FD, Baran N, Kedinger C. Leukemia inhibitory factor-dependent transcriptional activation in embryonic stem cells. The Journal of Cell Biology. 1997;138(6):1207–1217. doi: 10.1083/jcb.138.6.1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Matsuda T, Nakamura T, Nakao K, Arai T, Katsuki M, Heike T, Yokota T. STAT3 activation is sufficient to maintain an undifferentiated state of mouse embryonic stem cells. The EMBO Journal. 1999;18(15):4261–4269. doi: 10.1093/emboj/18.15.4261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Minami M, Inoue M, Wei S, Takeda K, Matsumoto M, Kishimoto T, Akira S. STAT3 activation is a critical step in gp130-mediated terminal differentiation and growth arrest of a myeloid cell line. Proceedings of the National Academy of Sciences of the United States of America. 1996;93(9):3963–3966. doi: 10.1073/pnas.93.9.3963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Yoshida T, Iwamoto T, Adachi K, Yokota T, Miyake Y, Hamaguchi M. Functional analysis of the effect of forced activation of STAT3 on M1 mouse leukemia cells. International Journal of Molecular Medicine. 2005;15(2):269–275. [PubMed] [Google Scholar]
  • 54.Yamanaka Y, Nakajima K, Fukada T, Hibi M, Hirano T. Differentiation and growth arrest signals are generated through the cytoplasmic region of gp130 that is essential for Stat3 activation. The EMBO Journal. 1996;15(7):1557–1565. [PMC free article] [PubMed] [Google Scholar]
  • 55.Silver JS, Hunter CA. gp130 at the nexus of inflammation, autoimmunity, and cancer. Journal of Leukocyte Biology. 2010;88(6):1145–1156. doi: 10.1189/jlb.0410217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Fischer P, Lehmann U, Sobota RM, Schmitz J, Niemand C, Linnemann S, Haan S, et al. The role of the inhibitors of interleukin-6 signal transduction SHP2 and SOCS3 for desensitization of interleukin-6 signalling. The Biochemical Journal. 2004;378(Pt 2):449–460. doi: 10.1042/BJ20030893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Lin C, Jackson AL, Guo J, Linsley PS, Eisenman RN. Myc-regulated microRNAs attenuate embryonic stem cell differentiation. The EMBO Journal. 2009;28(20):3157–3170. doi: 10.1038/emboj.2009.254. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Tarantino C, Paolella G, Cozzuto L, Minopoli G, Pastore L, Parisi S, Russo T. miRNA 34a, 100, and 137 modulate differentiation of mouse embryonic stem cells. The FASEB Journal. 2010;24(9):3255–3263. doi: 10.1096/fj.09-152207. [DOI] [PubMed] [Google Scholar]
  • 59.Oskowitz AZ, Lu J, Penfornis P, Ylostalo J, McBride J, Flemington EK, Prockop DJ, Pochampally R. Human multipotent stromal cells from bone marrow and microRNA: regulation of differentiation and leukemia inhibitory factor expression. Proceedings of the National Academy of Sciences of the United States of America. 2008;105(47):18372–18377. doi: 10.1073/pnas.0809807105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hao J, Li T, Qi X, Zhao D, Zhao G. WNT/beta-catenin pathway up-regulates Stat3 and converges on LIF to prevent differentiation of mouse embryonic stem cells. Developmental Biology. 2006;290(1):81–91. doi: 10.1016/j.ydbio.2005.11.011. [DOI] [PubMed] [Google Scholar]
  • 61.Ogawa K, Nishinakamura R, Iwamatsu Y, Shimosato D, Niwa H. Synergistic action of Wnt and LIF in maintaining pluripotency of mouse ES cells. Biochemical and Biophysical Research Communications. 2006;343(1):159–166. doi: 10.1016/j.bbrc.2006.02.127. [DOI] [PubMed] [Google Scholar]
  • 62.Boiani M, Schöler HR. Regulatory networks in embryo-derived pluripotent stem cells. Nature Reviews. Molecular Cell Biology. 2005;6(11):872–884. doi: 10.1038/nrm1744. [DOI] [PubMed] [Google Scholar]
  • 63.Pera MF, Tam PPL. Extrinsic regulation of pluripotent stem cells. Nature. 2010;465(7299):713–720. doi: 10.1038/nature09228. [DOI] [PubMed] [Google Scholar]
  • 64.Kochegarov A. Small molecules for stem cells. Expert Opinion on Therapeutic Patents. 2009;19(3):275–281. doi: 10.1517/13543770802709010. [DOI] [PubMed] [Google Scholar]
  • 65.Niwa H. Mouse ES cell culture system as a model of development. Development, Growth & Differentiation. 2010;52(3):275–283. doi: 10.1111/j.1440-169X.2009.01166.x. [DOI] [PubMed] [Google Scholar]
  • 66.Silva J, Smith A. Capturing pluripotency. Cell. 2008;132(4):532–536. doi: 10.1016/j.cell.2008.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Ying Q, Wray J, Nichols J, Batlle-Morera L, Doble B, Woodgett J, Cohen P, Smith A. The ground state of embryonic stem cell self-renewal. Nature. 2008;453(7194):519–523. doi: 10.1038/nature06968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Chambers I, Tomlinson SR. The transcriptional foundation of pluripotency. Development. 2009;136(14):2311–2322. doi: 10.1242/dev.024398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Yang W, Wei W, Shi C, Zhu J, Ying W, Shen Y, Ye X, et al. Pluripotin combined with leukemia inhibitory factor greatly promotes the derivation of embryonic stem cell lines from refractory strains. Stem Cells. 2009;27(2):383–389. doi: 10.1634/stemcells.2008-0974. [DOI] [PubMed] [Google Scholar]
  • 70.Chen L, Yang M, Dawes J, Khillan JS. Suppression of ES cell differentiation by retinol (vitamin A) via the overexpression of Nanog. Differentiation. 2007;75(8):682–693. doi: 10.1111/j.1432-0436.2007.00169.x. [DOI] [PubMed] [Google Scholar]
  • 71.Chen L, Khillan JS. A novel signaling by vitamin A/retinol promotes self renewal of mouse embryonic stem cells by activating PI3K/Akt signaling pathway via insulin-like growth factor-1 receptor. Stem Cells. 2010;28(1):57–63. doi: 10.1002/stem.251. [DOI] [PubMed] [Google Scholar]
  • 72.Chazaud C, Yamanaka Y, Pawson T, Rossant J. Early lineage segregation between epiblast and primitive endoderm in mouse blastocysts through the Grb2-MAPK pathway. Developmental Cell. 2006;10(5):615–624. doi: 10.1016/j.devcel.2006.02.020. [DOI] [PubMed] [Google Scholar]
  • 73.Carter MG, Stagg CA, Falco G, Yoshikawa T, Bassey UC, Aiba K, Sharova LV, Shaik N, Ko MS. An in situ hybridization-based screen for heterogeneously expressed genes in mouse ES cells. Gene Expression Patterns. 2008;8:181–198. doi: 10.1016/j.gep.2007.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Toyooka Y, Shimosato D, Murakami K, Takahashi K, Niwa H. Identification and characterization of subpopulations in undifferentiated ES cell culture. Development. 2008;135:909–918. doi: 10.1242/dev.017400. [DOI] [PubMed] [Google Scholar]
  • 75.Kim MO, Lee YJ, Han HJ. Involvement of Cx43 phosphorylation in 5′-N-ethylcarboxamide-induced migration and proliferation of mouse embryonic stem cells. Journal of Cellular Physiology. 2010;224(1):187–194. doi: 10.1002/jcp.22117. [DOI] [PubMed] [Google Scholar]
  • 76.Todorova MG, Soria B, Quesada I. Gap junctional intercellular communication is required to maintain embryonic stem cells in a non-differentiated and proliferative state. Journal of Cellular Physiology. 2008;214(2):354–362. doi: 10.1002/jcp.21203. [DOI] [PubMed] [Google Scholar]
  • 77.Wörsdörfer P, Maxeiner S, Markopoulos C, Kirfel G, Wulf V, Auth T, Urschel S, von Maltzahn J, Willecke K. Connexin expression and functional analysis of gap junctional communication in mouse embryonic stem cells. Stem Cells. 2008;26(2):431–439. doi: 10.1634/stemcells.2007-0482. [DOI] [PubMed] [Google Scholar]
  • 78.Wong RCB, Pera MF, Pébay A. Role of gap junctions in embryonic and somatic stem cells. Stem Cell Reviews. 2008;4(4):283–292. doi: 10.1007/s12015-008-9038-9. [DOI] [PubMed] [Google Scholar]
  • 79.Saito R, Yamasaki T, Nagai Y, Wu J, Kajiho H, Yokoi T, Noda E, et al. CrxOS maintains the self-renewal capacity of murine embryonic stem cells. Biochemical and Biophysical Research Communications. 2009;390(4):1129–1135. doi: 10.1016/j.bbrc.2009.09.118. [DOI] [PubMed] [Google Scholar]
  • 80.Lian I, Kim J, Okazawa H, Zhao J, Zhao B, Yu J, Chinnaiyan A, et al. The role of YAP transcription coactivator in regulating stem cell self-renewal and differentiation. Genes & Development. 2010;24(11):1106–1118. doi: 10.1101/gad.1903310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Fan Y, Melhem MF, Chaillet JR. Forced expression of the homeobox-containing gene Pem blocks differentiation of embryonic stem cells. Developmental Biology. 1999;210(2):481–496. doi: 10.1006/dbio.1999.9279. [DOI] [PubMed] [Google Scholar]
  • 82.Lim LS, Loh Y, Zhang W, Li Y, Chen X, Wang Y, Bakre M, Ng H, Stanton LW. Zic3 is required for maintenance of pluripotency in embryonic stem cells. Molecular Biology of the Cell. 2007;18(4):1348–1358. doi: 10.1091/mbc.E06-07-0624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Lim LS, Hong FH, Kunarso G, Stanton LW. The pluripotency regulator Zic3 is a direct activator of the Nanog promoter in ESCs. Stem Cells. 2010;28(11):1961–1969. doi: 10.1002/stem.527. [DOI] [PubMed] [Google Scholar]
  • 84.Galan-Caridad JM, Harel S, Arenzana TL, Hou ZE, Doetsch FK, Mirny LA, Reizis B. Zfx controls the self-renewal of embryonic and hematopoietic stem cells. Cell. 2007;129(2):345–357. doi: 10.1016/j.cell.2007.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Mu W, Munroe RJ, Barker AK, Schimenti JC. PDCD2 is essential for inner cell mass development and embryonic stem cell maintenance. Developmental Biology. 2010;347(2):279–288. doi: 10.1016/j.ydbio.2010.08.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Amleh A, Nair SJ, Sun J, Sutherland A, Hasty P, Li R. Mouse cofactor of BRCA1 (Cobra1) is required for early embryogenesis. PLoS ONE. 2009;4(4):e5034. doi: 10.1371/journal.pone.0005034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Hu G, Kim J, Xu Q, Leng Y, Orkin SH, Elledge SJ. A genome-wide RNAi screen identifies a new transcriptional module required for self-renewal. Genes & Development. 2009;23(7):837–848. doi: 10.1101/gad.1769609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Ding L, Paszkowski-Rogacz M, Nitzsche A, Slabicki MM, Heninger A, de Vries I, Kittler R, et al. A genome-scale RNAi screen for Oct4 modulators defines a role of the Paf1 complex for embryonic stem cell identity. Cell Stem Cell. 2009;4(5):403–415. doi: 10.1016/j.stem.2009.03.009. [DOI] [PubMed] [Google Scholar]
  • 89.Ivanova N, Dobrin R, Lu R, Kotenko I, Levorse J, DeCoste C, Schafer X, Lun Y, Lemischka IR. Dissecting self-renewal in stem cells with RNA interference. Nature. 2006;442(7102):533–538. doi: 10.1038/nature04915. [DOI] [PubMed] [Google Scholar]
  • 90.Loh Y, Zhang W, Chen X, George J, Ng H. Jmjd1a and Jmjd2c histone H3 Lys 9 demethylases regulate self-renewal in embryonic stem cells. Genes & Development. 2007;21(20):2545–2557. doi: 10.1101/gad.1588207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Gaspar-Maia A, Alajem A, Polesso F, Sridharan R, Mason MJ, Heidersbach A, Ramalho-Santos J, et al. Chd1 regulates open chromatin and pluripotency of embryonic stem cells. Nature. 2009;460(7257):863–868. doi: 10.1038/nature08212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Dejosez M, Krumenacker JS, Zitur LJ, Passeri M, Chu L, Songyang Z, Thomson JA, Zwaka TP. Ronin is essential for embryogenesis and the pluripotency of mouse embryonic stem cells. Cell. 2008;133(7):1162–1174. doi: 10.1016/j.cell.2008.05.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Dejosez M, Levine SS, Frampton GM, Whyte WA, Stratton SA, Barton MC, Gunaratne PH, Young RA, Zwaka TP. Ronin/Hcf-1 binds to a hyperconserved enhancer element and regulates genes involved in the growth of embryonic stem cells. Genes & Development. 2010;24(14):1479–1484. doi: 10.1101/gad.1935210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Smith KN, Singh AM, Dalton S. Myc represses primitive endoderm differentiation in pluripotent stem cells. Cell Stem Cell. 2010;7(3):343–354. doi: 10.1016/j.stem.2010.06.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Tay Y, Zhang J, Thomson AM, Lim B, Rigoutsos I. MicroRNAs to Nanog, Oct4 and Sox2 coding regions modulate embryonic stem cell differentiation. Nature. 2008;455:1124–1128. doi: 10.1038/nature07299. [DOI] [PubMed] [Google Scholar]
  • 96.Bernstein E, Kim SY, Carmell MA, Murchison EP, Alcorn H, Li MZ, Mills AA, Elledge SJ, Anderson KV, Hannon GJ. Dicer is essential for mouse development. Nature Genetics. 2003;35(3):215–217. doi: 10.1038/ng1253. [DOI] [PubMed] [Google Scholar]
  • 97.Kanellopoulou C, Muljo SA, Kung AL, Ganesan S, Drapkin R, Jenuwein T, Livingston DM, Rajewsky K. Dicer-deficient mouse embryonic stem cells are defective in differentiation and centromeric silencing. Genes & Development. 2005;19(4):489–501. doi: 10.1101/gad.1248505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Murchison EP, Partridge JF, Tam OH, Cheloufi S, Hannon GJ. Characterization of Dicer-deficient murine embryonic stem cells. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(34):12135–12140. doi: 10.1073/pnas.0505479102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Babiarz JE, Ruby JG, Wang Y, Bartel DP, Blelloch R. Mouse ES cells express endogenous shRNAs, siRNAs, and other Microprocessor-independent, Dicer-dependent small RNAs. Genes & Development. 2008;22(20):2773–2785. doi: 10.1101/gad.1705308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Wang Y, Medvid R, Melton C, Jaenisch R, Blelloch R. DGCR8 is essential for microRNA biogenesis and silencing of embryonic stem cell self-renewal. Nature Genetics. 2007;39:380–385. doi: 10.1038/ng1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Wang Y, Baskerville S, Shenoy A, Babiarz JE, Baehner L, Blelloch R. Embryonic stem cell-specific microRNAs regulate the G1-S transition and promote rapid proliferation. Nature Genetics. 2008;40:1478–1483. doi: 10.1038/ng.250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Niwa H, Miyazaki J, Smith AG. Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nature Genetics. 2000;24(4):372–376. doi: 10.1038/74199. [DOI] [PubMed] [Google Scholar]
  • 103.Rizzino A. Transcription factors that behave as master regulators during mammalian embryogenesis function as molecular rheostats. The Biochemical Journal. 2008;411(2):e5–e7. doi: 10.1042/BJ20080479. [DOI] [PubMed] [Google Scholar]
  • 104.Kopp JL, Ormsbee BD, Desler M, Rizzino A. Small increases in the level of Sox2 trigger the differentiation of mouse embryonic stem cells. Stem Cells. 2008;26(4):903–911. doi: 10.1634/stemcells.2007-0951. [DOI] [PubMed] [Google Scholar]
  • 105.Trouillas M, Saucourt C, Guillotin B, Gauthereau X, Ding L, Buchholz F, Doss MX, et al. Three LIF-dependent signatures and gene clusters with atypical expression profiles, identified by transcriptome studies in mouse ES cells and early derivatives. BMC Genomics. 2009;10:73. doi: 10.1186/1471-2164-10-73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Palmqvist L, Glover CH, Hsu L, Lu M, Bossen B, Piret JM, Humphries RK, Helgason CD. Correlation of murine embryonic stem cell gene expression profiles with functional measures of pluripotency. Stem Cells. 2005;23(5):663–680. doi: 10.1634/stemcells.2004-0157. [DOI] [PubMed] [Google Scholar]
  • 107.Schulz H, Kolde R, Adler P, Aksoy I, Anastassiadis K, Bader M, Billon N, et al. The FunGenES database: a genomics resource for mouse embryonic stem cell differentiation. PLoS ONE. 2009;4(9):e6804. doi: 10.1371/journal.pone.0006804. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Loh Y, Wu Q, Chew J, Vega VB, Zhang W, Chen X, Bourque G, et al. The Oct4 and Nanog transcription network regulates pluripotency in mouse embryonic stem cells. Nature Genetics. 2006;38(4):431–440. doi: 10.1038/ng1760. [DOI] [PubMed] [Google Scholar]
  • 109.Cinelli P, Casanova EA, Uhlig S, Lochmatter P, Matsuda T, Yokota T, Rülicke T, Ledermann B, Bürki K. Expression profiling in transgenic FVB/N embryonic stem cells overexpressing STAT3. BMC Developmental Biology. 2008;8:57. doi: 10.1186/1471-213X-8-57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Storm MP, Kumpfmueller B, Thompson B, Kolde R, Vilo J, Hummel O, Schulz H, Welham MJ. Characterization of the phosphoinositide 3-kinase-dependent transcriptome in murine embryonic stem cells: identification of novel regulators of pluripotency. Stem Cells. 2009;27(4):764–775. doi: 10.1002/stem.3. [DOI] [PubMed] [Google Scholar]
  • 111.Rathjen PD, Nichols J, Toth S, Edwards DR, Heath JK, Smith AG. Developmentally programmed induction of differentiation inhibiting activity and the control of stem cell populations. Genes & Development. 1990;4(12B):2308–2318. doi: 10.1101/gad.4.12b.2308. [DOI] [PubMed] [Google Scholar]
  • 112.Burdon T, Stracey C, Chambers I, Nichols J, Smith A. Suppression of SHP-2 and ERK signalling promotes self-renewal of mouse embryonic stem cells. Developmental Biology. 1999;210(1):30–43. doi: 10.1006/dbio.1999.9265. [DOI] [PubMed] [Google Scholar]
  • 113.Duval D, Trouillas M, Thibault C, Dembelé D, Diemunsch F, Reinhardt B, Mertz AL, Dierich A, Boeuf H. Apoptosis and differentiation commitment: novel insights revealed by gene profiling studies in mouse embryonic stem cells. Cell Death and Differentiation. 2006;13(4):564–575. doi: 10.1038/sj.cdd.4401789. [DOI] [PubMed] [Google Scholar]
  • 114.Boeuf H, Merienne K, Jacquot S, Duval D, Zeniou M, Hauss C, Reinhardt B, et al. The ribosomal S6 kinases, cAMP-responsive element-binding, and STAT3 proteins are regulated by different leukemia inhibitory factor signaling pathways in mouse embryonic stem cells. The Journal of Biological Chemistry. 2001;276(49):46204–46211. doi: 10.1074/jbc.M106718200. [DOI] [PubMed] [Google Scholar]
  • 115.Li Y, McClintick J, Zhong L, Edenberg HJ, Yoder MC, Chan RJ. Murine embryonic stem cell differentiation is promoted by SOCS-3 and inhibited by the zinc finger transcription factor Klf4. Blood. 2005;105(2):635–637. doi: 10.1182/blood-2004-07-2681. [DOI] [PubMed] [Google Scholar]
  • 116.Jiang J, Chan Y, Loh Y, Cai J, Tong G, Lim C, Robson P, Zhong S, Ng H. A core Klf circuitry regulates self-renewal of embryonic stem cells. Nature Cell Biology. 2008;10(3):353–360. doi: 10.1038/ncb1698. [DOI] [PubMed] [Google Scholar]
  • 117.Niwa H, Ogawa K, Shimosato D, Adachi K. A parallel circuit of LIF signalling pathways maintains pluripotency of mouse ES cells. Nature. 2009;460(7251):118–122. doi: 10.1038/nature08113. [DOI] [PubMed] [Google Scholar]
  • 118.Parisi S, Passaro F, Aloia L, Manabe I, Nagai R, Pastore L, Russo T. Klf5 is involved in self-renewal of mouse embryonic stem cells. Journal of Cell Science. 2008;121(Pt 16):2629–2634. doi: 10.1242/jcs.027599. [DOI] [PubMed] [Google Scholar]
  • 119.Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–676. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
  • 120.Zhang P, Andrianakos R, Yang Y, Liu C, Lu W. Kruppel-like factor 4 (Klf4) prevents embryonic stem (ES) cell differentiation by regulating Nanog gene expression. The Journal of Biological Chemistry. 2010;285(12):9180–9189. doi: 10.1074/jbc.M109.077958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Hall J, Guo G, Wray J, Eyres I, Nichols J, Grotewold L, Morfopoulou S, et al. Oct4 and LIF/Stat3 additively induce Krüppel factors to sustain embryonic stem cell self-renewal. Cell Stem Cell. 2009;5(6):597–609. doi: 10.1016/j.stem.2009.11.003. [DOI] [PubMed] [Google Scholar]
  • 122.Moreau JF, Bonneville M, Godard A, Gascan H, Gruart V, Moore MA, Soulillou JP. Characterization of a factor produced by human T cell clones exhibiting eosinophil-activating and burst-promoting activities. Journal of Immunology. 1987;138(11):3844–3849. [PubMed] [Google Scholar]
  • 123.Dührsen U, Metcalf D. A model system for leukemic transformation of immortalized hemopoietic cells in irradiated recipient mice. Leukemia. 1988;2(6):329–333. [PubMed] [Google Scholar]
  • 124.Leary AG, Wong GG, Clark SC, Smith AG, Ogawa M. Leukemia inhibitory factor differentiation-inhibiting activity/human interleukin for DA cells augments proliferation of human hematopoietic stem cells. Blood. 1990;75(10):1960–1964. [PubMed] [Google Scholar]
  • 125.Verfaillie C, McGlave P. Leukemia inhibitory factor/human interleukin for DA cells: a growth factor that stimulates the in vitro development of multipotential human hematopoietic progenitors. Blood. 1991;77(2):263–270. [PubMed] [Google Scholar]
  • 126.Metcalf D, Gearing DP. Fatal syndrome in mice engrafted with cells producing high levels of the leukemia inhibitory factor. Proceedings of the National Academy of Sciences of the United States of America. 1989;86(15):5948–5952. doi: 10.1073/pnas.86.15.5948. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Metcalf D, Nicola NA, Gearing DP. Effects of injected leukemia inhibitory factor on hematopoietic and other tissues in mice. Blood. 1990;76(1):50–56. [PubMed] [Google Scholar]
  • 128.Ware CB, Horowitz MC, Renshaw BR, Hunt JS, Liggitt D, Koblar SA, Gliniak BC, McKenna HJ, Papayannopoulou T, Thoma B. Targeted disruption of the low-affinity leukemia inhibitory factor receptor gene causes placental, skeletal, neural and metabolic defects and results in perinatal death. Development. 1995;121(5):1283–1299. doi: 10.1242/dev.121.5.1283. [DOI] [PubMed] [Google Scholar]
  • 129.Dagoneau N, Scheffer D, Huber C, Al-Gazali LI, Di Rocco M, Godard A, Martinovic J, et al. Null leukemia inhibitory factor receptor (LIFR) mutations in Stuve-Wiedemann/Schwartz-Jampel type 2 syndrome. American Journal of Human Genetics. 2004;74(2):298–305. doi: 10.1086/381715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.García-Gómez I, Elvira G, Zapata AG, Lamana ML, Ramírez M, Castro JG, Arranz MG, Vicente A, Bueren J, García-Olmo D. Mesenchymal stem cells: biological properties and clinical applications. Expert Opinion on Biological Therapy. 2010;10(10):1453–1468. doi: 10.1517/14712598.2010.519333. [DOI] [PubMed] [Google Scholar]
  • 131.Lorgeot V, Rougier F, Fixe P, Cornu E, Praloran V, Denizot Y. Spontaneous and inducible production of leukaemia inhibitory factor by human bone marrow stromal cells. Cytokine. 1997;9(10):754–758. doi: 10.1006/cyto.1997.0225. [DOI] [PubMed] [Google Scholar]
  • 132.Metcalfe SM. Axotrophin and leukaemia inhibitory factor (LIF) in transplantation tolerance. Philosophical Transactions of the Royal Society of London. Series B: Biological Sciences. 2005;360:1687–1694. doi: 10.1098/rstb.2005.1697. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Nasef A, Mazurier C, Bouchet S, Francois S, Chapel A, Thierry D, Gorin NC, Fouillard L. Leukemia inhibitory factor: role in human mesenchymal stem cells mediated immunosuppression. Cellular Immunology. 2008;253:16–22. doi: 10.1016/j.cellimm.2008.06.002. [DOI] [PubMed] [Google Scholar]
  • 134.Szilvassy SJ, Weller KP, Chen B, Juttner CA, Tsukamoto A, Hoffman R. Partially differentiated ex vivo expanded cells accelerate hematologic recovery in myeloablated mice transplanted with highly enriched long-term repopulating stem cells. Blood. 1996;88(9):3642–3653. [PubMed] [Google Scholar]
  • 135.Schraml E, Fehrer C, Brunauer R, Lepperdinger G, Chesnokova V, Schauenstein K. lin-Sca-1+ cells and age-dependent changes of their proliferation potential are reliant on mesenchymal stromal cells and are leukemia inhibitory factor dependent. Gerontology. 2008;54(5):312–323. doi: 10.1159/000161736. [DOI] [PubMed] [Google Scholar]
  • 136.Dhawan J, Rando TA. Stem cells in postnatal myogenesis: molecular mechanisms of satellite cell quiescence, activation and replenishment. Trends in Cell Biology. 2005;15(12):666–673. doi: 10.1016/j.tcb.2005.10.007. [DOI] [PubMed] [Google Scholar]
  • 137.Montarras D, Morgan J, Collins C, Relaix F, Zaffran S, Cumano A, Partridge T, Buckingham M. Direct isolation of satellite cells for skeletal muscle regeneration. Science. 2005;309(5743):2064–2067. doi: 10.1126/science.1114758. [DOI] [PubMed] [Google Scholar]
  • 138.Buckingham M, Montarras D. Skeletal muscle stem cells. Current Opinion in Genetics & Development. 2008;18(4):330–336. doi: 10.1016/j.gde.2008.06.005. [DOI] [PubMed] [Google Scholar]
  • 139.Austin L, Burgess AW. Stimulation of myoblast proliferation in culture by leukaemia inhibitory factor and other cytokines. Journal of the Neurological Sciences. 1991;101(2):193–197. doi: 10.1016/0022-510X(91)90045-9. [DOI] [PubMed] [Google Scholar]
  • 140.Barnard W, Bower J, Brown MA, Murphy M, Austin L. Leukemia inhibitory factor (LIF) infusion stimulates skeletal muscle regeneration after injury: injured muscle expresses lif mRNA. Journal of the Neurological Sciences. 1994;123:108–113. doi: 10.1016/0022-510X(94)90211-9. [DOI] [PubMed] [Google Scholar]
  • 141.Diao Y, Wang X, Wu Z. SOCS1, SOCS3, and PIAS1 promote myogenic differentiation by inhibiting the leukemia inhibitory factor-induced JAK1/STAT1/STAT3 pathway. Molecular and Cellular Biology. 2009;29(18):5084–5093. doi: 10.1128/MCB.00267-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Yang Y, Xu Y, Li W, Wang G, Song Y, Yang G, Han X, Du Z, Sun L, Ma K. STAT3 induces muscle stem cell differentiation by interaction with myoD. Cytokine. 2009;46(1):137–141. doi: 10.1016/j.cyto.2008.12.015. [DOI] [PubMed] [Google Scholar]
  • 143.Spangenburg EE, Booth FW. Leukemia inhibitory factor restores the hypertrophic response to increased loading in the LIF(−/−) mouse. Cytokine. 2006;34(3–4):125–130. doi: 10.1016/j.cyto.2006.05.001. [DOI] [PubMed] [Google Scholar]
  • 144.Gearing DP, Ziegler SF, Comeau MR, Friend D, Thoma B, Cosman D, Park L, Mosley B. Proliferative responses and binding properties of hematopoietic cells transfected with low-affinity receptors for leukemia inhibitory factor, oncostatin M, and ciliary neurotrophic factor. Proceedings of the National Academy of Sciences of the United States of America. 1994;91(3):1119–1123. doi: 10.1073/pnas.91.3.1119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Bazan E, Alonso FJ, Redondo C, Lopez-Toledano MA, Alfaro JM, Reimers D, Herranz AS, et al. In vitro and in vivo characterization of neural stem cells. Histology and Histopathology. 2004;19:1261–1275. doi: 10.14670/HH-19.1261. [DOI] [PubMed] [Google Scholar]
  • 146.Abrous DN, Koehl M, Le Moal M. Adult neurogenesis: from precursors to network and physiology. Physiological Reviews. 2005;85(2):523–569. doi: 10.1152/physrev.00055.2003. [DOI] [PubMed] [Google Scholar]
  • 147.Suh H, Deng W, Gage FH. Signaling in adult neurogenesis. Annual Review of Cell and Developmental Biology. 2009;25:253–275. doi: 10.1146/annurev.cellbio.042308.113256. [DOI] [PubMed] [Google Scholar]
  • 148.Andersen RK, Zimmer J, Wahlberg LU, Meyer M. Effect of leukemia inhibitory factor on long-term propagation of precursor cells derived from rat forebrain subventricular zone and ventral mesencephalon. Experimental Neurology. 2008;211(1):301–310. doi: 10.1016/j.expneurol.2008.02.007. [DOI] [PubMed] [Google Scholar]
  • 149.Müller S, Chakrapani BPS, Schwegler H, Hofmann H, Kirsch M. Neurogenesis in the dentate gyrus depends on ciliary neurotrophic factor and signal transducer and activator of transcription 3 signaling. Stem Cells. 2009;27(2):431–441. doi: 10.1634/stemcells.2008-0234. [DOI] [PubMed] [Google Scholar]
  • 150.Sun Y, Pollard S, Conti L, Toselli M, Biella G, Parkin G, Willatt L, Falk A, Cattaneo E, Smith A. Long-term tripotent differentiation capacity of human neural stem (NS) cells in adherent culture. Molecular and Cellular Neurosciences. 2008;38(2):245–258. doi: 10.1016/j.mcn.2008.02.014. [DOI] [PubMed] [Google Scholar]
  • 151.Covey MV, Levison SW. Leukemia inhibitory factor participates in the expansion of neural stem/progenitors after perinatal hypoxia/ischemia. Neuroscience. 2007;148(2):501–509. doi: 10.1016/j.neuroscience.2007.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Ishibashi T, Lee PR, Baba H, Fields RD. Leukemia inhibitory factor regulates the timing of oligodendrocyte development and myelination in the postnatal optic nerve. Journal of Neuroscience Research. 2009;87(15):3343–3355. doi: 10.1002/jnr.22173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Yamanaka S. Patient-specific pluripotent stem cells become even more accessible. Cell Stem Cell. 2010;7(1):1–2. doi: 10.1016/j.stem.2010.06.009. [DOI] [PubMed] [Google Scholar]
  • 154.Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–872. doi: 10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
  • 155.Elefanty AG, Blelloch R, Passegué E, Wernig M, Mummery CL. On the streets of San Francisco: highlights from the ISSCR Annual Meeting 2010. Cell Stem Cell. 2010;7(4):443–450. doi: 10.1016/j.stem.2010.09.002. [DOI] [PubMed] [Google Scholar]
  • 156.Yoshida Y, Takahashi K, Okita K, Ichisaka T, Yamanaka S. Hypoxia enhances the generation of induced pluripotent stem cells. Cell Stem Cell. 2009;5(3):237–241. doi: 10.1016/j.stem.2009.08.001. [DOI] [PubMed] [Google Scholar]
  • 157.Zhao T, Xu Y. p53 and stem cells: new developments and new concerns. Trends in Cell Biology. 2010;20(3):170–175. doi: 10.1016/j.tcb.2009.12.004. [DOI] [PubMed] [Google Scholar]
  • 158.Cotterman R, Knoepfler PS. N-Myc regulates expression of pluripotency genes in neuroblastoma including lif, klf2, klf4, and lin28b. PLoS ONE. 2009;4(6):e5799. doi: 10.1371/journal.pone.0005799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Auernhammer CJ, Melmed S. Leukemia-inhibitory factor-neuroimmune modulator of endocrine function. Endocrine Reviews. 2000;21(3):313–345. doi: 10.1210/er.21.3.313. [DOI] [PubMed] [Google Scholar]
  • 160.Brivanlou AH, Darnell JE. Signal transduction and the control of gene expression. Science. 2002;295(5556):813–818. doi: 10.1126/science.1066355. [DOI] [PubMed] [Google Scholar]
  • 161.Pires-daSilva A, Sommer RJ. The evolution of signalling pathways in animal development. Nature Reviews. Genetics. 2003;4(1):39–49. doi: 10.1038/nrg977. [DOI] [PubMed] [Google Scholar]
  • 162.Huising MO, Kruiswijk CP, Flik G. Phylogeny and evolution of class-I helical cytokines. The Journal of Endocrinology. 2006;189(1):1–25. doi: 10.1677/joe.1.06591. [DOI] [PubMed] [Google Scholar]
  • 163.Hombría JCG, Brown S. The fertile field of Drosophila Jak/STAT signalling. Current Biology. 2002;12(16):R569–R575. doi: 10.1016/S0960-9822(02)01057-6. [DOI] [PubMed] [Google Scholar]
  • 164.Horiuchi H, Furusawa S, Matsuda H. Maintenance of chicken embryonic stem cells in vitro. Methods in Molecular Biology. 2006;329:17–34. doi: 10.1385/1-59745-037-5:17. [DOI] [PubMed] [Google Scholar]
  • 165.Fujiki K, Nakao M, Dixon B. Molecular cloning and characterisation of a carp (Cyprinus carpio) cytokine-like cDNA that shares sequence similarity with IL-6 subfamily cytokines CNTF, OSM and LIF. Developmental and Comparative Immunology. 2003;27(2):127–136. doi: 10.1016/S0145-305X(02)00074-5. [DOI] [PubMed] [Google Scholar]
  • 166.Abe T, Mikekado T, Haga S, Kisara Y, Watanabe K, Kurokawa T, Suzuki T. Identification, cDNA cloning, and mRNA localization of a zebrafish ortholog of leukemia inhibitory factor. Comparative Biochemistry and Physiology. Part B: Biochemistry & Molecular Biology. 2007;147(1):38–44. doi: 10.1016/j.cbpb.2006.12.019. [DOI] [PubMed] [Google Scholar]
  • 167.Hanington PC, Belosevic M. Interleukin-6 family cytokine M17 induces differentiation and nitric oxide response of goldfish (Carassius auratus L.) macrophages. Developmental and Comparative Immunology. 2007;31(8):817–829. doi: 10.1016/j.dci.2006.12.001. [DOI] [PubMed] [Google Scholar]
  • 168.Duong CV, Geissen M, Rohrer H. The developmental expression of vasoactive intestinal peptide (VIP) in cholinergic sympathetic neurons depends on cytokines signaling through LIFRbeta-containing receptors. Development. 2002;129(6):1387–1396. doi: 10.1242/dev.129.6.1387. [DOI] [PubMed] [Google Scholar]
  • 169.Hanington PC, Patten SA, Reaume LM, Waskiewicz AJ, Belosevic M, Ali DW. Analysis of leukemia inhibitory factor and leukemia inhibitory factor receptor in embryonic and adult zebrafish (Danio rerio) Developmental Biology. 2008;314(2):250–260. doi: 10.1016/j.ydbio.2007.10.012. [DOI] [PubMed] [Google Scholar]
  • 170.Nishinakamura R, Matsumoto Y, Matsuda T, Ariizumi T, Heike T, Asashima M, Yokota T. Activation of Stat3 by cytokine receptor gp130 ventralizes Xenopus embryos independent of BMP-4. Developmental Biology. 1999;216(2):481–490. doi: 10.1006/dbio.1999.9518. [DOI] [PubMed] [Google Scholar]
  • 171.Nichane M, Ren X, Bellefroid EJ. Self-regulation of Stat3 activity coordinates cell-cycle progression and neural crest specification. The EMBO Journal. 2010;29(1):55–67. doi: 10.1038/emboj.2009.313. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Brons IG, Smithers LE, Trotter MW, Rugg-Gunn P, Sun B, Lopes SMCDS, Howlett SK, et al. Derivation of pluripotent epiblast stem cells from mammalian embryos. Nature. 2007;448:191–195. doi: 10.1038/nature05950. [DOI] [PubMed] [Google Scholar]
  • 173.Tesar PJ, Chenoweth JG, Brook FA, Davies TJ, Evans EP, Mack DL, Gardner RL, McKay RD. New cell lines from mouse epiblast share defining features with human embryonic stem cells. Nature. 2007;448:196–199. doi: 10.1038/nature05972. [DOI] [PubMed] [Google Scholar]
  • 174.Bao S, Tang F, Li X, Hayashi K, Gillich A, Lao K, Surani MA. Epigenetic reversion of post-implantation epiblast to pluripotent embryonic stem cells. Nature. 2009;461:1292–1295. doi: 10.1038/nature08534. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Guo G, Yang J, Nichols J, Hall JS, Eyres I, Mansfield W, Smith A. Klf4 reverts developmentally programmed restriction of ground state pluripotency. Development. 2009;136:1063–1069. doi: 10.1242/dev.030957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.De Matos DG, Miller K, Scott R, Tran CA, Kagan D, Nataraja SG, Clark A, Palmer S. Leukemia inhibitory factor induces cumulus expansion in immature human and mouse oocytes and improves mouse two-cell rate and delivery rates when it is present during mouse in vitro oocyte maturation. Fertility and Sterility. 2008;90:2367–2375. doi: 10.1016/j.fertnstert.2007.10.061. [DOI] [PubMed] [Google Scholar]
  • 177.Kanatsu-Shinohara M, Inoue K, Ogonuki N, Miki H, Yoshida S, Toyokuni S, Lee J, Ogura A, Shinohara T. Leukemia inhibitory factor enhances formation of germ cell colonies in neonatal mouse testis culture. Biology of Reproduction. 2007;76:55–62. doi: 10.1095/biolreprod.106.055863. [DOI] [PubMed] [Google Scholar]
  • 178.Marwood M, Visser K, Salamonsen LA, Dimitriadis E. Interleukin-11 and leukemia inhibitory factor regulate the adhesion of endometrial epithelial cells: implications in fertility regulation. Endocrinology. 2009;150:2915–2923. doi: 10.1210/en.2008-1538. [DOI] [PubMed] [Google Scholar]
  • 179.Mikhaylova IV, Jääskeläinen T, Jääskeläinen J, Palvimo JJ, Voutilainen R. Leukemia inhibitory factor as a regulator of steroidogenesis in human NCI-H295R adrenocortical cells. The Journal of Endocrinology. 2008;199:435–444. doi: 10.1677/JOE-08-0377. [DOI] [PubMed] [Google Scholar]
  • 180.Metcalf D, Hilton D, Nicola NA. Leukemia inhibitory factor can potentiate murine megakaryocyte production in vitro. Blood. 1991;77:2150–2153. [PubMed] [Google Scholar]
  • 181.Nasef A, Mazurier C, Bouchet S, François S, Chapel A, Thierry D, Gorin N, Fouillard L. Leukemia inhibitory factor: Role in human mesenchymal stem cells mediated immunosuppression. Cellular Immunology. 2008;253:16–22. doi: 10.1016/j.cellimm.2008.06.002. [DOI] [PubMed] [Google Scholar]
  • 182.Sun L, Ma K, Wang H, Xiao F, Gao Y, Zhang W, Wang K, Gao X, Ip N, Wu Z. JAK1-STAT1-STAT3, a key pathway promoting proliferation and preventing premature differentiation of myoblasts. The Journal of Cell Biology. 2007;179:129–138. doi: 10.1083/jcb.200703184. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Alter J, Rozentzweig D, Bengal E. Inhibition of myoblast differentiation by tumor necrosis factor alpha is mediated by c-Jun N-terminal kinase 1 and leukemia inhibitory factor. The Journal of Biological Chemistry. 2008;283:23224–23234. doi: 10.1074/jbc.M801379200. [DOI] [PubMed] [Google Scholar]
  • 184.Paradis H, Gendron RL. LIF transduces contradictory signals on capillary outgrowth through induction of stat3 and (P41/43)MAP kinase. Journal of Cell Science. 2000;113(Pt 23):4331–4339. doi: 10.1242/jcs.113.23.4331. [DOI] [PubMed] [Google Scholar]
  • 185.Murphy M, Reid K, Hilton DJ, Bartlett PF. Generation of sensory neurons is stimulated by leukemia inhibitory factor. Proceedings of the National Academy of Sciences of the United States of America. 1991;88:3498–3501. doi: 10.1073/pnas.88.8.3498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Andersen RK, Widmer HR, Zimmer J, Wahlberg LU, Meyer M. Leukemia inhibitory factor favours neurogenic differentiation of long-term propagated human midbrain precursor cells. Neuroscience Letters. 2009;464:203–208. doi: 10.1016/j.neulet.2009.08.050. [DOI] [PubMed] [Google Scholar]
  • 187.Carter DA, Dick AD, Mayer EJ. CD133+ adult human retinal cells remain undifferentiated in Leukaemia Inhibitory Factor (LIF) BMC Ophthalmology. 2009;9:1. doi: 10.1186/1471-2415-9-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Chen H, Ma H, Sytwu H, Wang H, Chen CV, Liu S, et al. Neural stem cells secrete factors that promote auditory cell proliferation via a leukemia inhibitory factor signaling pathway. Journal of Neuroscience Research. 2010;88(15):3308–3318. doi: 10.1002/jnr.22492. [DOI] [PubMed] [Google Scholar]
  • 189.Barasch J, Yang J, Ware CB, Taga T, Yoshida K, Erdjument-Bromage H, Tempst P, Parravicini E, Malach S, Aranoff T. Mesenchymal to epithelial conversion in rat metanephros is induced by LIF. Cell. 1999;99:377–386. doi: 10.1016/S0092-8674(00)81524-X. [DOI] [PubMed] [Google Scholar]
  • 190.Kanda J, Uchiyama T, Tomosugi N, Higuchi M, Uchiyama T, Kawabata H. Oncostatin M and leukemia inhibitory factor increase hepcidin expression in hepatoma cell lines. International Journal of Hematology. 2009;90:545–552. doi: 10.1007/s12185-009-0443-x. [DOI] [PubMed] [Google Scholar]
  • 191.Lumaki F, Basso SM, Orlando R. Cytokines, thyroid diseases and thyroid cancer. Cytokine. 2010;50(3):299–233. doi: 10.1016/j.cyto.2010.03.005. [DOI] [PubMed] [Google Scholar]
  • 192.Quaglino A, Schere-Levy C, Romorini L, Meiss RP, Kordon EC. Mouse mammary tumors display Stat3 activation dependent on leukemia inhibitory factor signaling. Breast Cancer Research. 2007;9:R69. doi: 10.1186/bcr1777. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Wysoczynski M, Miekus K, Jankowski K, Wanzeck J, Bertolone S, Janowska-Wieczorek A, Ratajczak J, Ratajczak MZ. Leukemia inhibitory factor: a newly identified metastatic factor in rhabdomyosarcomas. Cancer Research. 2007;67:2131–2140. doi: 10.1158/0008-5472.CAN-06-1021. [DOI] [PubMed] [Google Scholar]
  • 194.McKenzie RC, Szepietowski J. Cutaneous leukemia inhibitory factor and its potential role in the development of skin tumors. Dermatologic Surgery. 2004;30:279–290. doi: 10.1111/j.1524-4725.2004.30087.x. [DOI] [PubMed] [Google Scholar]
  • 195.Maruta S, Takiguchi S, Ueyama M, Kataoka Y, Oda Y, Tsuneyoshi M, Iguchi H. A role for leukemia inhibitory factor in melanoma-induced bone metastasis. Clinical & Experimental Metastasis. 2009;26:133–141. doi: 10.1007/s10585-008-9223-x. [DOI] [PubMed] [Google Scholar]
  • 196.Peñuelas S, Anido J, Prieto-Sánchez RM, Folch G, Barba I, Cuartas I, García-Dorado D, Poca MA, Sahuquillo J, Baselga J, Seoane J. TGF-[beta] Increases Glioma-Initiating Cell Self-Renewal through the Induction of LIF in Human Glioblastoma. Cancer Cell. 2009;15:315–327. doi: 10.1016/j.ccr.2009.02.011. [DOI] [PubMed] [Google Scholar]
  • 197.Kurek JB, Bower JJ, Romanella M, Koentgen F, Murphy M, Austin L. The role of leukemia inhibitory factor in skeletal muscle regeneration. Muscle Nerve. Muscle & Nerve. 1997;20:815–822. doi: 10.1002/(SICI)1097-4598(199707)20:7<815::AID-MUS5>3.0.CO;2-A. [DOI] [PubMed] [Google Scholar]
  • 198.Kubota Y, Hirashima M, Kishi K, Stewart CL, Suda T. Leukemia inhibitory factor regulates microvessel density by modulating oxygen-dependent VEGF expression in mice. Journal of Clinical Investigation. 2008;118:2393–2403. doi: 10.1172/JCI34882. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Bozec A, Bakiri L, Hoebertz A, Eferl R, Schilling AF, Komnenovic V, Scheuch H, Priemel M, Stewart CL, Amling M, Wagner EF. Osteoclast size is controlled by Fra-2 through LIF/LIF-receptor signalling and hypoxia. Nature. Nature. 2008;454:221–225. doi: 10.1038/nature07019. [DOI] [PubMed] [Google Scholar]
  • 200.Sugiura S, Lahav R, Han J, Kou SY, Banner LR, de Pablo F, Patterson PH. Leukaemia inhibitory factor is required for normal inflammatory responses to injury in the peripheral and central nervous systems in vivo and is chemotactic for macrophages in vitro. The European Journal of Neuroscience. 2000;12:457–466. doi: 10.1046/j.1460-9568.2000.00922.x. [DOI] [PubMed] [Google Scholar]
  • 201.Bugga L, Gadient RA, Kwan K, Stewart CL, Patterson PH. Analysis of neuronal and glial phenotypes in brains of mice deficient in leukemia inhibitory factor. Journal of Neurobiology. 1998;36:509–524. doi: 10.1002/(SICI)1097-4695(19980915)36:4<509::AID-NEU5>3.0.CO;2-#. [DOI] [PubMed] [Google Scholar]
  • 202.Marriott MP, Emery B, Cate HS, Binder MD, Kemper D, Wu Q, Kolbe S, Gordon IR, Wang H, Egan G, Murray S, Butzkueven H, Kilpatrick TJ. Leukemia inhibitory factor signaling modulates both central nervous system demyelination and myelin repair. Glia. 2008;56:686–698. doi: 10.1002/glia.20646. [DOI] [PubMed] [Google Scholar]
  • 203.Cohen JE, Fields RD. Activity-dependent neuron-glial signaling by ATP and leukemiainhibitory factor promotes hippocampal glial cell development. Neuron Glia Biology. 2008;4:43–55. doi: 10.1017/S1740925X09000076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Bürgi S, Samardzija M, Grimm C. Endogenous leukemia inhibitory factor protects photoreceptor cells against light-induced degeneration. Molecular Vision. 2009;15:1631–1637. [PMC free article] [PubMed] [Google Scholar]
  • 205.Kirsch M, Trautmann N, Ernst M, Hofmann H. Involvement of gp130-associated cytokine signaling in Müller cell activation following optic nerve lesion. Glia. 2010;58:768–779. doi: 10.1002/glia.20961. [DOI] [PubMed] [Google Scholar]
  • 206.Bauer S, Rasika S, Han J, Mauduit C, Raccurt M, Morel G, Jourdan F, Benahmed M, Moyse E, Patterson PH. Leukemia inhibitory factor is a key signal for injury-induced neurogenesis in the adult mouse olfactory epithelium. The Journal of Neuroscience. 2003;23:1792–1803. doi: 10.1523/JNEUROSCI.23-05-01792.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Banner LR, Patterson PH, Allchorne A, Poole S, Woolf CJ. Leukemia inhibitory factor is an anti-inflammatory and analgesic cytokine. The Journal of Neuroscience. 1998;18:5456–5462. doi: 10.1523/JNEUROSCI.18-14-05456.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Chesnokova V, Kariagina A, Melmed S. Opposing effects of pituitary leukemia inhibitory factor and SOCS-3 on the ACTH axis response to inflammation. American Journal of Physiology. Endocrinology and Metabolism. 2002;282:E1110–E1118. doi: 10.1152/ajpendo.00442.2001. [DOI] [PubMed] [Google Scholar]

Articles from Stem Cell Reviews are provided here courtesy of Springer

RESOURCES