Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
Add more filters

Publication year range
1.
Annu Rev Cell Dev Biol ; 30: 647-75, 2014.
Article in English | MEDLINE | ID: mdl-25288119

ABSTRACT

Mouse embryonic stem (ES) cells perpetuate in vitro the broad developmental potential of naïve founder cells in the preimplantation embryo. ES cells self-renew relentlessly in culture but can reenter embryonic development seamlessly, differentiating on schedule to form all elements of the fetus. Here we review the properties of these remarkable cells. Arising from the stability, homogeneity, and equipotency of ES cells, we consider the concept of a pluripotent ground state. We evaluate the authenticity of ES cells in relation to cells in the embryo and examine their utility for dissecting mechanisms that confer pluripotency and that execute fate choice. We summarize current knowledge of the transcription factor circuitry that governs the ES cell state and discuss the opportunity to expose molecular logic further through iterative computational modeling and experimentation. Finally, we present a perspective on unresolved questions, including the challenge of deriving ground state pluripotent stem cells from non-rodent species.


Subject(s)
Embryonic Stem Cells/cytology , Animals , Asymmetric Cell Division , Blastocyst/cytology , Cell Culture Techniques , Cell Lineage , Cells, Cultured , Cellular Reprogramming , Coculture Techniques , Culture Media , Culture Media, Serum-Free , Embryonal Carcinoma Stem Cells/cytology , Embryonic Stem Cells/physiology , Fibroblasts/cytology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Genes, Reporter , Germ Layers/cytology , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Intercellular Signaling Peptides and Proteins/physiology , Leukemia Inhibitory Factor/physiology , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , Transcription Factors/pharmacology , Transcription Factors/physiology
2.
Development ; 146(24)2019 12 16.
Article in English | MEDLINE | ID: mdl-31740534

ABSTRACT

Embryonic stem cells (ESCs) exist in at least two states that transcriptionally resemble different stages of embryonic development. Naïve ESCs resemble peri-implantation stages and primed ESCs the pre-gastrulation epiblast. In mouse, primed ESCs give rise to definitive endoderm in response to the pathways downstream of Nodal and Wnt signalling. However, when these pathways are activated in naïve ESCs, they differentiate to a cell type resembling early primitive endoderm (PrE), the blastocyst-stage progenitor of the extra-embryonic endoderm. Here, we apply this context dependency to human ESCs, showing that activation of Nodal and Wnt signalling drives the differentiation of naïve pluripotent cells toward extra-embryonic PrE, or hypoblast, and these can be expanded as an in vitro model for naïve extra-embryonic endoderm (nEnd). Consistent with observations made in mouse, human PrE differentiation is dependent on FGF signalling in vitro, and we show that, by inhibiting FGF receptor signalling, we can simplify naïve pluripotent culture conditions, such that the inhibitor requirements closer resemble those used in mouse. The expandable nEnd cultures reported here represent stable extra-embryonic endoderm, or human hypoblast, cell lines.This article has an associated 'The people behind the papers' interview.


Subject(s)
Endoderm/embryology , Leukemia Inhibitory Factor/physiology , Nodal Signaling Ligands/physiology , Pluripotent Stem Cells/physiology , Wnt Signaling Pathway/physiology , Animals , Cells, Cultured , Embryo, Mammalian , Embryonic Development/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Endoderm/cytology , Endoderm/metabolism , Gene Expression Regulation, Developmental , Germ Layers/cytology , Germ Layers/physiology , Humans , Leukemia Inhibitory Factor/metabolism , Mice , Nodal Signaling Ligands/metabolism , Signal Transduction/physiology
3.
Gynecol Endocrinol ; 37(8): 725-729, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33974468

ABSTRACT

AIM: Study the effect of low-dose aspirin on the endometrial receptivity in endometriosis rat models. MATERIALS AND METHODS: This study is to explore the expressions of progesterone receptor and LIF among three groups of endometriosis rat models: control group (n = 12), EMs group (n = 15), and aspirin group (n = 17). The expressions of progesterone receptor (PR), PRA, PRB, and leukemia inhibitory factor receptor (LIFR) in eutopic endometrium were determined using immunohistochemistry technology, western blot, and qRT-PCR. The levels of LIF in eutopic endometrium and serum were detected by western blot, qRT-PCR, and ELISA. RESULTS: The expressions of PR, PRA, and PRB protein were significantly increased in the eutopic endometrium after low-dose aspirin treatment, and the level of PRB mRNA was also increased while the ratio of PRA/PRB mRNA was decreased in the eutopic endometrium. The levels of LIF in eutopic endometrium and serum were increased compared with the untreated endometriosis rats. However, the expression of LIFR was not statistically different among the three groups. CONCLUSIONS: The results suggest that the low-dose aspirin treatment could downregulate progesterone resistance and increase the expression of LIF of endometriosis rats during the implantation window, which could improve endometrial receptivity and enhance the pregnant rate of endometriosis. It may provide a potential treatment method for endometriosis-related infertility.


Subject(s)
Aspirin/administration & dosage , Embryo Implantation/physiology , Endometriosis/drug therapy , Leukemia Inhibitory Factor/drug effects , Progesterone/physiology , Receptors, Progesterone/analysis , Animals , Endometriosis/complications , Endometriosis/metabolism , Endometrium/chemistry , Female , Infertility, Female/drug therapy , Infertility, Female/etiology , Leukemia Inhibitory Factor/analysis , Leukemia Inhibitory Factor/physiology , Leukemia Inhibitory Factor Receptor alpha Subunit/analysis , Leukemia Inhibitory Factor Receptor alpha Subunit/physiology , Male , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Progesterone/drug effects
4.
Dev Biol ; 445(2): 145-155, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30448440

ABSTRACT

The mammary gland (MG) is a unique organ responsible for milk synthesis, secretion, and involution to prepare the gland for subsequent lactation. The mammary epithelial cells (MECs), which are the milk synthesizing units of the MG, proliferate, differentiate, undergo apoptosis and regenerate following a cyclic pathway of lactation - involution - lactation, fine-tuning these molecular events through hormones, growth factors and other regulatory molecules. The developmental stages of the MG are embryonic, prepubertal, pubertal, pregnancy, lactation and involution, with major developmental processes occurring after puberty. The involution stage includes interesting physiological processes such as MEC apoptosis, matrix remodeling, and the generation of cells regaining the shape of a virgin MG. Signal transducer and activator of transcription 3 (STAT3) is the established master regulator of this process and aberrant expression of STAT3 leads to subnormal involution and may induce neoplasia. Several studies have reported on the molecular mechanism of MG involution with substantial knowledge being gained about this process; however, a deep understanding of this phenomenon has yet to be attained. This review focuses deeply on the molecular details of post-lactational regression, the signaling pathways involved in the lactation-involution cycle, and the latest developments in STAT3-associated MG neoplasia. Deep insight into the involution process will pave the way towards understanding the biology, apoptosis, and oncogenesis of the MG.


Subject(s)
Mammary Glands, Animal/growth & development , Mammary Glands, Animal/physiology , Animals , Apoptosis/genetics , Breast Neoplasms/etiology , Cytokines/genetics , Cytokines/physiology , Disease Progression , Epithelial Cells/cytology , Epithelial Cells/physiology , Extracellular Matrix/physiology , Female , Glycolipids/metabolism , Glycoproteins/metabolism , Humans , Lactation/genetics , Lactation/physiology , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/physiology , Lipid Droplets , Mammary Glands, Animal/anatomy & histology , Mice , MicroRNAs/genetics , Models, Biological , Pregnancy , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/physiology , Signal Transduction , Transforming Growth Factor beta/physiology
5.
Sheng Li Xue Bao ; 72(1): 105-114, 2020 Feb 25.
Article in Zh | MEDLINE | ID: mdl-32099988

ABSTRACT

Embryo implantation is crucial for the establishment and maintenance of successful pregnancy and requires the synchronization between implantation-competent blastocyst and receptive uterus. In assisted reproductive technologies, recognition of uterine receptivity is the limiting factor for improving pregnancy rate. It has been previously reported that embryo implantation involves the activation and inactivation of numerous signaling molecules which may influence the proliferation and differentiation of uterine epithelial cells, epithelial polarity, luminal closure, embryo orientation, epithelial-stromal interactions, gland development, etc. Here we summarize the function of estrogen, progesterone, leukemia inhibitory factor (LIF), microRNA (miRNA), channel protein and signaling pathways in embryo implantation and explore their regulatory network to provide theoretical basis for the treatment of infertility and development of safe and efficient contraceptives.


Subject(s)
Embryo Implantation , Uterus/physiology , Blastocyst/physiology , Estrogens/physiology , Female , Humans , Leukemia Inhibitory Factor/physiology , MicroRNAs/genetics , Pregnancy , Progesterone/physiology , Signal Transduction
6.
Development ; 142(20): 3488-99, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26395492

ABSTRACT

Embryonic stem cells (ESCs) are pluripotent cell lines that can be maintained indefinitely in an early developmental state. ESC culture conditions almost always require the cytokine LIF to maintain self-renewal. As ESCs are not homogeneous but contain multiple populations reminiscent of the blastocyst, identifying the target cells of LIF is necessary to understand the propagation of pluripotency. We recently found that LIF acts under self-renewing conditions to stimulate the fraction of ESCs that express extraembryonic markers, but has little impact on pluripotent gene expression. Here, we report that LIF has two distinct roles: it blocks early epiblast (Epi) differentiation, and it supports the expansion of primitive endoderm (PrE)-primed ESCs and PrE in vivo. We find that activation of JAK/STAT signalling downstream of LIF occurs initially throughout the pre-implantation embryo, but later marks the PrE. Moreover, the addition of LIF to cultured embryos increases the GATA6(+) PrE population, whereas inhibition of JAK/STAT signalling reduces both NANOG(+) epiblast and GATA6(+) PrE. The reduction of the NANOG(+) Epi might be explained by its precocious differentiation to later Epi derivatives, whereas the increase in PrE is mediated both by an increase in proliferation and inhibition of PrE apoptosis that is normally triggered in embryos with an excess of GATA6(+) cells. Thus, it appears that the relative size of the PrE is determined by the number of LIF-producing cells in the embryo. This suggests a mechanism by which the embryo adjusts the relative ratio of the primary lineages in response to experimental manipulation.


Subject(s)
Blastocyst/cytology , Endoderm/cytology , Gene Expression Regulation, Developmental , Leukemia Inhibitory Factor/physiology , Animals , Apoptosis , Cell Differentiation , Cell Lineage , Cytokines/metabolism , Embryonic Development , Embryonic Stem Cells/cytology , Female , Flow Cytometry , GATA6 Transcription Factor/metabolism , Gene Expression Profiling , Interleukin-6/metabolism , Janus Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Phenotype , Pluripotent Stem Cells/cytology , STAT3 Transcription Factor/metabolism , Time Factors
7.
Adv Exp Med Biol ; 1099: 125-139, 2018.
Article in English | MEDLINE | ID: mdl-30306520

ABSTRACT

Opioid analgesics remain the most effective and widely used analgesics for the management of moderate to severe pain, including cancer pain and chronic non-cancer pain. However, the efficacy of long-term opioid analgesics is attenuated by tolerance and/or hyperalgesia after long-term use, preventing adequate pain relief under stable opioid dosages for chronic pain patients. Classical neuron-centered concepts about tolerance, such as internalization of opioid receptors, upregulation of N-methyl-D-aspartate receptor function, or downregulation of glutamate transporter activity, can only partially explain the phenomenon of tolerance. Recent evidence revealing glial activation and upregulation of inflammatory mediators in the rodent central nervous system has confirmed the pivotal role of neuroinflammation in neuropathic pain or opioid tolerance, or both. However, human evidence is still sparse.Based on our clinical practice, we conducted translational research by investigating the cerebrospinal fluid (CSF) cytokine and chemokine profiles of opioid-tolerant patients after research ethic committee approval. CSF samples from opioid-tolerant patients and opioid-naive subjects were compared. We found CXCL1, CXCL12, and leukemia inhibitory factor (LIF) were significantly upregulated among the opioid-tolerant patients and positively correlated with the opioid dosage.We translated these findings back to lab animal experiment; after induction of tolerance by morphine infusion, the spinal cord expression of CXCL1, CXCL12, and LIF were all upregulated. Although CXCL1 and CXCL12 infusion alone did not affect baseline tail-flick latency, morphine analgesic efficacy dropped significantly after intrathecal infusion of CXCL1 and CXCL12. After establishing tolerance by intrathecal continuous infusion of morphine, tolerance development was accelerated by co-administration of CXCL1 and CXCL12. In parallel, the effect was attenuated by co-administration of CXCL1- or CXCL12-neutralizing antibody or concordant receptor antagonists.On the contrary, although chronic morphine administration still induced LIF upregulation in rat spinal cords, intrathecal injection of LIF potentiated the analgesic action of morphine and delayed the development of morphine tolerance. Upregulation of endogenously released LIF by long-term use of opioids might counterbalance the tolerance induction effects of other pro-inflammatory cytokines.CXCL1, CXCL12, and LIF are upregulated in both opioid-tolerant patients and rodents. The onset and extent of opioid tolerance were affected by modulating the intrathecal CXCL1/CXCR2, CXCL12/CXCR4, and LIF signaling and could be novel drug targets for the treatment of opioid tolerance.


Subject(s)
Analgesics, Opioid/pharmacology , Chemokine CXCL12/physiology , Chemokine CXCL1/physiology , Drug Tolerance , Inflammation/physiopathology , Leukemia Inhibitory Factor/physiology , Animals , Humans , Rats , Spinal Cord/drug effects
8.
Am J Physiol Cell Physiol ; 313(5): C487-C500, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28768641

ABSTRACT

IL-6 and leukemia inhibitory factor (LIF), members of the IL-6 family of cytokines, play recognized paradoxical roles in skeletal muscle mass regulation, being associated with both growth and atrophy. Overload or muscle contractions can induce a transient increase in muscle IL-6 and LIF expression, which has a regulatory role in muscle hypertrophy. However, the cellular mechanisms involved in this regulation have not been completely identified. The induction of mammalian target of rapamycin complex 1 (mTORC1)-dependent myofiber protein synthesis is an established regulator of muscle hypertrophy, but the involvement of the IL-6 family of cytokines in this process is poorly understood. Therefore, we investigated the acute effects of IL-6 and LIF administration on mTORC1 signaling and protein synthesis in C2C12 myotubes. The role of glycoprotein 130 (gp130) receptor and downstream signaling pathways, including phosphoinositide 3-kinase (PI3K)-Akt-mTORC1 and signal transducer and activator of transcription 3 (STAT3)-suppressor of cytokine signaling 3 (SOCS3), was investigated by administration of specific siRNA or pharmaceutical inhibitors. Acute administration of IL-6 and LIF induced protein synthesis, which was accompanied by STAT3 activation, Akt-mTORC1 activation, and increased SOCS3 expression. This induction of protein synthesis was blocked by both gp130 siRNA knockdown and Akt inhibition. Interestingly, STAT3 inhibition or Akt downstream mTORC1 signaling inhibition did not fully block the IL-6 or LIF induction of protein synthesis. SOCS3 siRNA knockdown increased basal protein synthesis and extended the duration of the protein synthesis induction by IL-6 and LIF. These results demonstrate that either IL-6 or LIF can activate gp130-Akt signaling axis, which induces protein synthesis via mTORC1-independent mechanisms in cultured myotubes. However, IL-6- or LIF-induced SOCS3 negatively regulates the activation of myotube protein synthesis.


Subject(s)
Interleukin-6/physiology , Muscle Fibers, Skeletal/metabolism , Myoblasts/metabolism , Protein Biosynthesis/physiology , Animals , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cells, Cultured , Cytokines/pharmacology , Cytokines/physiology , Interleukin-6/pharmacology , Leukemia Inhibitory Factor/pharmacology , Leukemia Inhibitory Factor/physiology , Mice , Muscle Fibers, Skeletal/drug effects , Myoblasts/drug effects , Protein Biosynthesis/drug effects
9.
Hepatology ; 64(5): 1606-1622, 2016 11.
Article in English | MEDLINE | ID: mdl-27533020

ABSTRACT

Increasing evidence supports a role for N-myc downstream-regulated gene 2 (NDRG2) deregulation in tumorigenesis. We investigated the roles and mechanisms of NDRG2 in human cholangiocarcinoma (CCA) progression. In the present study, expression of NDRG2, microRNA (miR)-181c and leukemia inhibitory factor (LIF) in human CCA and adjacent nontumor tissues were examined. The effects of NDRG2 on CCA tumor growth and metastasis were determined both in vivo and in vitro. The role of the NDRG2/LIF/miR-181c signaling pathway in cholangiocarcinogenesis and metastasis were investigated both in vivo and in vitro. The results showed that human CCA tissues exhibited decreased levels of NDRG2 and increased levels of miR-181c and LIF compared with nontumor tissues. NDRG2 could inhibit CCA cell proliferation, chemoresistance, and metastasis both in vitro and in vivo. We found that NDRG2 is a target gene of miR-181c, and the down-regulation of NDRG2 was attributed to miR-181c overexpression in CCA. Furthermore, miR-181c can be activated by LIF treatment, whereas NDRG2 could inhibit LIF transcription through disrupting the binding between Smad, small mothers against decapentaplegic complex and LIF promoter. Down-regulation of NDRG2 and overexpression of miR-181c or LIF are significantly associated with a poorer overall survival (OS) in CCA patients. Finally, we found that a combination of NDRG2, miR-181c, and LIF expression is a strong predictor of prognosis in CCA patients. CONCLUSION: These results establish the counteraction between NDRG2 and LIF/miR-181c as a key mechanism that regulates cholangiocarcinogenesis and metastasis. Our results elucidated a novel pathway in NDRG2-mediated inhibition of cholangiocarcinogenesis and metastasis and suggest new therapeutic targets, including NDRG2, LIF, miR-181c, and transforming growth factor beta, in CCA prevention and treatment. (Hepatology 2016;64:1606-1622).


Subject(s)
Bile Duct Neoplasms/etiology , Cholangiocarcinoma/etiology , Feedback, Physiological , Leukemia Inhibitory Factor/physiology , MicroRNAs/physiology , Proteins/physiology , Tumor Suppressor Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Disease Progression , Female , Humans , Male , Mice , Middle Aged , Signal Transduction
10.
Br J Anaesth ; 117(4): 512-520, 2016 Oct.
Article in English | MEDLINE | ID: mdl-28077540

ABSTRACT

BACKGROUND: The efficacy of opioids typically decreases after long-term use owing to the development of tolerance. Glial activation and the upregulation of proinflammatory cytokines are related to the induction of tolerance. We investigated the effect of leukemia inhibitory factor (LIF) on morphine analgesia and tolerance. METHODS: LIF concentrations in rat spinal cords were measured by polymerase chain reaction (PCR) and enzyme-linked immunosorbent assay (ELISA) after morphine administration. LIF distribution was examined using confocal microscopy. To evaluate the effects of LIF on morphine analgesia and tolerance, LIF was intrathecally administered 30 min before morphine injection. The analgesic effect of morphine was evaluated by measuring tail-flick latency. Human LIF concentrations from the cerebrospinal fluid (CSF) of opioid tolerant patients were also determined by specific ELISA. RESULTS: Chronic morphine administration upregulated LIF concentrations in rat spinal cords. Intrathecal injection of LIF potentiated the analgesic action of morphine. Patch clamp recording of spinal cord slices showed that LIF enhanced DAMGO ([D-Ala2, N-MePhe4, Gly-ol]-enkephalin)-induced outward potassium current. The development of tolerance was markedly suppressed by exogenous LIF, whereas neutralizing the endogenously released LIF with anti-LIF antibodies accelerated the tolerance induction. Moreover, LIF concentrations in the CSF of opioid-tolerant patients were higher than those in the opioid-naive controls. CONCLUSIONS: Intrathecal administration of LIF potentiated morphine antinociceptive activity and attenuated the development of morphine tolerance. Upregulation of endogenously released LIF by long-term use of opioids might counterbalance the tolerance induction effects of other proinflammatory cytokines. LIF might be a novel drug candidate for inhibiting opioid tolerance induction.


Subject(s)
Analgesics, Opioid/pharmacology , Leukemia Inhibitory Factor/physiology , Morphine/pharmacology , Animals , Cytokines/analysis , Drug Tolerance , Humans , Male , Rats , Rats, Sprague-Dawley
11.
Rev Neurosci ; 26(4): 443-59, 2015.
Article in English | MEDLINE | ID: mdl-25879318

ABSTRACT

Leukemia inhibitory factor (LIF) is a multi-function cytokine that has various effects on different tissues and cell types in rodents and humans; however, its insufficiency has a relatively mild impact. This could explain why only some aspects of LIF activity are in the time-light, whereas other aspects are not well known. In this review, the LIF structure, signaling pathway, and primary roles in the development and function of an organism are reviewed, and the effects of LIF on stem cell growth and differentiation, which are important for its use in cell culturing, are described. The focus is on the roles of LIF in central nervous system development and on the modulation of its physiological functions as well as the involvement of LIF in the pathogenesis of brain diseases and injuries. Finally, LIF and its signaling pathway are discussed as potential targets of therapeutic interventions to influence both negative phenomena and regenerative processes following brain injury.


Subject(s)
Leukemia Inhibitory Factor/physiology , Nervous System Diseases/pathology , Nervous System Physiological Phenomena , Animals , Humans , Leukemia Inhibitory Factor/genetics , Nervous System/embryology , Nervous System/growth & development
12.
Zhongguo Dang Dai Er Ke Za Zhi ; 16(9): 933-8, 2014 Sep.
Article in Zh | MEDLINE | ID: mdl-25229963

ABSTRACT

OBJECTIVE: To study the changes of endogenous leukemia inhibitory factor (LIF) in neonatal rats with periventricular leukomalacia (PVL). METHODS: A PVL model of 3-day-old Wistar rats was prepared by left carotid artery ligation followed by 6% oxygen for 4 hours. The rats were sacrificed at 1, 3, 7, 14 and 28 days of hypoxia ischemia (HI), and the brain tissues were sampled. Real-Time PCR and Western blot methods were applied to analyze the expression of LIF mRNA and protein. Double staining immunofluorescence was used to detect the co-expression of LIF and GFAP. RESULTS: At 1, 3 and 7 days of HI, LIF protein level in the PVL group was higher than in the control group (P<0.01). In the PVL group, the LIF protein level on the third day after HI reached a peak and was higher than the other time points (P<0.01). The change of LIF mRNA expression showed the same tendency with LIF protein. The double staining immunofluorescence showed a co-expression of LIF and GFAP. CONCLUSIONS: LIF mRNA and LIF protein expression in astrocytes show a trend of initial increase followed by steady decline in neonatal rats with PVL, suggesting that endogenous LIF may participate in the repair of PVL.


Subject(s)
Leukemia Inhibitory Factor/physiology , Leukomalacia, Periventricular/metabolism , Animals , Animals, Newborn , Disease Models, Animal , Female , Glial Fibrillary Acidic Protein/analysis , Leukemia Inhibitory Factor/analysis , Leukemia Inhibitory Factor/genetics , Leukomalacia, Periventricular/pathology , Male , RNA, Messenger/analysis , Rats , Rats, Wistar
13.
J Neurosci ; 32(6): 2100-9, 2012 Feb 08.
Article in English | MEDLINE | ID: mdl-22323722

ABSTRACT

New CNS neurons and glia are generated throughout adulthood from endogenous neural stem and progenitor cells. These progenitors can respond to injury, but their ability to proliferate, migrate, differentiate, and survive is usually insufficient to replace lost cells and restore normal function. Potentiating the progenitor response with exogenous factors is an attractive strategy for the treatment of nervous system injuries and neurodegenerative and demyelinating disorders. Previously, we reported that delivery of leukemia inhibitory factor (LIF) to the CNS stimulates the self-renewal of neural stem cells and the proliferation of parenchymal glial progenitors. Here we identify these parenchymal glia as oligodendrocyte (OL) progenitor cells (OPCs) and show that LIF delivery stimulates their proliferation through the activation of gp130 receptor signaling within these cells. Importantly, this effect of LIF on OPC proliferation can be harnessed to enhance the generation of OLs that express myelin proteins and reform nodes of Ranvier in the context of chronic demyelination in the adult mouse hippocampus. Our findings, considered together with the known beneficial effects of LIF on OL and neuron survival, suggest that LIF has both reparative and protective activities that make it a promising potential therapy for CNS demyelinating disorders and injuries.


Subject(s)
Cell Proliferation , Hippocampus/physiology , Leukemia Inhibitory Factor/physiology , Myelin Sheath/metabolism , Oligodendroglia/physiology , Stem Cells/physiology , Animals , Cell Proliferation/drug effects , Female , Hippocampus/cytology , Hippocampus/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Myelin Sheath/pathology , Neurogenesis/physiology , Oligodendroglia/pathology , Ranvier's Nodes/pathology , Ranvier's Nodes/physiology , Stem Cells/pathology
14.
Stem Cells ; 30(3): 561-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22213097

ABSTRACT

Oxygen levels in tissues including the embryonic brain are lower than those in the atmosphere. We reported previously that Notch signal activation induces demethylation of astrocytic genes, conferring astrocyte differentiation ability on midgestational neural precursor cells (mgNPCs). Here, we show that the oxygen sensor hypoxia-inducible factor 1α (HIF1α) plays a critical role in astrocytic gene demethylation in mgNPCs by cooperating with the Notch signaling pathway. Expression of constitutively active HIF1α and a hyperoxic environment, respectively, promoted and impeded astrocyte differentiation in the developing brain. Our findings suggest that hypoxia contributes to the appropriate scheduling of mgNPC fate determination.


Subject(s)
Brain/growth & development , Epigenesis, Genetic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neural Stem Cells/physiology , Receptors, Notch/metabolism , Signal Transduction , Animals , Astrocytes/metabolism , Astrocytes/physiology , Brain/cytology , Brain/metabolism , Cell Differentiation , Cell Hypoxia , Cells, Cultured , DNA Methylation , Gene Expression Regulation, Developmental , Glial Fibrillary Acidic Protein/genetics , Leukemia Inhibitory Factor/physiology , Mice , Mice, Inbred ICR , Neural Stem Cells/metabolism , Oxygen/metabolism
15.
Stem Cells ; 30(11): 2450-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22969033

ABSTRACT

Totipotency of embryonic stem cells (ESCs) is controlled at the transcriptional level by a handful of transcription factors (TFs) that promote stemness and prevent differentiation. One of the most enriched DNA elements in promoters and enhancers of genes specifically active in ESCs is the CCAAT box, which is recognized by NF-Y, a trimer with histone-like subunits--NF-YB/NF--YC--and the sequence-specific NF-YA. We show that the levels of the short NF-YA isoform--NF-YAs--is high in mouse ESCs (mESCs) and drops after differentiation; a dominant negative mutant affects expression of important stem cells genes, directly and indirectly. Protein transfections of TAT-NF-YAs stimulate growth and compensate for withdrawal of leukemia inhibitory factor (LIF) in cell cultures. Bioinformatic analysis identifies NF-Y sites as highly enriched in genomic loci of stem TFs in ESCs. Specifically, 30%-50% of NANOG peaks have NF-Y sites and indeed NF-Y-binding is required for NANOG association to DNA. These data indicate that NF-Y belongs to the restricted circle of TFs that govern mESCs, and, specifically, that NF-YAs is the active isoform in these cells.


Subject(s)
CCAAT-Binding Factor/metabolism , Embryonic Stem Cells/metabolism , Animals , CCAAT-Binding Factor/genetics , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells/physiology , Gene Expression , Gene Expression Regulation , Gene Regulatory Networks , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Leukemia Inhibitory Factor/physiology , Mice , Nanog Homeobox Protein , Promoter Regions, Genetic , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism
16.
Stem Cells ; 30(11): 2387-99, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22899336

ABSTRACT

Various growth factor cocktails have been used to proliferate and then differentiate human neural progenitor (NP) cells derived from embryonic stem cells (ESC) for in vitro and in vivo studies. However, the cytokine leukemia inhibitory factor (LIF) has been largely overlooked. Here, we demonstrate that LIF significantly enhanced in vitro survival and promoted differentiation of human ESC-derived NP cells. In NP cells, as well as NP-derived neurons, LIF reduced caspase-mediated apoptosis and reduced both spontaneous and H2O2-induced reactive oxygen species in culture. In vitro, NP cell proliferation and the yield of differentiated neurons were significantly higher in the presence of LIF. In NP cells, LIF enhanced cMyc phosphorylation, commonly associated with self-renewal/proliferation. Also, in differentiating NP cells LIF activated the phosphoinositide 3-kinase and signal transducer and activator of transcription 3 pathways, associated with cell survival and reduced apoptosis. When differentiated in LIF+ media, neurite outgrowth and ERK1/2 phosphorylation were potentiated together with increased expression of gp130, a component of the LIF receptor complex. NP cells, pretreated in vitro with LIF, were effective in reducing infarct volume in a model of focal ischemic stroke but LIF did not lead to significantly improved initial NP cell survival over nontreated NP cells. Our results show that LIF signaling significantly promotes human NP cell proliferation, survival, and differentiation in vitro. Activated LIF signaling should be considered in cell culture expansion systems for future human NP cell-based therapeutic transplant studies.


Subject(s)
Embryonic Stem Cells/physiology , Leukemia Inhibitory Factor/physiology , Nerve Growth Factors/physiology , Neural Stem Cells/physiology , Neurons/physiology , Animals , Apoptosis , Cell Differentiation , Cell Proliferation , Cell Survival , Cells, Cultured , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/therapy , Leukemia Inhibitory Factor/administration & dosage , Male , Mice , Mice, Inbred C57BL , Nerve Growth Factors/administration & dosage , Neural Stem Cells/metabolism , Neural Stem Cells/transplantation , Neurites/metabolism , Neurites/physiology , Neurons/metabolism , Phosphorylation , Protein Processing, Post-Translational , Reactive Oxygen Species/metabolism , Signal Transduction , Transcriptional Activation
17.
Mol Cell Biochem ; 377(1-2): 107-19, 2013 May.
Article in English | MEDLINE | ID: mdl-23430356

ABSTRACT

Adipose-derived stem cells (ASCs) have been defined as cells that undergo sustained in vitro growth and have multilineage differentiation potential. However, the identity and purification of ASCs has proved elusive due to the lack of specific markers and poor understanding of their physiological roles. Here, we prospectively isolated and identified a restricted homogeneous subpopulation of ASCs (Lin(-)CD271(+)Sca-1(+)) from mouse adipose tissues on the basis of cell-surface markers. Individual ASCs generated colony-forming unit-fibroblast at a high frequency and could differentiate into adipocytes, osteoblasts, and chondrocytes in vitro. Expansion of ASCs in a large quantity was feasible in medium supplemented with fibroblast growth factor-2 and leukemia inhibitory factor, without loss of adipogenic and osteogenic differentiation capacity. Moreover, we found that the transplanted ASCs can differentiate into adipocytes in adipogenic microenvironment in vivo and osteoblasts in osteogenic microenvironment in vivo. Thus we proved that Lin, CD271, and Sca-1 could be used as the specific markers to purify ASCs from adipose tissue. The method we established to identify ASCs as defined in vivo entities will help develop ASCs transplantation as a new therapeutic strategy for bone regeneration and adipose tissue regeneration in clinic.


Subject(s)
Adipogenesis , Antigens, Ly/metabolism , Membrane Proteins/metabolism , Naphthalenes/metabolism , Osteogenesis , Stem Cells/physiology , Abdominal Fat/cytology , Adapalene , Animals , Bone Regeneration , Cell Proliferation , Cells, Cultured , Fibroblast Growth Factor 2/physiology , Flow Cytometry , Hydroxyapatites/chemistry , Leukemia Inhibitory Factor/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Side-Population Cells/metabolism , Stem Cell Transplantation , Tissue Engineering , Tissue Scaffolds/chemistry
18.
Dev Neurosci ; 34(5): 449-62, 2012.
Article in English | MEDLINE | ID: mdl-23258129

ABSTRACT

Stem cells rely on extracellular signals produced by the niche, which dictate their ability to self-renew, expand and differentiate. It is essential to have sensitive and reproducible methods of either quantifying or isolating these stem cells and progenitors to understand their intrinsic properties and how extrinsic signals regulate their development. However, stem cells are difficult to distinguish from multipotential progenitors, which may look and act like them. Here we define a 4-color flow cytometry panel using CD133, LeX, CD140a, NG2 to define a neural stem cell (NSC) as well as 4 classes of multipotential progenitors and 3 classes of bipotential progenitors, several of which have not been described previously. We performed gain and loss of function studies for leukemia inhibitory factor (LIF) and showed a depletion of NSCs, a subset of multipotential neural precursors and immature oligodendrocytes in LIF null mice. Gain of function studies showed that LIF increased the abundance of these precursors. Our studies also show that these NPs have differential requirements for LIF and ciliary neurotrophic factor (CNTF) and for epidermal growth factor (EGF), fibroblast growth factor (FGF-2) and platelet-derived growth factor (PDGF) for their propagation in vitro. Surprisingly, the related cytokine, CNTF, was less potent than LIF in increasing the NSCs and more potent than LIF in increasing the PDGF responsive multipotential precursors. Finally, we show that LIF increases the expression of the core transcription factors: Klf4, Fbx15, Nanog, Sox2 and c-Myc. Altogether our FACS (fluorescence-activated cell sorter) analyses reveal that the neonatal subventricular zone is far more heterogeneous than previously suspected and our studies provide new insights into the signals and mechanisms that regulate their self-renewal and proliferation.


Subject(s)
Cerebral Ventricles/physiology , Homeostasis/physiology , Leukemia Inhibitory Factor/physiology , Neural Stem Cells/physiology , Stem Cells/physiology , AC133 Antigen , Animals , Animals, Newborn , Antigens/genetics , Antigens/metabolism , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Proliferation , Cell Separation , Cerebral Ventricles/cytology , Flow Cytometry , Fucosyltransferases/pharmacology , Glycoproteins/genetics , Glycoproteins/metabolism , Kruppel-Like Factor 4 , Leukemia Inhibitory Factor/genetics , Mice , Mice, Inbred C57BL , Oligodendroglia/physiology , Peptides/genetics , Peptides/metabolism , Phenotype , Proteoglycans/genetics , Proteoglycans/metabolism , Real-Time Polymerase Chain Reaction , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Signal Transduction
19.
Biol Reprod ; 86(2): 54, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22053093

ABSTRACT

It is well established that syncytium formation involves the fusion of mononucleated trophoblasts into a multinucleated structure and the secretion of hormonal factors, such as human chorionic gonadotropin (hCG). These morphological and biochemical changes are regulated by a plethora of ligands, which upon binding to specific receptors trigger the activation of many signaling pathways, such as janus kinase/signal transducer and activator of transcription (JAK/STAT) and the mitogen-activated protein (MAP) kinase extracellular signal-regulated kinases 1 and 2 (MAPK3/1). We used the forskolin-induced syncytialization of trophoblastlike BeWo cells to characterize at the cellular level the effect mediated by leukemia inhibitory factor (LIF) on trophoblast differentiation and to describe its action at the molecular level. Forskolin induces both hCG secretion and BeWo cell syncytial fusion. Although LIF had no effect on the undifferentiated state of the cells, the cytokine generated a strong reduction in forskolin-induced hCG release. In contrast to its effect on hCG secretion, LIF exerts a synergistic effect toward forskolin-induced fusion. LIF reduced hormonal production through a STAT1- and STAT3-dependent mechanism, whereas MAPK3/1 was not involved in this process. However, both types of signaling molecules were required to mediate the action of LIF in forskolin-induced cell fusion. These data provide novel insights into the regulation of trophoblast cell differentiation by LIF and describe for the first time the molecular mechanism underlying the effect of the cytokine.


Subject(s)
Cell Differentiation/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Janus Kinases/physiology , Leukemia Inhibitory Factor/physiology , Mitogen-Activated Protein Kinases/physiology , STAT Transcription Factors/physiology , Signal Transduction/physiology , Trophoblastic Tumor, Placental Site/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Choriocarcinoma/pathology , Colforsin/pharmacology , Female , Humans , Janus Kinase 1/physiology , Janus Kinase 2/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Pregnancy , STAT1 Transcription Factor/physiology , STAT3 Transcription Factor/physiology , Signal Transduction/drug effects , Uterine Neoplasms/pathology
20.
Mol Hum Reprod ; 18(4): 204-15, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22053055

ABSTRACT

The epithelial cell surface of the endometrium undergoes substantial biochemical changes to allow embryo attachment and implantation in early pregnancy. We hypothesized that tissue macrophages influence these events to promote uterine receptivity. To investigate the role of macrophages in regulating epithelial cell expression of genes linked to glycan-mediated embryo adhesion, Ishikawa, RL95-2 and HEC1A endometrial epithelial cells were cultured alone or with unactivated or lipopolysaccharide-activated monocytic U937 cells, separated using transwell inserts. Expression of mRNAs encoding two α1,2-fucosyltransferases (FUT1, FUT2) was increased in all three epithelial cell lines following co-culture with U937 cells, and was associated with increased fucosylation of cell surface glycoproteins detected using lectins from Ulex europaeus (UEA-1) and Dolichos biflorus (DBA). FUT1 induction by U937 cells also occurred in primary endometrial epithelial cells collected in luteal but not proliferative phase. Activation of the interleukin-6 (IL6)/leukemia inhibitory factor (LIF) cytokine signaling pathway with phosphorylation of STAT3 and elevated SOCS3 mRNA expression was evident in epithelial cells stimulated by U937 co-culture. Several recombinant macrophage-secreted cytokines exerted stimulatory or inhibitory effects on FUT1 and FUT2 mRNA expression, and the macrophage-derived cytokine LIF partially replicated the effects of U937 cells on both FUT1 and FUT2 expression and UEA-1 and DBA lectin reactivity in Ishikawa cells. These results suggest that macrophage-derived factors including LIF might facilitate development of an implantation-receptive endometrium by regulating surface glycan structures in epithelial cells. Abnormal phenotypes or altered abundance of uterine macrophages could contribute to the pathophysiology of primary unexplained infertility in women.


Subject(s)
Endometrium/metabolism , Fucosyltransferases/genetics , Gene Expression Regulation , Macrophages/physiology , Cells, Cultured , Coculture Techniques , Cytokines/physiology , Embryo Implantation , Epithelial Cells/metabolism , Female , Fucosyltransferases/metabolism , Humans , Interleukin-6/metabolism , Interleukin-6/physiology , Leukemia Inhibitory Factor/metabolism , Leukemia Inhibitory Factor/physiology , RNA, Messenger/metabolism , U937 Cells , Galactoside 2-alpha-L-fucosyltransferase
SELECTION OF CITATIONS
SEARCH DETAIL