Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Int J Mol Sci ; 22(14)2021 Jul 12.
Article in English | MEDLINE | ID: mdl-34299091

ABSTRACT

The differentiation of human pluripotent stem cells (hPSCs) to neural stem cells (NSCs) is the key initial event in neurogenesis and is thought to be dependent on the family of Wnt growth factors, their receptors and signaling proteins. The delineation of the transcriptional pathways that mediate Wnt-induced hPSCs to NSCs differentiation is vital for understanding the global genomic mechanisms of the development of NSCs and, potentially, the creation of new protocols in regenerative medicine. To understand the genomic mechanism of Wnt signaling during NSCs development, we treated hPSCs with Wnt activator (CHIR-99021) and leukemia inhibitory factor (LIF) in a chemically defined medium (N2B27) to induce NSCs, referred to as CLNSCs. The CLNSCs were subcultured for more than 40 passages in vitro; were positive for AP staining; expressed neural progenitor markers such as NESTIN, PAX6, SOX2, and SOX1; and were able to differentiate into three neural lineage cells: neurons, astrocytes, and oligodendrocytes in vitro. Our transcriptome analyses revealed that the Wnt and Hedgehog signaling pathways regulate hPSCs cell fate decisions for neural lineages and maintain the self-renewal of CLNSCs. One interesting network could be the deregulation of the Wnt/ß-catenin signaling pathway in CLNSCs via the downregulation of c-MYC, which may promote exit from pluripotency and neural differentiation. The Wnt-induced spinal markers HOXA1-4, HOXA7, HOXB1-4, and HOXC4 were increased, however, the brain markers FOXG1 and OTX2, were absent in the CLNSCs, indicating that CLNSCs have partial spinal cord properties. Finally, a CLNSC simple culture condition, when applied to hPSCs, supports the generation of NSCs, and provides a new and efficient cell model with which to untangle the mechanisms during neurogenesis.


Subject(s)
Biomarkers/analysis , Neural Stem Cells/cytology , Neurogenesis , Neurons/cytology , Pluripotent Stem Cells/cytology , Transcriptome , Wnt Signaling Pathway , Cell Differentiation , Cells, Cultured , Humans , Leukemia Inhibitory Factor/administration & dosage , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism
2.
PLoS One ; 16(2): e0243727, 2021.
Article in English | MEDLINE | ID: mdl-33534866

ABSTRACT

In vitro embryo production systems are limited by their inability to consistently produce embryos with the competency to develop to the blastocyst stage, survive cryopreservation, and establish a pregnancy. Previous work identified a combination of three cytokines [fibroblast growth factor 2 (FGF2), leukemia inhibitory factor (LIF), and insulin-like growth factor 1 (IGF1)], called FLI, that we hypothesize improve preimplantation development of bovine embryos in vitro. To test this hypothesis, FLI was supplemented into oocyte maturation or embryo culture medium. Embryos were produced in vitro using abattoir-derived oocytes and fertilized with sperm from a single bull known to have high fertility. After an 18-20 h fertilization period, putative zygotes were cultured in synthetic oviductal fluid (SOF) for 8 days. The addition of FLI to the oocyte maturation medium increased (P < 0.05) the dissociation of transzonal projections at 12, 18, and 24 h of maturation, as well as, the proportion of oocytes that reached the metaphase II stage of meiosis. Additionally, lipid content was decreased (P < 0.05) in the blastocyst stage embryo. The addition of FLI during the culture period increased development to the blastocyst stage, cytoskeleton integrity, and survival following slow freezing, as well as, decreased post thaw cell apoptosis (P < 0.05). In conclusion, the supplementation of these cytokines in vitro has the potential to alleviate some of the challenges associated with the cryo-survival of in vitro produced bovine embryos through improving embryo development and embryo quality.


Subject(s)
Cattle/embryology , Cryopreservation/veterinary , Embryo, Mammalian/embryology , Fibroblast Growth Factor 2 , Insulin-Like Growth Factor I , Leukemia Inhibitory Factor , Animals , Blastocyst/cytology , Blastocyst/drug effects , Blastocyst/ultrastructure , Cryopreservation/methods , Embryo, Mammalian/drug effects , Embryo, Mammalian/ultrastructure , Female , Fertilization in Vitro/methods , Fertilization in Vitro/veterinary , Fibroblast Growth Factor 2/administration & dosage , Fibroblast Growth Factor 2/pharmacology , In Vitro Oocyte Maturation Techniques/methods , In Vitro Oocyte Maturation Techniques/veterinary , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/pharmacology , Leukemia Inhibitory Factor/administration & dosage , Leukemia Inhibitory Factor/pharmacology , Pregnancy
3.
J Reprod Immunol ; 142: 103195, 2020 11.
Article in English | MEDLINE | ID: mdl-32927320

ABSTRACT

The functional competence of leukemia inhibitory factor (LIF), as immunocontraceptive vaccine in mice, was investigated. Balb/c mice were divided into two groups of vaccinated and controls. The recombinant human LIF (rhLIF) protein and phosphate buffer saline was emulsified with Freund's adjuvant and injected into vaccinated and control groups, respectively. Theinhibition of implantation was evaluated in mice uterine. The concentration of secreted interferon-γ (IFN-γ) and interleukin (IL)-4 were measured in cultured splenocyte of mice stimulated by rhLIF. The expressions of immune responsive gene 1 (IRG-1), cochlin (COCH), amphiregulin(Ar), and heparin-binding EGF-like growth factor (HB-EGF) genes were determined. Mice were assessed for inhibition of fertility after delivery, reversibility of immune response against rhLIF, and survival rate. Active immunization of mice with rhLIF resulted in reduction of the implantation and fertility rate up to 80.49% and 75%, respectively. All mice produced a high titer of anti-rhLIF antibodies in serums and vaginal fluids washes after 16 weeks; however, these antibodies were cleared from vaginal fluid washes after six months. A significant down-regulation in mRNA levels of IRG-1, Ar and HB-EGF was observed in vaccinated group compared to controls; however, no significant change in the expression profile of cochlin gene was detected. The results showed that rhLIF prevented pregnancy in a high percentage of female mice. Although the immunization of female Balb/c mice with rhLIF inhibited fertility and expression of genes associated with this molecule, further studies are needed to support this protein as a suitable candidate for contraceptive vaccine in mammals.


Subject(s)
Contraception, Immunologic/methods , Fertility/immunology , Leukemia Inhibitory Factor/administration & dosage , Vaccines, Contraceptive/administration & dosage , Amphiregulin/genetics , Animals , Down-Regulation/genetics , Down-Regulation/immunology , Extracellular Matrix Proteins/genetics , Female , Fertility/genetics , Heparin-binding EGF-like Growth Factor/genetics , Hydro-Lyases/genetics , Leukemia Inhibitory Factor/immunology , Mice , Models, Animal , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , Vaccines, Contraceptive/immunology , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/immunology
4.
Exp Neurol ; 330: 113324, 2020 08.
Article in English | MEDLINE | ID: mdl-32320698

ABSTRACT

Neonatal hypoxic-ischemic encephalopathy remains the most important neurological problem of the newborn. Delays in diagnosing perinatal brain injuries are common, preventing access to acute therapies. Therefore, there is a critical need for therapeutic strategies that are beneficial when delivered beyond 24 h after birth. Here we show that Leukemia Inhibitory Factor (LIF) functions as an essential injury-induced neurotrophic cytokine in the CNS and that non-invasively administering LIF as late as 3 days after a hypoxic-ischemic insult improves neurological function. Using a mouse model of late preterm brain injury we show that astroglial and microglial/macrophage reactivity to hypoxia-ischemia was diminished at 3 days of recovery, but then exacerbated at 2 weeks of recovery in LIF haplodeficient mice. There also were significantly more CD68+/Iba-1+ cells in the ipsilateral striatum in LIF-Het mice compared to WT mice at 2 weeks of recovery. This desynchronized glial response was accompanied by increased neuronal cell death in the striatum and neocortex (Fluorojade C), hypomyelination (reduced MBP staining and thinner external capsule), increased extent of brain damage (Nissl) and diminished neurological function on sensorimotor tests. To our surprise, injured LIF-Het mice had ~7-fold higher IGF-1 levels than injured WT mice at 3 days after HI injury. Intranasally administered LIF activated the Jak-Stat-3 pathway both within the subventricular zone and the neocortex at 30 min after administration. When delivered with a delay of 3 days after the insult, intranasal LIF reduced the extent of brain injury by ~60%, attenuated astrogliosis and microgliosis in striatum, improved subcortical white matter thickness, increased numbers of Olig2+ cells in corpus callosum and improved performance on sensorimotor tests at 2 weeks of recovery. These studies provide key pre-clinical data recommending LIF administration as a neuroprotectant and regenerative cytokine and they highlight the feasibility of pursuing new therapeutics targeting the tertiary phase of neurodegeneration for hypoxic-ischemic encephalopathies.


Subject(s)
Brain/drug effects , Hypoxia-Ischemia, Brain/pathology , Leukemia Inhibitory Factor/administration & dosage , Nerve Regeneration/drug effects , Neuroprotective Agents/administration & dosage , Administration, Intranasal , Animals , Animals, Newborn , Brain/pathology , Mice
5.
Reprod Fertil Dev ; 31(6): 1157-1165, 2019 May.
Article in English | MEDLINE | ID: mdl-31030728

ABSTRACT

In order to help elucidate the process of epiblast and trophoblast cell differentiation in bovine embryos invitro, we attempted to develop a suitable culture medium to allow extended embryo culture. Day 7 bovine blastocysts developed in conventional medium were cultured further in embryonic stem cell medium with or without leukaemia inhibitory factor (LIF) until Day 23. At Day 14, the expression of octamer-binding transcription factor 3/4 (OCT3/4) and VIMENTIN was significantly higher in embryos cultured with than without LIF, but embryonic disc formation was not observed. Although expression of SRY (sex determining region Y)-box 17 (SOX17) mRNA was significantly lower in Day 14 embryos cultured with and without LIF than in invivo embryos, hypoblast cells formed just inside the trophoblast cells of the invitro-cultured embryos. On Day 23, expression of placental lactogen (PL) and prolactin-related protein 1 (PRP1) was not affected by LIF in invitro-cultured embryos, levels of both genes were significantly lower in the invitro than invivo embryos. Similar to invivo embryos, binucleate cell clusters seen in Day 23invitro-cultured embryos were composed of PL-negative and -positive cells. These results suggest that our culture system partially reproduced the differentiation process of trophoblast cells invivo.


Subject(s)
Cell Differentiation/physiology , Embryonic Development/physiology , Leukemia Inhibitory Factor/administration & dosage , Animals , Cattle , Cell Differentiation/drug effects , Culture Media , Embryo Culture Techniques , Embryo, Mammalian , Embryonic Development/drug effects , Embryonic Stem Cells , Octamer Transcription Factor-3/metabolism , Trophoblasts/metabolism , Vimentin/metabolism
6.
Mol Med Rep ; 19(3): 2377-2385, 2019 03.
Article in English | MEDLINE | ID: mdl-30664218

ABSTRACT

Leukemia inhibitory factor (LIF) is a multifunctional cytokine. The present study aimed to determine the expression and effects of LIF on nucleus pulposus generation. Degenerated nucleus pulposus samples were obtained from animal models and patients with lumbar intervertebral disc herniation. Degradation scores of intervertebral discs were evaluated via magnetic resonance imaging (MRI) and histology, and the protein expression levels of LIF were detected. Furthermore, cultured primary human degenerated nucleus pulposus cells (DNPCs) were stimulated with various concentrations of recombinant human LIF protein (rhLIF), and aggrecan and collagen type II α1 (COL2α1) protein expression levels were detected by western blotting. In addition, aggrecan expression was determined by toluidine blue staining. The effects of rhLIF on proliferation and apoptosis of DNPCs were evaluated by Cell Counting Kit­8 and flow cytometry, respectively. The results revealed that the degradation scores of intervertebral discs were significantly associated with modeling time, as determined by MRI and histology. In addition, the protein expression levels of LIF were initially increased in patients with lumbar disc herniation and in rabbit models, particularly in the 2­week modeling group; however, its expression decreased with the progression of disc degeneration. Notably, LIF expression in each modeling group was higher than that in the control and 0 week modeling group. The in vitro study revealed that the protein expression levels of aggrecan and COL2α1 were significantly increased in response to rhLIF, in a dose­dependent manner, and statistical differences were identified between the treatment groups and control group. The results of toluidine blue staining were consistent with this finding. Although rhLIF had no effect on proliferation, it inhibited apoptosis of DNPCs in a concentration­dependent manner. In conclusion, LIF was upregulated during the process of intervertebral disc degeneration, and may promote the expression of extracellular matrix components. It may also be hypothesized that LIF acts as a potential protective factor by inhibiting apoptosis of DNPCs without affecting cell proliferation.


Subject(s)
Intervertebral Disc Degeneration/drug therapy , Leukemia Inhibitory Factor/genetics , Nucleus Pulposus/drug effects , Recombinant Proteins/administration & dosage , Aggrecans/genetics , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Collagen Type II/genetics , Female , Flow Cytometry , Gene Expression Regulation/drug effects , Humans , Intervertebral Disc/diagnostic imaging , Intervertebral Disc/metabolism , Intervertebral Disc/physiopathology , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/physiopathology , Intervertebral Disc Displacement/drug therapy , Intervertebral Disc Displacement/genetics , Intervertebral Disc Displacement/physiopathology , Leukemia Inhibitory Factor/administration & dosage , Magnetic Resonance Imaging , Male , Nucleus Pulposus/diagnostic imaging , Nucleus Pulposus/growth & development , Nucleus Pulposus/pathology , Rabbits , Recombinant Proteins/genetics
7.
Mol Ther ; 26(10): 2407-2417, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30078764

ABSTRACT

Retinal degenerations are a large cluster of diseases characterized by the irreversible loss of light-sensitive photoreceptors that impairs the vision of 9.1 million people in the US. An attractive treatment option is to use gene therapy to deliver broad-spectrum neuroprotective factors. However, this approach has had limited clinical translation because of the inability to control transgene expression. To address this problem, we generated an adeno-associated virus vector named RPF2 that was engineered to express domains of leukemia inhibitory factor fused to the destabilization domain of bacterial dihydrofolate reductase. Fusion proteins containing the destabilization domain are degraded in mammalian cells but can be stabilized with the binding of the drug trimethoprim. Our data show that expression levels of RPF2 are tightly regulated by the dose of trimethoprim and can be reversed by trimethoprim withdrawal. We further show that stabilized RPF2 can protect photoreceptors and prevent blindness in treated mice.


Subject(s)
Genetic Therapy , Leukemia Inhibitory Factor/genetics , Retinal Degeneration/therapy , Animals , Dependovirus/genetics , Gene Expression Regulation/drug effects , Humans , Leukemia Inhibitory Factor/administration & dosage , Mice , Neuroprotection/genetics , Photoreceptor Cells/drug effects , Photoreceptor Cells/pathology , Retina/drug effects , Retina/pathology , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Tetrahydrofolate Dehydrogenase/genetics , Transgenes/drug effects , Trimethoprim/administration & dosage
8.
Chin Med J (Engl) ; 131(1): 75-81, 2018 Jan 05.
Article in English | MEDLINE | ID: mdl-29271384

ABSTRACT

BACKGROUND: Leukemia inhibitory factor (LIF) has been reported to possess various pharmacological effects, including displaying vascular and neuroprotective properties, during retinal disease. The aim of this study was to investigate the vascular and structural changes in the retina of diabetic mice and to explore whether LIF prevents experimental diabetes-induced retinal injury in the early stages. METHODS: Diabetes was induced in C57Bl/6J mice with streptozotocin (STZ) injections. Successful diabetic animal models were randomly separated into two groups: the diabetic group (n = 15) and the LIF-treated group (n = 15). Normal C57BL/6 mice served as the normal control group (n = 14). Recombinant human LIF was intravitreally injected 8 weeks after the diabetic model was successfully established. Retinas were collected and evaluated using histological and immunohistochemical techniques, and flat-mounted retinas and Western blotting were performed at 18 weeks after the induction of diabetes and 2 days after the intravitreal injection of LIF. The analysis of variance test were used. RESULTS: Histological analysis showed that there were fewer retinal ganglion cells (RGCs) and the inner nuclear layer (INL) became thinner in the diabetic model group (RGC 21.8 ± 4.0 and INL 120.2 ± 4.6 µm) compared with the normal control group (RGC 29.0 ± 6.7, t = -3.02, P = 0.007; INL 150.7 ± 10.6 µm, t = -8.88, P < 0.001, respectively). After LIF treatment, the number of RGCs (26.9 ± 5.3) was significantly increased (t = 3.39, P = 0.030) and the INL (134.5 ± 14.2 µm) was thicker compared to the diabetic group (t = 2.75, P = 0.013). In the anti-Brn-3a-labeled retinas, the number of RGCs in the LIF-treated group (3926.0 ± 143.9) was obviously increased compared to the diabetic group (3507.7 ± 286.1, t = 2.38, P = 0.030), while no significance was found between the LIF-treated group and the control group (4188.3 ± 114.7, t = -2.47, P = 0.069). Flat-mounted retinas demonstrated that a disorganized, dense distribution of the vessel was prominent in the diabetic model group. Vessel distribution in the LIF-treated mouse group was typical and the thickness was uniform. The levels of phosphosignal transducer and activator of transcription 3 activation were obviously higher in the LIF-injected retinas than those in the diabetic control group (t = 3.85, P = 0.019) and the normal control (t = -3.20, P = 0.019). CONCLUSION: The present study provides evidence that LIF treatment protects the integrity of the vasculature and prevents retinal injury in the early stages of diabetic retinopathy in STZ-induced diabetic models.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetic Retinopathy/prevention & control , Leukemia Inhibitory Factor/therapeutic use , Retinal Vessels/drug effects , Streptozocin , Animals , Blood Glucose , Cell Count , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/chemically induced , Diabetic Retinopathy/pathology , Humans , Intravitreal Injections , Leukemia Inhibitory Factor/administration & dosage , Male , Mice , Mice, Inbred C57BL , Recombinant Proteins/therapeutic use , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Retinal Vessels/pathology
9.
Obesity (Silver Spring) ; 24(11): 2361-2367, 2016 11.
Article in English | MEDLINE | ID: mdl-27663886

ABSTRACT

OBJECTIVE: High-fat diet (HFD) is known to induce low-grade hypothalamic inflammation. Whether inflammation occurs in other brain areas remains unknown. This study tested the effect of short-term HFD on cytokine gene expression and identified leukemia inhibitory factor (LIF) as a responsive cytokine in the brain stem. Thus, functional and cellular effects of LIF in the brain stem were investigated. METHODS: Male rats were fed chow or HFD for 3 days, and then gene expression was analyzed in different brain regions for IL-1ß, IL-6, TNF-α, and LIF. The effect of intracerebroventricular injection of LIF on chow intake and body weight was also tested. Patch clamp recording was performed in the nucleus tractus solitarius (NTS). RESULTS: HFD increased pontine TNF-α mRNA while downregulating LIF in all major parts of the brain stem, but not in the hypothalamus or hippocampus. LIF injection into the cerebral aqueduct suppressed food intake without conditioned taste aversion, suggesting that LIF can induce anorexia via lower brain regions without causing malaise. In the NTS, a key brain stem nucleus for food intake regulation, LIF induced acute changes in neuronal excitability. CONCLUSIONS: HFD-induced downregulation of anorexic LIF in the brain stem may provide a permissive condition for HFD overconsumption. This may be at least partially mediated by the NTS.


Subject(s)
Anorexia/physiopathology , Brain Stem/metabolism , Diet, High-Fat/adverse effects , Down-Regulation , Leukemia Inhibitory Factor/physiology , Animals , Body Weight/drug effects , Eating/drug effects , Hypothalamus/metabolism , Inflammation/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Leukemia Inhibitory Factor/administration & dosage , Male , RNA, Messenger/metabolism , Rats , Solitary Nucleus/metabolism , Tumor Necrosis Factor-alpha/metabolism
10.
J Anim Sci ; 94(7): 2789-97, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27482666

ABSTRACT

Leukemia inhibitory factor (LIF) has been suggested to function as a potent inhibitor of feed intake in rodents. In sheep, intravenous injection of lipopolysaccharide (LPS) resulted in an increase in gene expression for LIF in the arcuate nucleus ( < 0.01). In the same experiment, agouti related protein (AgRP) expression was elevated ( < 0.05) but there were no effects on proopiomelanocortin expression. Another group of sheep were provided intracerebroventricular (ICV) injections of LIF at 250, 500, 1,000, and 2,500 ng per sheep. Cumulative feed intake was inhibited by the 1,000- and 2,500-ng doses at 8 and 10 h after ICV injection ( < 0.03). All doses of LIF elevated temperature above 40°C, indicating a fever. When AgRP was intracerebroventricularly injected before LIF, there was no effect of LIF to reduce feed intake, suggesting the LIF inhibition of feed intake is consistent with the concept that the effect is mediated by the melanocortin-4 receptor. In an experiment to determine whether endocrine and metabolic effects of LIF were similar to reported effects of LPS, sheep were intracerebroventricularly injected with 2,500 ng LIF, and blood samples were collected at 10-min intervals for 6 h for assay of LH, samples from the first 3 h were assayed for GH, and samples at 30-min intervals were assayed for glucose and free fatty acids. The effect of treatment and treatment × time interaction was significant, indicating elevated plasma free fatty acids ( < 0.03 and < 0.001, respectively) and glucose ( < 0.01 and < 0.0001, respectively). There was also a treatment × time interaction on circulating concentrations of LH such that LIF caused LH to decrease ( < 0.0001). Additionally, there was a tendency for LIF treatment to increase circulating concentrations of GH (P = 0.0874). The effects of LIF on feed intake and other parameters was similar to the effects of LPS and leads to a hypothesis that LIF expression in response to LPS may be a component of the mechanism for feed intake inhibition and perhaps for changes in selected hormone and metabolites in disease models.


Subject(s)
Eating/drug effects , Leukemia Inhibitory Factor/pharmacology , Lipopolysaccharides/toxicity , Sheep/physiology , Agouti-Related Protein/administration & dosage , Agouti-Related Protein/pharmacology , Animals , Appetite/drug effects , Eating/physiology , Fatty Acids, Nonesterified/blood , Gene Expression Regulation , Leukemia Inhibitory Factor/administration & dosage , Luteinizing Hormone , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Time Factors
11.
Adv Exp Med Biol ; 854: 411-8, 2016.
Article in English | MEDLINE | ID: mdl-26427439

ABSTRACT

Caveolin-1 (Cav-1), the scaffolding protein of caveolae, is expressed in several retinal cell types and is associated with ocular pathologies. Cav-1 modulates neuroinflammatory/neuroprotective responses to central nervous system injury. We have shown that loss of Cav-1 results in a blunted cytokine response in retinas challenged with inflammatory stimuli. As neuroinflammatory and neuroprotective signaling overlap in their cytokine production and downstream signaling pathways, we hypothesized that loss of Cav-1 may also suppress neuroprotective signaling in the retina. To test this, we subjected mice in which Cav-1 was deleted specifically in the retina to a neurodegenerative insult induced by sodium iodate (NaIO3) and measured STAT3 activation, a measure of neuroprotective signaling. Our results show that Cav-1 ablation blunts STAT3 activation induced by NaIO3. STAT3 activation in response to intravitreal administration of the IL-6 family cytokine, leukemia inhibitory factor (LIF), was not affected by Cav-1 deletion indicating a competent gp130 receptor response. Thus, Cav-1 modulates neuroprotective signaling by regulating the endogenous production of neuroprotective factors.


Subject(s)
Caveolin 1/genetics , Neuroprotection/genetics , Retina/metabolism , Signal Transduction/genetics , Animals , Blotting, Western , Caveolin 1/deficiency , Female , Immunohistochemistry , Injections, Intraperitoneal , Iodates/administration & dosage , Iodates/pharmacology , Leukemia Inhibitory Factor/administration & dosage , Leukemia Inhibitory Factor/pharmacology , Male , Mice , Mice, Knockout , Neuroprotection/drug effects , Retina/drug effects , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
12.
PLoS One ; 9(12): e116031, 2014.
Article in English | MEDLINE | ID: mdl-25542011

ABSTRACT

OBJECTIVES: To explore whether LIF could promote the proliferation of neural precursor cells (NPCs) and to analyze the correlation between increased NPCs and FluoroGold (FG) labeled neurons in mice after spinal cord injury (SCI). METHODS: Motor behavior was assessed using Rotarod and Platform Hang tests; neurons in the corticospinal and rubrospinal systems were labeled with FG, NPCs were immustained with nestin-FITC conjugate. The numbers of FG-labeled neurons and NPCs were estimated, and the correlation between FG-labeled neurons and NPCs was assessed. RESULTS: Mice in the SCI group showed negligible recovery of locomotor behavior; in contrast, mice in the LIF group showed a statically significant improvement. Both FG-labeled neurons and NPCs were significantly increased in the LIF group compared to the SCI group, and this increase in FG-labeled neurons and NPCs showed a clear association above the lesion level. CONCLUSIONS: LIF could promote locomotive behaviors in mice post-SCI by encouraging the proliferation of NPCs; LIF may in fact be a potential cytokine for the induction of NPCs post-SCI.


Subject(s)
Leukemia Inhibitory Factor/therapeutic use , Neural Stem Cells/cytology , Neurons/cytology , Spinal Cord Injuries/therapy , Animals , Cell Proliferation , Female , Leukemia Inhibitory Factor/administration & dosage , Locomotion , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/pathology , Neurogenesis , Neurons/pathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
13.
Dis Model Mech ; 7(10): 1193-203, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25085990

ABSTRACT

The endogenous reparative capacity of the adult human brain is low, and chronic neurodegenerative disorders of the central nervous system represent one of the greatest areas of unmet clinical need in the developing world. Novel therapeutic strategies to treat them include: (i) growth factor delivery to boost endogenous repair and (ii) replacement cell therapy, including replacing dopaminergic neurons to treat Parkinson's disease (PD). However, these approaches are restricted not only by rapid degradation of growth factors, but also by the limited availability of cells for transplant and the poor survival of implanted cells that lack the necessary stromal support. We therefore hypothesised that provision of a transient artificial stroma for paracrine delivery of pro-survival factors could overcome both of these issues. Using leukaemia inhibitory factor (LIF) - a proneural, reparative cytokine - formulated as target-specific poly(lactic-co-glycolic acid) (PLGA) nano-particles (LIF-nano-stroma), we discovered that attachment of LIF-nano-stroma to freshly isolated fetal dopaminergic cells improved their survival fourfold: furthermore, in vivo, the number of surviving human fetal dopaminergic cells tended to be higher at 3 months after grafting into the striatum of nude rats, compared with controls treated with empty nanoparticles. In addition, we also analysed the effect of a novel nano-stroma incorporating XAV939 (XAV), a potent inhibitor of the developmentally important Wnt-ß-catenin signalling pathway, to investigate whether it could also promote the survival and differentiation of human fetal dopaminergic precursors; we found that the numbers of both tyrosine-hydroxylase-positive neurons (a marker of dopaminergic neurons) and total neurons were increased. This is the first demonstration that LIF-nano-stroma and XAV-nano-stroma each have pro-survival effects on human dopaminergic neurons, with potential value for target-specific modulation of neurogenic fate in cell-based therapies for PD.


Subject(s)
Drug Carriers , Heterocyclic Compounds, 3-Ring/administration & dosage , Leukemia Inhibitory Factor/administration & dosage , Nanoparticles , Parkinson Disease/therapy , Signal Transduction , Wnt Proteins/metabolism , beta Catenin/metabolism , Dopamine/administration & dosage , Humans , Microscopy, Electron, Scanning
14.
Biol Reprod ; 91(3): 66, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25031358

ABSTRACT

The establishment of a receptive uterus is the prime requirement for embryo implantation. In mice, the E2-induced cytokine leukemia inhibitory factor (LIF) is essential in switching the uterine luminal epithelium (LE) from a nonreceptive to a receptive state. Here we define the LIF-mediated switch using array analysis and informatics to identify LIF-induced changes in gene expression and annotated signaling pathways specific to the LE. We compare gene expression profiles at 0, 1, 3, and 6 h, following LIF treatment. During the first hour, the JAK-STAT signaling pathway is activated and the expression of 54 genes declines, primarily affecting LE cytoskeletal and chromatin organization as well as a transient reduction in the progesterone, TGFbetaR1, and ACVR1 receptors. Simultaneously 256 genes increase expression, of which 42 are transcription factors, including Sox, Kfl, Hes, Hey, and Hox families. Within 3 h, the expression of 3987 genes belonging to more than 25 biological process pathways was altered. We confirmed the mRNA and protein distribution of key genes from 10 pathways, including the Igf-1, Vegf, Toll-like receptors, actin cytoskeleton, ephrin, integrins, TGFbeta, Wnt, and Notch pathways. These data identify novel LIF-activated pathways in the LE and define the molecular basis between the refractory and receptive uterine phases. More broadly, these findings highlight the staggering capacity of a single cytokine to induce a dynamic and complex network of changes in a simple epithelium essential to mammalian reproduction and provide a basis for identifying new routes to regulating female reproduction.


Subject(s)
Embryo Implantation , Endometrium/metabolism , Gene Expression Regulation, Developmental , Leukemia Inhibitory Factor/metabolism , Signal Transduction , Animals , Blotting, Western , Chromatin Assembly and Disassembly , Computational Biology , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Dinoprostone/administration & dosage , Dinoprostone/metabolism , Endometrium/cytology , Endometrium/enzymology , Female , Gene Expression Profiling , Injections, Intraperitoneal , Leukemia Inhibitory Factor/administration & dosage , Leukemia Inhibitory Factor/genetics , Mice, Inbred C3H , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction
15.
Biomed Res Int ; 2013: 140639, 2013.
Article in English | MEDLINE | ID: mdl-24371815

ABSTRACT

The generation and application of porcine induced pluripotent stem cells (iPSCs) may enable the testing for safety and efficacy of therapy in the field of human regenerative medicine. Here, the generation of iPSCs from the Massachusetts General Hospital miniature pig (MGH minipig) established for organ transplantation studies is reported. Fibroblasts were isolated from the skin of the ear of a 10-day-old MGH minipig and transduced with a cocktail of six human factors: POU5F1, NANOG, SOX2, C-MYC, KLF4, and LIN28. Two distinct types of iPSCs were generated that were positive for alkaline phosphatase activity, as well as the classical pluripotency markers: Oct4, Nanog, Sox2, and the surface marker Ssea-1. Only one of two porcine iPSC lines differentiated into three germ layers both in vitro and in vivo. Western blot analysis showed that the porcine iPSCs were dependent on LIF or BMP-4 to sustain self-renewal and pluripotency. In conclusion, the results showed that human pluripotent factors could reprogram porcine ear fibroblasts into the pluripotent state. These cells may provide a useful source of cells that could be used for the treatment of degenerative and genetic diseases and agricultural research and application.


Subject(s)
Bone Morphogenetic Protein 4/administration & dosage , Cell Differentiation/drug effects , Induced Pluripotent Stem Cells/cytology , Leukemia Inhibitory Factor/administration & dosage , Animals , Bone Morphogenetic Protein 4/metabolism , Hospitals, General , Humans , Induced Pluripotent Stem Cells/metabolism , Kruppel-Like Factor 4 , Leukemia Inhibitory Factor/metabolism , Massachusetts , Regenerative Medicine , Swine , Swine, Miniature
16.
Cancer Lett ; 339(1): 144-51, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23856028

ABSTRACT

Medullary thyroid carcinoma (MTC) is a neoplasm of the endocrine system, which originates from parafollicular C-cells of the thyroid gland. For MTC therapy, the Food and Drug Administration recently approved vandetanib and cabozantinib, multi-kinase inhibitors targeting RET and other tyrosine kinase receptors of vascular endothelial growth factor, epidermal growth factor, or hepatocyte growth factor. Nevertheless, not all patients with the progressive MTC respond to these drugs, requiring the development of additional therapeutic modalities that have distinct activity. Previously, we reported that expression of activated Ras or Raf in the human MTC cell lines, TT and MZ-CRC-1, can induce growth arrest and RET downregulation via a leukemia inhibitory factor (LIF)-mediated autocrine/paracrine loop. In this study, we aimed to evaluate bacterially-produced recombinant human LIF for its efficacy to suppress human MTC xenografts in mice. Here, we report that, consistent with its effects in vitro, locally or systemically administered recombinant LIF effectively suppressed growth of TT and MZ-CRC-1 xenografts in mice. Further, as predicted from its effects in TT and MZ-CRC-1 cell cultures in vitro, recombinant LIF activated the JAK/STAT pathway and downregulated RET and E2F1 expression in tumors in mice. These results suggest that LIF is a potent cytostatic agent for MTC cells, which regulates unique mechanisms that are not targeted by currently available therapeutic agents.


Subject(s)
Leukemia Inhibitory Factor/pharmacology , Recombinant Proteins , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Animals , Carcinoma, Neuroendocrine , Cell Line, Tumor , Disease Models, Animal , E2F1 Transcription Factor/metabolism , Female , Humans , Janus Kinases/metabolism , Leukemia Inhibitory Factor/administration & dosage , Mice , Proto-Oncogene Proteins c-ret/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Thyroid Neoplasms/drug therapy , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
17.
Reprod Domest Anim ; 48(4): 569-76, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23320894

ABSTRACT

This study examined the effects of supplementation of ES-like cell culture medium with bone morphogenetic protein (BMP)-4 (0, 10, 20 or 100 ng/ml) or Noggin (250, 500 or 750 ng/ml) or TGF-ß1 (0, 0.1, 1 or 10 ng/ml) or SB431542 (0, 10, 25 or 50 µm), an inhibitor of TGF-ß1 signalling, on survival, colony area and expression level of pluripotency genes in buffalo ES-like cells at passage 40-80, under different culture conditions. BMP-4 supplementation significantly reduced (p < 0.05) colony survival rate, percentage increase in colony area and relative mRNA abundance of OCT4, whereas that of NANOG and SOX-2 was increased significantly (p < 0.05). Noggin supplementation did not affect the colony survival rate and percentage increase in colony area in the presence of FGF-2 and LIF. In the presence of FGF-2 alone, it significantly reduced (p < 0.05) the relative mRNA abundance of OCT4 and SOX-2 and increased (p < 0.05) that of NANOG. Supplementation with TGF-ß1 at 1.0 ng/ml but not at other concentrations increased colony survival rate but had no effect on percentage increase in colony area at any concentration. Supplementation with SB-431542 decreased (p < 0.05) colony survival rate at 50 µm but not at other concentrations. The percentage increase in colony area was lower (p < 0.05) with 10 µm SB-431542 than that in the controls, whereas at higher concentrations of 25 or 50 µm, SB-431542 decreased (p < 0.05) the colony size instead of increasing it. In conclusion, these results suggest that BMP-4 induces differentiation in buffalo ES-like cells, whereas TGF-ß/activin/nodal pathway may not be playing a crucial role in maintaining pluripotency in these cells.


Subject(s)
Buffaloes/embryology , Embryonic Stem Cells/physiology , Transforming Growth Factor beta1/administration & dosage , Animals , Benzamides/administration & dosage , Bone Morphogenetic Proteins/administration & dosage , Carrier Proteins/administration & dosage , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Culture Media , Dioxoles/administration & dosage , Fibroblast Growth Factor 2/administration & dosage , Leukemia Inhibitory Factor/administration & dosage , Transforming Growth Factor beta1/antagonists & inhibitors
18.
Histochem Cell Biol ; 139(1): 13-34, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22926285

ABSTRACT

Skeletal muscle regeneration in pathology and following injury requires the coordinated actions of inflammatory cells and myogenic cells to remove damaged tissue and rebuild syncytial muscle cells, respectively. Following contusion injury to muscle, the cytokine leukemia inhibitor factor (LIF) is up-regulated and knockout of Lif negatively impacts on morphometric parameters of muscle regeneration. Although it was speculated that LIF regulates muscle regeneration through direct effects on myogenic cells, the inflammatory effects of LIF have not been examined in regenerating skeletal muscle. Therefore, the expression and function of LIF was examined using the antagonist MH35-BD during specific inflammatory and myogenic stages of notexin-induced muscle regeneration in mice. LIF protein and mRNA were up-regulated in two distinct phases following intramuscular injection of notexin into tibialis anterior muscles. The first phase of LIF up-regulation coincided with the increased expression of pro-inflammatory cytokines; the second phase coincided with myogenic differentiation and formation of new myotubes. Administration of the LIF receptor antagonist MH35-BD during the second phase of LIF up-regulation had no significant effects on transcript expression of genes required for myogenic differentiation or associated with inflammation; there were no significant differences in morphometric parameters of the regenerating muscle. Conversely, when MH35-BD was administered during the acute inflammatory phase, increased gene transcripts for the pro-inflammatory cytokines Tnf (Tumor necrosis factor), Il1b (Interleukin-1ß) and Il6 (Interleukin-6) alongside an increase in the number of Ly6G positive neutrophils infiltrating the muscle were observed. This was followed by a reduction in Myog (Myogenin) mRNA, which is required for myogenic differentiation, and the subsequent number of myotubes formed was significantly decreased in MH35-BD-treated groups compared to sham. Thus, antagonism of the LIF receptor during the inflammatory phase of skeletal muscle regeneration appeared to induce an inflammatory response that inhibited subsequent myotube formation. We propose that the predominant role of LIF in skeletal muscle regeneration appears to be in regulating the inflammatory response rather than directly effecting myogenic cells.


Subject(s)
Inflammation/metabolism , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Muscle Development , Muscle, Skeletal/metabolism , Regeneration , Signal Transduction , Animals , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Elapid Venoms/pharmacology , Gene Expression Regulation , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Inflammation Mediators/metabolism , Injections, Intraperitoneal , Leukemia Inhibitory Factor/administration & dosage , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , Leukemia Inhibitory Factor Receptor alpha Subunit/antagonists & inhibitors , Macrophages/immunology , Macrophages/metabolism , Male , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Inbred C57BL , Muscle Development/drug effects , Muscle, Skeletal/drug effects , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Mutation , Myogenin/genetics , Myogenin/metabolism , Neutrophils/immunology , Neutrophils/metabolism , RNA, Messenger/metabolism , Recombinant Proteins/administration & dosage , Regeneration/drug effects , Signal Transduction/drug effects , Time Factors
19.
Contraception ; 87(6): 813-23, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23121824

ABSTRACT

BACKGROUND: Leukemia inhibitory factor (LIF) is a cytokine with an essential role in the preparation of the endometrium for implantation. Previous studies demonstrated that PEGLA, a LIF receptor antagonist (LA) conjugated with polyethylene glycol (PEG), effectively prevents implantation in mice, identifying PEGLA as a potential contraceptive for women. STUDY DESIGN: Adult female cynomolgus macaques were used to determine the optimal route of administration to deliver PEGLA to the uterine endometrium. Endometrial explants were used to examine the ability of PEGLA to block LIF action at endometrial cells. RESULTS: Both intramuscular and subcutaneous PEGLA administration resulted in peak serum PEGLA 24 h after administration; serum PEGLA was detectable throughout the 144-h sampling period. In contrast, serum PEGLA was near or below the limit of detection after vaginal administration. After intramuscular administration, PEGLA was localized to both luminal and glandular epithelial cells of the uterine endometrium, and PEGLA was measurable in endometrial lysates. PEGLA administration reduced endometrial signal transducer and activator of transcription protein 3 (STAT3) phosphorylation in vivo and in vitro. PEGLA also blocked LIF's ability to elevate expression of cochlin, insulin-like growth factor-binding protein 3, vascular endothelial growth factor A, and cyclooxygenase-2 (also known as PTGS2) in endometrial explants in vitro. CONCLUSIONS: PEGLA was delivered to the non-human primate uterine endometrium with systemic administration, and PEGLA blocked LIF actions associated with implantation. Blocking LIF receptor activity with the antagonist PEGLA may prevent pregnancy in women and provide a novel alternative to currently-available hormonal and barrier contraceptives.


Subject(s)
Contraceptive Agents, Female/administration & dosage , Endometrium/drug effects , Gene Expression Regulation/drug effects , Leukemia Inhibitory Factor/antagonists & inhibitors , Polyethylene Glycols/administration & dosage , Administration, Intravaginal , Animals , Contraceptive Agents, Female/blood , Contraceptive Agents, Female/pharmacokinetics , Contraceptive Agents, Female/pharmacology , Endometrium/cytology , Endometrium/metabolism , Female , Immunohistochemistry , Injections, Intramuscular , Injections, Subcutaneous , Leukemia Inhibitory Factor/administration & dosage , Leukemia Inhibitory Factor/blood , Leukemia Inhibitory Factor/pharmacokinetics , Leukemia Inhibitory Factor/pharmacology , Macaca fascicularis , Metabolic Clearance Rate , Osmolar Concentration , Phosphorylation/drug effects , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/pharmacology , Protein Processing, Post-Translational/drug effects , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Tissue Culture Techniques , Tissue Distribution
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...