Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Int Immunopharmacol ; 116: 109762, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36702076

ABSTRACT

Autoimmune hepatitis (AIH) is increasingly affecting human health but pharmacotherapies remain to be identified. Growing evidence reveals that ferroptosis, a newly recognized form of programmed cell death, is critical for AIH. However, the exact mechanisms of the ferroptotic cascade remain elusive. Data in this study showed that ferroptosis aggravation was associated with protectively-elevated fibroblast growth factor 4 (FGF4) expression in Concanavalin A (ConA)-induced AIH liver injury, with these effects being effectively reversed by Ferrostatin-1 (Fer-1). Moreover, hepatic Fgf4 depletion was more susceptible to lipid peroxidation and iron accumulation, as well as hepatic lesion and inflammation caused by ConA administration. Conversely, treatment with non-mitogenic recombinant FGF4 (rFGF4) mitigated liver damage and hepatocellular ferroptosis while being accompanied by the upregulation of CDGSH iron-sulfur domain-containing protein 3 (CISD3) in vivo and in vitro. Furthermore, CISD3 overexpression exhibited stronger resistance to ferroptosis while CISD3 knockdown reduced ferroptotic biomarkers cystine/glutamate transporter (xCT) and glutathione peroxidase 4(GPX4) in rFGF4-treated Erastin-induced AML12 cells. In addition, rFGF4 significantly enhanced the levels of heme oxygenase 1 (HO-1) and nuclear factor erythroid 2-related factor 2 (Nrf2) in ConA-induced AIH mice. Overall, this study showed that FGF4 can act as a phylactic role in AIH progression, with rFGF4 treatment inhibiting ferroptosis of hepatocytes by increasing CISD3 levels and activating Nrf2/HO-1 signaling.


Subject(s)
Ferroptosis , Hepatitis, Autoimmune , Mice , Humans , Animals , Iron/metabolism , Hepatitis, Autoimmune/drug therapy , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Fibroblast Growth Factor 4/pharmacology , Hepatocytes/metabolism
2.
Hepatology ; 76(4): 1105-1120, 2022 10.
Article in English | MEDLINE | ID: mdl-35152446

ABSTRACT

BACKGROUND AND AIMS: NAFLD represents an increasing health problem in association with obesity and diabetes with no effective pharmacotherapies. Growing evidence suggests that several FGFs play important roles in diverse aspects of liver pathophysiology. Here, we report a previously unappreciated role of FGF4 in the liver. APPROACH AND RESULTS: Expression of hepatic FGF4 is inversely associated with NAFLD pathological grades in both human patients and mouse models. Loss of hepatic Fgf4 aggravates hepatic steatosis and liver damage resulted from an obesogenic high-fat diet. By contrast, pharmacological administration of recombinant FGF4 mitigates hepatic steatosis, inflammation, liver damage, and fibrogenic markers in mouse livers induced to develop NAFLD and NASH under dietary challenges. Such beneficial effects of FGF4 are mediated predominantly by activating hepatic FGF receptor (FGFR) 4, which activates a downstream Ca2+ -Ca2+ /calmodulin-dependent protein kinase kinase beta-dependent AMP-activated protein kinase (AMPK)-Caspase 6 signal axis, leading to enhanced fatty acid oxidation, reduced hepatocellular apoptosis, and mitigation of liver damage. CONCLUSIONS: Our study identifies FGF4 as a stress-responsive regulator of liver pathophysiology that acts through an FGFR4-AMPK-Caspase 6 signal pathway, shedding light on strategies for treating NAFLD and associated liver pathologies.


Subject(s)
Non-alcoholic Fatty Liver Disease , AMP-Activated Protein Kinases/metabolism , Animals , Caspase 6/metabolism , Caspase 6/pharmacology , Diet, High-Fat/adverse effects , Fatty Acids/metabolism , Fibroblast Growth Factor 4/metabolism , Fibroblast Growth Factor 4/pharmacology , Fibroblast Growth Factor 4/therapeutic use , Humans , Liver/pathology , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/pathology , Receptors, Fibroblast Growth Factor/metabolism , Receptors, Fibroblast Growth Factor/therapeutic use
3.
Nat Commun ; 12(1): 7256, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34907199

ABSTRACT

Several members of the FGF family have been identified as potential regulators of glucose homeostasis. We previously reported that a low threshold of FGF-induced FGF receptor 1c (FGFR1c) dimerization and activity is sufficient to evoke a glucose lowering activity. We therefore reasoned that ligand identity may not matter, and that besides paracrine FGF1 and endocrine FGF21, other cognate paracrine FGFs of FGFR1c might possess such activity. Indeed, via a side-by-side testing of multiple cognate FGFs of FGFR1c in diabetic mice we identified the paracrine FGF4 as a potent anti-hyperglycemic FGF. Importantly, we found that like FGF1, the paracrine FGF4 is also more efficacious than endocrine FGF21 in lowering blood glucose. We show that paracrine FGF4 and FGF1 exert their superior glycemic control by targeting skeletal muscle, which expresses copious FGFR1c but lacks ß-klotho (KLB), an obligatory FGF21 co-receptor. Mechanistically, both FGF4 and FGF1 upregulate GLUT4 cell surface abundance in skeletal muscle in an AMPKα-dependent but insulin-independent manner. Chronic treatment with rFGF4 improves insulin resistance and suppresses adipose macrophage infiltration and inflammation. Notably, unlike FGF1 (a pan-FGFR ligand), FGF4, which has more restricted FGFR1c binding specificity, has no apparent effect on food intake. The potent anti-hyperglycemic and anti-inflammatory properties of FGF4 testify to its promising potential for use in the treatment of T2D and related metabolic disorders.


Subject(s)
Fibroblast Growth Factor 4/pharmacology , Hypoglycemic Agents/pharmacology , Muscle, Skeletal/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Kinase , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Fibroblast Growth Factor 4/administration & dosage , Fibroblast Growth Factor 4/metabolism , Fibroblast Growth Factors/administration & dosage , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/pharmacology , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/metabolism , Inflammation , Insulin Resistance , Ligands , Macrophages/drug effects , Macrophages/metabolism , Mice , Muscle, Skeletal/metabolism , Obesity/drug therapy , Obesity/metabolism , Paracrine Communication , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction/drug effects
4.
Cells ; 10(10)2021 10 14.
Article in English | MEDLINE | ID: mdl-34685725

ABSTRACT

Mature cardiomyocytes (CMs) obtained from human pluripotent stem cells (hPSCs) have been required for more accurate in vitro modeling of adult-onset cardiac disease and drug discovery. Here, we found that FGF4 and ascorbic acid (AA) induce differentiation of BG01 human embryonic stem cell-cardiogenic mesoderm cells (hESC-CMCs) into mature and ventricular CMs. Co-treatment of BG01 hESC-CMCs with FGF4+AA synergistically induced differentiation into mature and ventricular CMs. FGF4+AA-treated BG01 hESC-CMs robustly released acute myocardial infarction (AMI) biomarkers (cTnI, CK-MB, and myoglobin) into culture medium in response to hypoxic injury. Hypoxia-responsive genes and potential cardiac biomarkers proved in the diagnosis and prognosis of coronary artery diseases were induced in FGF4+AA-treated BG01 hESC-CMs in response to hypoxia based on transcriptome analyses. This study demonstrates that it is feasible to model hypoxic stress in vitro using hESC-CMs matured by soluble factors.


Subject(s)
Ascorbic Acid/pharmacology , Cell Differentiation , Fibroblast Growth Factor 4/pharmacology , Human Embryonic Stem Cells/pathology , Models, Biological , Myocytes, Cardiac/pathology , Stress, Physiological , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Hypoxia/drug effects , Cell Line , Culture Media/pharmacology , Gene Expression Regulation/drug effects , Heart Ventricles/pathology , Human Embryonic Stem Cells/drug effects , Humans , Myocardial Infarction/pathology , Myocytes, Cardiac/drug effects , Stress, Physiological/drug effects , Transcriptome/genetics
5.
Cell Rep ; 26(2): 407-414.e5, 2019 01 08.
Article in English | MEDLINE | ID: mdl-30625323

ABSTRACT

Trophoblast stem (TS) cells are increasingly used as a model system for studying placentation and placental disorders. However, practical limitations of genetic manipulation have posed challenges for genetic analysis using TS cells. Here, we report the generation of mouse parthenogenetic haploid TS cells (haTSCs) and show that supplementation with FGF4 and inhibition of Rho-associated protein kinase (ROCK) enable the maintenance of their haploidy and developmental potential. The resulting haTSCs have 20 chromosomes, exhibit typical expression features of TS cells, possess the multipotency to differentiate into specialized trophoblast cell types, and can chimerize E13.5 and term placentas. We also demonstrate the capability of the haTSCs to undergo genetic manipulation and facilitate genome-wide screening in the trophoblast lineage. We expect that haTSCs will offer a powerful tool for studying functional genomics and placental biology.


Subject(s)
Embryonic Stem Cells/cytology , Haploidy , Trophoblasts/cytology , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Female , Fibroblast Growth Factor 4/pharmacology , Karyotype , Mice , Primary Cell Culture/methods , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
6.
Nat Protoc ; 14(1): 28-50, 2019 01.
Article in English | MEDLINE | ID: mdl-30470820

ABSTRACT

The human stomach contains two primary domains: the corpus, which contains the fundic epithelium, and the antrum. Each of these domains has distinct cell types and functions, and therefore each presents with unique disease pathologies. Here, we detail two protocols to differentiate human pluripotent stem cells (hPSCs) into human gastric organoids (hGOs) that recapitulate both domains. Both protocols begin with the differentiation of hPSCs into definitive endoderm (DE) using activin A, followed by the generation of free-floating 3D posterior foregut spheroids using FGF4, Wnt pathway agonist CHIR99021 (CHIR), BMP pathway antagonist Noggin, and retinoic acid. Embedding spheroids in Matrigel and continuing 3D growth in epidermal growth factor (EGF)-containing medium for 4 weeks results in antral hGOs (hAGOs). To obtain fundic hGOs (hFGOs), spheroids are additionally treated with CHIR and FGF10. Induced differentiation of acid-secreting parietal cells in hFGOs requires temporal treatment of BMP4 and the MEK inhibitor PD0325901 for 48 h on protocol day 30. In total, it takes ~34 d to generate hGOs from hPSCs. To date, this is the only approach that generates functional human differentiated gastric cells de novo from hPSCs.


Subject(s)
Cell Culture Techniques , Endoderm/cytology , Epithelial Cells/cytology , Gastric Fundus/cytology , Organoids/cytology , Pluripotent Stem Cells/cytology , Pyloric Antrum/cytology , Activins/pharmacology , Benzamides/pharmacology , Carrier Proteins/pharmacology , Cell Differentiation , Collagen/chemistry , Culture Media/chemistry , Culture Media/pharmacology , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Drug Combinations , Endoderm/drug effects , Endoderm/metabolism , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fibroblast Growth Factor 10/pharmacology , Fibroblast Growth Factor 4/pharmacology , Gastric Fundus/metabolism , Humans , Laminin/chemistry , Organ Specificity , Organoids/drug effects , Organoids/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Proteoglycans/chemistry , Pyloric Antrum/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , Tretinoin/pharmacology , Wnt Signaling Pathway/drug effects
7.
Stem Cells Dev ; 28(4): 290-302, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30526365

ABSTRACT

In mice, trophoblast stem (TS) cells are derived from the polar trophectoderm of blastocysts. TS cells cultured in the presence of fibroblast growth factor 4 (Fgf4) are in an undifferentiated state and express undifferentiated marker genes such as Cdx2. After removing Fgf4 from the culture medium, TS cells drastically reduce the expression of undifferentiated marker genes, stop cell proliferation, and differentiate into all trophoblast cell subtypes. To clarify the roles of the parental genomes in placentation, we previously established TS cells from androgenetic embryos (AGTS cells). AGTS cells are in the undifferentiated state when cultured with Fgf4 and express undifferentiated marker genes. After removing Fgf4, AGTS cells differentiate into trophoblast giant cells (TGCs), but not into spongiotrophoblast cells, and some of the AGTS cells continue to proliferate. In this study, we investigated the differentiation potency of AGTS cells by analyzing the expression of undifferentiated marker genes and all trophoblast cell subtype-specific genes. After removing Fgf4, some undifferentiated marker genes (Cdx2, Eomes and Elf5) continued to be expressed. Interestingly, TGCs differentiated from AGTS cells also expressed Cdx2, but not Prl3d1. Moreover, the expression of Gcm1 and Synb was induced after the differentiation, indicating that AGTS cells preferentially differentiated into labyrinth progenitor cells. Cdx2 knockdown resulted in increased Prl3d1 expression, suggesting that Fgf4-independent Cdx2 expression inhibited the functional TGCs. Moreover, Fgf4-independent Cdx2 expression was activated by Gab1, one of the paternally expressed imprinted genes via the mitogen-activated protein kinase kinase (MEK)-extracellular signal regulated protein kinase (ERK) pathway. These results suggested that the paternal genome activates the MEK-ERK pathway without the Fgf4 signal, accelerates the differentiation into labyrinth progenitor cells and controls the function of TGCs.


Subject(s)
Cell Differentiation , Mouse Embryonic Stem Cells/cytology , Trophoblasts/cytology , Animals , CDX2 Transcription Factor/genetics , CDX2 Transcription Factor/metabolism , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fibroblast Growth Factor 4/pharmacology , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , MAP Kinase Signaling System , Mice , Mouse Embryonic Stem Cells/drug effects , Mouse Embryonic Stem Cells/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
8.
Oncotarget ; 7(45): 74015-74030, 2016 Nov 08.
Article in English | MEDLINE | ID: mdl-27677589

ABSTRACT

Several fibroblast growth factor (FGF) isoforms act to stimulate epithelial-mesenchymal transition (EMT) during cancer progression. FGF4 and FGF7 are two ligands of FGF receptor 2 (FGFR2). Using two lung adenocarcinoma (ADC) cell lines, A549 and H1299, we showed that FGF4, but not FGF7, altered cell morphology, promoted EMT-associated protein expression, and enhanced cell proliferation, migration/invasion and colony initiation. In addition, FGF4 increased store-operated calcium entry (SOCE) and expression of the calcium signal-associated protein Orai1. The SOCE inhibitor 2,5-di-tert-butylhydroquinone (BHQ) or Orai1 knockdown reversed all of the EMT-promoting effects of FGF4. BHQ also inhibited FGF4-induced EMT in a mouse xenograft model. Finally, 60 human lung ADC samples and 21 sets of matched specimens (primary and metastatic foci in lymph nodes from one patient) were used to confirm the clinicopathologic significance of FGF4 and its correlation with E-cadherin, Vimentin and Orai1 expression. Our study thus shows that FGF4 induces EMT by elevating SOCE in lung ADC.


Subject(s)
Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Calcium/metabolism , Epithelial-Mesenchymal Transition , Fibroblast Growth Factor 4/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Adult , Aged , Animals , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Epithelial-Mesenchymal Transition/drug effects , Female , Fibroblast Growth Factor 4/pharmacology , Fibroblast Growth Factor 7/metabolism , Fibroblast Growth Factor 7/pharmacology , Gene Knockdown Techniques , Humans , Lung Neoplasms/genetics , Male , Mice , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Phenotype , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Vimentin/metabolism , Xenograft Model Antitumor Assays
9.
Rom J Morphol Embryol ; 56(3): 1043-50, 2015.
Article in English | MEDLINE | ID: mdl-26662137

ABSTRACT

Wharton's jelly mesenchymal stromal cells (WJ-MSCs) derived from human umbilical cords could be an appropriate candidate for hepatocyte replacement therapy. Improvement of the efficiency of the cell expression of liver specific genes can be considered in finding new transplantation resources. The present study aimed to differentiate WJ-MSCs toward hepatocyte-like cells on collagen film in the presence of hepatogenic factors, including fibroblast growth factor 4 (FGF4), hepatocyte growth factor (HGF), and insulin-like growth factor-1 (IGF-1). MSCs derived from Wharton's jelly explants were characterized by flow cytometry. Then, the cells were cultured in the presence of hepatogenic media with or without FGF4 on 2D collagen films for 21 days. The expression of liver-specific genes was evaluated by real-time polymerase chain reaction (RT-PCR) and immunocytochemistry. The functional assays were performed by Periodic Acid-Schiff (PAS) staining and Indocyanin Green (ICG) uptake. The cultures pre-exposed to FGF4 expressed higher levels of endodermal markers, such as albumin, compared to the control cultures. Also, cytokeratin 18 expression was significantly increased in FGF4-treated cells. However, the expression level of other liver-specific markers was not influenced by exposure to hepatogenic media with or without FGF4. In conclusion, it was demonstrated that FGF4 could induce the differentiation of WJ-MSCs toward endoderm. Despite the morphological changes and increase in PAS reaction, WJ-MSCs could not differentiate into hepatocytes by hepatogenic media consisting of IGF-1.


Subject(s)
Fibroblast Growth Factor 4/pharmacology , Hepatocytes/cytology , Mesenchymal Stem Cells/cytology , Wharton Jelly/cytology , Animals , Antigens, CD/metabolism , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Shape/drug effects , Cells, Cultured , Flow Cytometry , Hepatocytes/drug effects , Humans , Immunohistochemistry , Infant, Newborn , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Rats , Real-Time Polymerase Chain Reaction
10.
Stem Cell Res Ther ; 6: 89, 2015 May 09.
Article in English | MEDLINE | ID: mdl-25956970

ABSTRACT

INTRODUCTION: Advances in tendon engineering with mesenchymal stem cells (MSCs) are hindered by a need for cues to direct tenogenesis, and markers to assess tenogenic state. We examined the effects of factors involved in embryonic tendon development on adult MSCs, and compared MSC responses to that of embryonic tendon progenitor cells (TPCs), a model system of tenogenically differentiating cells. METHODS: Murine MSCs and TPCs subjected to cyclic tensile loading, transforming growth factor-ß2 (TGFß2), and fibroblast growth factor-4 (FGF4) in vitro were assessed for proliferation and mRNA levels of scleraxis, TGFß2, tenomodulin, collagen type I and elastin. RESULTS: Before treatment, scleraxis and elastin levels in MSCs were lower than in TPCs, while other tendon markers expressed at similar levels in MSCs as TPCs. TGFß2 alone and combined with loading were tenogenic based on increased scleraxis levels in both MSCs and TPCs. Loading alone had minimal effect. FGF4 downregulated tendon marker levels in MSCs but not in TPCs. Select tendon markers were not consistently upregulated with scleraxis, demonstrating the importance of characterizing a profile of markers. CONCLUSIONS: Similar responses as TPCs to specific treatments suggest MSCs have tenogenic potential. Potentially shared mechanisms of cell function between MSCs and TPCs should be investigated in longer term studies.


Subject(s)
Embryonic Stem Cells/drug effects , Fibroblast Growth Factor 4/pharmacology , Mesenchymal Stem Cells/drug effects , Tendons/cytology , Tensile Strength , Transforming Growth Factor beta2/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Marrow Cells/cytology , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Down-Regulation/drug effects , Elastin/genetics , Elastin/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblast Growth Factor 4/genetics , Fibroblast Growth Factor 4/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Transgenic , Recombinant Proteins/biosynthesis , Recombinant Proteins/isolation & purification , Recombinant Proteins/pharmacology , Transcriptome/drug effects , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta2/metabolism
11.
Stem Cell Res ; 13(3 Pt A): 478-91, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25239494

ABSTRACT

Dysfunctional stem cell differentiation into placental lineages is associated with gestational diseases. Of the differentiated lineages available to trophoblast stem cells (TSC), elevated O2 and mitochondrial function are necessary to placental lineages at the maternal-placental surface and important in the etiology of preeclampsia. TSC lineage imbalance leads to embryonic failure during uterine implantation. Stress at implantation exacerbates stem cell depletion by decreasing proliferation and increasing differentiation. In an implantation site O2 is normally ~2%. In culture, exposure to 2% O2 and fibroblast growth factor 4 (FGF4) enabled the highest mouse TSC multipotency and proliferation. In contrast, hypoxic stress (0.5% O2) initiated the most TSC differentiation after 24h despite exposure to FGF4. However, hypoxic stress supported differentiation poorly after 4-7 days, despite FGF4 removal. At all tested O2 levels, FGF4 maintained Warburg metabolism; mitochondrial inactivity and aerobic glycolysis. However, hypoxic stress suppressed mitochondrial membrane potential and maintained low mitochondrial cytochrome c oxidase (oxidative phosphorylation/OxPhos), and high pyruvate kinase M2 (glycolysis) despite FGF4 removal. Inhibiting OxPhos inhibited optimum differentiation at 20% O2. Moreover, adding differentiation-inducing hyperosmolar stress failed to induce differentiation during hypoxia. Thus, differentiation depended on OxPhos at 20% O2; hypoxic and hyperosmolar stresses did not induce differentiation at 0.5% O2. Hypoxia-limited differentiation and mitochondrial inhibition and activation suggest that differentiation into two lineages of the labyrinthine placenta requires O2>0.5-2% and mitochondrial function. Stress-activated protein kinase increases an early lineage and suppresses later lineages in proportion to the deviation from optimal O2 for multipotency, thus it is the first enzyme reported to prioritize differentiation.


Subject(s)
Cell Hypoxia , Mitochondria/metabolism , Stem Cells/cytology , Trophoblasts/cytology , Adenosine Triphosphate/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/drug effects , Cell Lineage , Cell Proliferation/drug effects , Cells, Cultured , DNA-Binding Proteins , Electron Transport Complex IV/metabolism , Female , Fibroblast Growth Factor 4/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/metabolism , Neuropeptides/metabolism , Placenta/cytology , Pregnancy , Pregnancy Proteins/metabolism , Pyruvate Kinase/metabolism , Reactive Oxygen Species/metabolism , Stem Cells/metabolism , Transcription Factors
12.
Genet Mol Res ; 13(1): 415-24, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24535868

ABSTRACT

Our research demonstrated the potential for mouse bone marrow mesenchymal stem cells (mBMMSCs) to differentiate into hepatocytes in vitro and in vivo. However, the exact mechanism of this process remains unknown. In this study, we investigated the role of the mitogen-activated protein kinase (MAPK) cell-signaling pathway in the differentiation of mBMMSCs into hepatocytes. mBMMSCs were isolated from femurs and tibias, and hepatic differentiation was induced in Isove's modified Eagle's medium supplemented with 10% fetal bovine serum, containing human growth factor and fibroblast growth factor 4. After seven days of induction, morphological characteristics were examined. For inhibition of signaling molecular activities, the inhibitors p38 (SB203580), ERK1/2 (U0126), and MSK1 (H89) were added to the differentiation medium. Real-time polymerase chain reaction and Western blot analysis were used to evaluate the gene expression profiles and protein expression of several markers, including the early specific markers of hepatocytes (AFP and FOXa2), phosphorylated-p38 (p-p38), phosphorylated-ERK1/2 (p-ERK1/2), and phosphorylated- MSK1 (p-MSK1). Expressions of p-p38, p-ERK1/2, and p-MSK1 were effectively inhibited by their respective inhibitors. Expressions of early specific markers, AFP and FOXa2, in the p38, ERK1/2, and MSK1 inhibitor-treated groups were significantly decreased compared to those of the cytokine-induced control. Notably, the expressions of AFP and FOXa2 in the p38 inhibitor group were more obviously reduced than those in the ERK1/2 inhibitor group. The MAPK signaling pathway, especially p38, is sufficient to drive differentiation of mBMMSCs into hepatocytes. This could increase the efficiency of hepatocyte differentiation, which would benefit clinical applications.


Subject(s)
Cell Differentiation/drug effects , Fibroblast Growth Factor 4/pharmacology , Hepatocytes/cytology , MAP Kinase Signaling System , Mesenchymal Stem Cells/cytology , Animals , Hepatocyte Nuclear Factor 3-beta/genetics , Hepatocyte Nuclear Factor 3-beta/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred ICR , Receptors, Peptide/genetics , Receptors, Peptide/metabolism , alpha-Fetoproteins/genetics , alpha-Fetoproteins/metabolism
13.
Biochem Biophys Res Commun ; 445(1): 16-22, 2014 Feb 28.
Article in English | MEDLINE | ID: mdl-24491556

ABSTRACT

Mesenchymal stem cells (MSCs) are an active topic of research in regenerative medicine due to their ability to secrete a variety of growth factors and cytokines that promote healing of damaged tissues and organs. In addition, these secreted growth factors and cytokines have been shown to exert an autocrine effect by regulating MSC proliferation and differentiation. We found that expression of EGF, FGF-4 and HGF were down-regulated during serial passage of bone marrow-derived mesenchymal stem cells (BMSCs). Proliferation and differentiation potentials of BMSCs treated with these growth factors for 2 months were evaluated and compared to BMSCs treated with FGF-2, which increased proliferation of BMSCs. FGF-2 and -4 increased proliferation potentials at high levels, about 76- and 26-fold, respectively, for 2 months, while EGF and HGF increased proliferation of BMSCs by less than 2.8-fold. Interestingly, differentiation potential, especially adipogenesis, was maintained only by HGF treatment. Treatment with FGF-2 rapidly induced activation of AKT and later induced ERK activation. The basal level of phosphorylated ERK increased during serial passage of BMSCs treated with FGF-2. The expression of LC3-II, an autophagy marker, was gradually increased and the population of senescent cells was increased dramatically at passage 7 in non-treated controls. But FGF-2 and FGF-4 suppressed LC3-II expression and down-regulated senescent cells during long-term (i.e. 2month) cultures. Taken together, depletion of growth factors during serial passage could induce autophagy, senescence and down-regulation of stemness (proliferation via FGF-2/-4 and differentiation via HGF) through suppression of AKT and ERK signaling.


Subject(s)
Bone Marrow Cells/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , Mesenchymal Stem Cells/drug effects , Adult , Blotting, Western , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Epidermal Growth Factor/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 2/pharmacology , Fibroblast Growth Factor 4/pharmacology , Hepatocyte Growth Factor/pharmacology , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Microtubule-Associated Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Young Adult
14.
Nature ; 505(7485): 676-80, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24476891

ABSTRACT

We recently discovered an unexpected phenomenon of somatic cell reprogramming into pluripotent cells by exposure to sublethal stimuli, which we call stimulus-triggered acquisition of pluripotency (STAP). This reprogramming does not require nuclear transfer or genetic manipulation. Here we report that reprogrammed STAP cells, unlike embryonic stem (ES) cells, can contribute to both embryonic and placental tissues, as seen in a blastocyst injection assay. Mouse STAP cells lose the ability to contribute to the placenta as well as trophoblast marker expression on converting into ES-like stem cells by treatment with adrenocorticotropic hormone (ACTH) and leukaemia inhibitory factor (LIF). In contrast, when cultured with Fgf4, STAP cells give rise to proliferative stem cells with enhanced trophoblastic characteristics. Notably, unlike conventional trophoblast stem cells, the Fgf4-induced stem cells from STAP cells contribute to both embryonic and placental tissues in vivo and transform into ES-like cells when cultured with LIF-containing medium. Taken together, the developmental potential of STAP cells, shown by chimaera formation and in vitro cell conversion, indicates that they represent a unique state of pluripotency.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Placenta/cytology , Trophoblasts/cytology , Adrenocorticotropic Hormone/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Lineage/drug effects , Cellular Reprogramming/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Female , Fibroblast Growth Factor 4/pharmacology , Induced Pluripotent Stem Cells/drug effects , Leukemia Inhibitory Factor/pharmacology , Mice , Mice, Inbred ICR , Placenta/drug effects , Pregnancy , Trophoblasts/drug effects
15.
PLoS One ; 8(8): e71641, 2013.
Article in English | MEDLINE | ID: mdl-23967228

ABSTRACT

Fibroblast growth factor-4 (FGF4) is expressed in embryonic stages and in adult tissues, where it plays critical roles in modulating multiple cellular functions. However, the exact roles of FGF4 on proliferation and differentiation of embryonic stem cells (ESCs) are not completely understood. Exogenous addition of FGF4 stimulated proliferation of mouse ESCs (mESCs), as proven by the increases in DNA synthesis and cell cycle regulatory protein induction. These increases were almost completely inhibited by pre-treating cells with anti-FGF4 antibody. FGF4 also activated c-Jun N-terminal kinase (JNK) and extracellular-signal regulated kinase (ERK) signaling, but not p38 kinase. Blockage of JNK signaling by SP600125 or by transfection with its specific siRNA significantly inhibited FGF4-stimulated cell proliferation through the suppression of c-Jun induction and activator protein-1 (AP-1) activity. However, ERK or p38 kinase inhibitor did not affect FGF4-stimulated proliferation in mESCs. FGF4 suppressed osteogenic differentiation of mESCs by inhibiting expression of transcription factors involved in bone formation. Further, exogenous FGF4 addition stimulated proliferation of human periodontal ligament stem cells (hPDLSCs) and bone marrow mesenchymal stem cells (BMMSCs) via activation of ERK signaling. FGF4 also augmented mineralization of hPDLSCs, but not of BMMSCs. Collectively, it is suggested that FGF4 triggers proliferation of stem cells by activating MAPK-mediated signaling, while it affects differently osteogenic differentiation according to the origins of stem cells.


Subject(s)
Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fibroblast Growth Factor 4/pharmacology , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction/drug effects , Animals , Antibodies, Monoclonal/pharmacology , Calcification, Physiologic/drug effects , Cell Cycle Proteins/metabolism , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Embryonic Stem Cells/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factor 4/antagonists & inhibitors , Gene Expression Regulation/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis/drug effects , Phosphorylation , Protein Binding , Sp7 Transcription Factor , Transcription Factor AP-1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
16.
J Biol Chem ; 288(41): 29760-70, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-23950180

ABSTRACT

Microglossia is a congenital birth defect in humans and adversely impacts quality of life. In vertebrates, tongue muscle derives from the cranial mesoderm, whereas tendons and connective tissues in the craniofacial region originate from cranial neural crest (CNC) cells. Loss of transforming growth factor ß (TGFß) type II receptor in CNC cells in mice (Tgfbr2(fl/fl);Wnt1-Cre) causes microglossia due to a failure of cell-cell communication between cranial mesoderm and CNC cells during tongue development. However, it is still unclear how TGFß signaling in CNC cells regulates the fate of mesoderm-derived myoblasts during tongue development. Here we show that activation of the cytoplasmic and nuclear tyrosine kinase 1 (ABL1) cascade in Tgfbr2(fl/fl);Wnt1-Cre mice results in a failure of CNC-derived cell differentiation followed by a disruption of TGFß-mediated induction of growth factors and reduction of myogenic cell proliferation and differentiation activities. Among the affected growth factors, the addition of fibroblast growth factor 4 (FGF4) and neutralizing antibody for follistatin (FST; an antagonist of bone morphogenetic protein (BMP)) could most efficiently restore cell proliferation, differentiation, and organization of muscle cells in the tongue of Tgfbr2(fl/fl);Wnt1-Cre mice. Thus, our data indicate that CNC-derived fibroblasts regulate the fate of mesoderm-derived myoblasts through TGFß-mediated regulation of FGF and BMP signaling during tongue development.


Subject(s)
Muscles/metabolism , Neural Crest/metabolism , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Tongue/metabolism , Animals , Cell Proliferation/drug effects , Cells, Cultured , Female , Fibroblast Growth Factor 4/genetics , Fibroblast Growth Factor 4/metabolism , Fibroblast Growth Factor 4/pharmacology , Follistatin/genetics , Follistatin/metabolism , Follistatin/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Developmental , Humans , Immunoblotting , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Knockout , Muscles/embryology , Neural Crest/cytology , Neural Crest/embryology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/genetics , Skull/embryology , Skull/metabolism , Tongue/cytology , Tongue/embryology , Transforming Growth Factor beta/pharmacology
17.
Biosci Biotechnol Biochem ; 77(1): 173-7, 2013.
Article in English | MEDLINE | ID: mdl-23291761

ABSTRACT

Fibroblast growth factor 4 (FGF4) is considered a crucial gene in the development of mammalian embryos. Here we identified common amino acid sequences predicted from coding exons of the FGF4 gene in five pigs of two breeds, and HispFGF4, a 6× histidine-tagged porcine FGF4, was produced in Escherichia coli. HispFGF4 was purified efficiently from the supernatant of cell lysate by heparin column chromatography. In a porcine embryonic fibroblast cell line, HispFGF4 showed significant mitogenic activities at concentrations as low as 0.001 nM (p<0.01). To the best of our knowledge, this is the first report describing the complete nucleotide sequence of coding exons for the porcine FGF4 protein in two breeds, together with the production of a recombinant, bioactive porcine FGF4 derivative.


Subject(s)
Exons , Fibroblast Growth Factor 4/genetics , Open Reading Frames , Recombinant Fusion Proteins/genetics , Swine/genetics , Animals , Base Sequence , Breeding , Cloning, Molecular , Escherichia coli/genetics , Fibroblast Growth Factor 4/metabolism , Fibroblast Growth Factor 4/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Molecular Sequence Data , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Sequence Alignment , Swine/embryology
18.
Dev Biol ; 373(2): 359-72, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23123966

ABSTRACT

Preimplantation development culminates with the emergence of three distinct populations: the inner cell mass, primitive endoderm and trophectoderm. Here, we define the mechanisms underlying the requirement of Suds3 in pre/peri-implantation development. Suds3 knockdown blastocysts exhibit a failure of both trophectoderm proliferation as well as a conspicuous lack of primitive endoderm. Expression of essential lineage factors Nanog, Sox2, Cdx2, Eomes, Elf5 and Sox17 are severely reduced in the absence of Suds3. Importantly, we document deficient FGF4/ERK signaling and show that exogenous FGF4 rescues primitive endoderm formation and trophectoderm proliferation in Suds3 knockdown blastocysts. We also show that Hdac1 knockdown reduces Sox2/FGF4/ERK signaling in blastocysts. Collectively, these data define a role for Suds3 in activation of FGF4/ERK signaling and determine an essential molecular role of Suds3/Sin3/HDAC complexes in lineage specification in vivo.


Subject(s)
Body Patterning , Cell Lineage , Repressor Proteins/deficiency , Animals , Blastocyst Inner Cell Mass/cytology , Blastocyst Inner Cell Mass/drug effects , Blastocyst Inner Cell Mass/metabolism , Body Patterning/drug effects , Body Patterning/genetics , Cell Lineage/drug effects , Cell Lineage/genetics , Ectoderm/cytology , Ectoderm/drug effects , Ectoderm/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibroblast Growth Factor 4/metabolism , Fibroblast Growth Factor 4/pharmacology , GATA6 Transcription Factor/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Gene Knockdown Techniques , HMGB Proteins/genetics , HMGB Proteins/metabolism , Histone Deacetylase 1/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Nanog Homeobox Protein , Repressor Proteins/genetics , Repressor Proteins/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , SOXF Transcription Factors/genetics , SOXF Transcription Factors/metabolism , Signal Transduction/drug effects
19.
Reproduction ; 144(5): 625-32, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22956519

ABSTRACT

Several fibroblast growth factors (FGFs), including FGF1, FGF4 and FGF10, alter ovarian granulosa cell function. These ligands exhibit different patterns of receptor activation, and their mechanisms of action on granulosa cells remain unknown. The objective of this study was to identify the major pathways and target genes activated by FGF1, FGF4 and FGF10 in primary oestrogenic granulosa cells cultured under serum-free conditions. FGF1 and FGF4 increased levels of mRNA encoding Sprouty family members, SPRY2 and SPRY4, and the orphan nuclear receptors NR4A1 and NR4A3. Both FGF1 and FGF4 decreased levels of mRNA encoding SPRY3 and the pro-apoptotic factor BAX. FGF1 but not FGF4 stimulated expression of the cell cycle regulator, GADD45B. In contrast, FGF10 altered the expression of none of these genes. Western blot demonstrated that FGF4 activated ERK1/2 and Akt signalling rapidly and transiently, whereas FGF10 elicited a modest and delayed activation of ERK1/2. These data show that FGF1 and FGF4 activate typical FGF signalling pathways in granulosa cells, whereas FGF10 activates atypical pathways.


Subject(s)
Cattle , Fibroblast Growth Factors/pharmacology , Gene Expression/drug effects , Granulosa Cells/metabolism , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Female , Fibroblast Growth Factor 1/pharmacology , Fibroblast Growth Factor 10/pharmacology , Fibroblast Growth Factor 4/pharmacology , Granulosa Cells/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Orphan Nuclear Receptors/genetics , RNA, Messenger/analysis , Signal Transduction/drug effects , Signal Transduction/genetics , bcl-2-Associated X Protein/genetics
20.
Virol J ; 9: 6, 2012 Jan 06.
Article in English | MEDLINE | ID: mdl-22225618

ABSTRACT

BACKGROUND: The EB peptide is a 20-mer that was previously shown to have broad spectrum in vitro activity against several unrelated viruses, including highly pathogenic avian influenza, herpes simplex virus type I, and vaccinia, the prototypic orthopoxvirus. To expand on this work, we evaluated EB for in vitro activity against the zoonotic orthopoxviruses cowpox and monkeypox and for in vivo activity in mice against vaccinia and cowpox. FINDINGS: In yield reduction assays, EB had an EC50 of 26.7 µM against cowpox and 4.4 µM against monkeypox. The EC50 for plaque reduction was 26.3 µM against cowpox and 48.6 µM against monkeypox. A scrambled peptide had no inhibitory activity against either virus. EB inhibited cowpox in vitro by disrupting virus entry, as evidenced by a reduction of the release of virus cores into the cytoplasm. Monkeypox was also inhibited in vitro by EB, but at the attachment stage of infection. EB showed protective activity in mice infected intranasally with vaccinia when co-administered with the virus, but had no effect when administered prophylactically one day prior to infection or therapeutically one day post-infection. EB had no in vivo activity against cowpox in mice. CONCLUSIONS: While EB did demonstrate some in vivo efficacy against vaccinia in mice, the limited conditions under which it was effective against vaccinia and lack of activity against cowpox suggest EB may be more useful for studying orthopoxvirus entry and attachment in vitro than as a therapeutic against orthopoxviruses in vivo.


Subject(s)
Antiviral Agents/pharmacology , Cowpox virus/drug effects , Cowpox/drug therapy , Ectromelia virus/drug effects , Fibroblast Growth Factor 4/pharmacology , Oligopeptides/pharmacology , Vaccinia/drug therapy , Animals , Antiviral Agents/administration & dosage , Disease Models, Animal , Fibroblast Growth Factor 4/administration & dosage , Humans , Mice , Microbial Sensitivity Tests , Oligopeptides/administration & dosage , Survival Analysis , Treatment Outcome , Viral Load , Viral Plaque Assay , Virus Attachment/drug effects , Virus Internalization/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...