Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 14981, 2024 07 01.
Article in English | MEDLINE | ID: mdl-38951546

ABSTRACT

Gulf War Illness (GWI) describes a series of symptoms suffered by veterans of the Gulf war, consisting of cognitive, neurological and gastrointestinal dysfunctions. Two chemicals associated with GWI are the insecticide permethrin (PER) and the nerve gas prophylactic pyridostigmine-bromide (PB). In this study we assessed the effects of PER and PB exposure on the pathology and subsequent alcohol (EtOH)-induced liver injury, and the influence of a macrophage depletor, PLX3397, on EtOH-induced liver damage in PER/PB-treated mice. Male C57BL/6 mice were injected daily with vehicle or PER/PB for 10 days, followed by 4 months recovery, then treatment with PLX3397 and a chronic-plus-single-binge EtOH challenge for 10 days. PER/PB exposure resulted in the protracted increase in liver transaminases in the serum and induced chronic low-level microvesicular steatosis and inflammation in GWI vs Naïve mice up to 4 months after cessation of exposure. Furthermore, prior exposure to PER/PB also resulted in exacerbated response to EtOH-induced liver injury, with enhanced steatosis, ductular reaction and fibrosis. The enhanced EtOH-induced liver damage in GWI-mice was attenuated by strategies designed to deplete macrophages in the liver. Taken together, these data suggest that exposure to GWI-related chemicals may alter the liver's response to subsequent ethanol exposure.


Subject(s)
Ethanol , Mice, Inbred C57BL , Persian Gulf Syndrome , Pyridostigmine Bromide , Animals , Persian Gulf Syndrome/chemically induced , Persian Gulf Syndrome/pathology , Male , Pyridostigmine Bromide/pharmacology , Mice , Ethanol/adverse effects , Ethanol/toxicity , Permethrin/toxicity , Liver/drug effects , Liver/pathology , Insecticides/toxicity , Insecticides/adverse effects , Disease Models, Animal
2.
Res Sq ; 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38313276

ABSTRACT

Gulf War Illness (GWI) describes a series of symptoms suffered by veterans of the Gulf war consisting of cognitive, neurological and gastrointestinal dysfunctions. Two chemicals associated with GWI are the insecticide permethrin (PER) and the nerve gas prophylactic pyridostigmine-bromide (PB). In this study we assessed the effects of PER and PB exposure on pathology and subsequent alcohol (EtOH)-induced liver injury, and the influence of a macrophage depletor, PLX3397, on EtOH-induced liver damage in PER/PB- treated mice. Male C57BL/6 mice were injected daily with vehicle or PER/PB for 10 days, followed by 4 months recovery, then treatment with PLX3397 and a chronic-plus-single-binge EtOH challenge for 10 days. PER/PB exposure resulted in the protracted increase in liver transaminases in the serum and induced chronic low-level microvesicular steatosis and inflammation in GWI vs Naïve mice up to 4 months after cessation of exposure. Furthermore, prior exposure to PER/PB also resulted in exacerbated response to EtOH-induced liver injury, with enhanced steatosis, ductular reaction and fibrosis. The enhanced EtOH-induced liver damage in GWI-mice was attenuated by strategies designed to deplete macrophages in the liver. Taken together, these data suggest that exposure to GWI-related chemicals may alter the liver's response to subsequent ethanol exposure.

3.
Am J Pathol ; 192(3): 484-502, 2022 03.
Article in English | MEDLINE | ID: mdl-34896073

ABSTRACT

Leptin is an adipokine with roles in food intake and energy metabolism through its actions on neurons in the hypothalamus. The role of leptin in obesity and cardiovascular disorders is well documented. However, its influence on liver conditions such as cholestasis is poorly understood. The effects of exogenous leptin and leptin-neutralizing antibody on biliary hyperplasia, hepatic fibrosis, and inflammation in the multidrug resistance protein 2 knockout (Mdr2KO) mouse model of cholestasis were assessed by quantifying markers specific for cholangiocytes, activated hepatic stellate cells (HSCs), and cytokines. Serum and hepatic leptin were increased in Mdr2KO mice compared with FVB/NJ (FVBN) controls, and exogenous leptin enhanced biliary hyperplasia and liver fibrosis in Mdr2KO and FVBN mice. Leptin administration increased hepatic expression of C-C motif chemokine ligand 2 and IL-6 in Mdr2KO mice. In contrast, leptin-neutralizing antibody reduced intrahepatic bile duct mass and decreased HSC activation in Mdr2KO mice compared with FVBN controls. Sex-related differences were noted, with female Mdr2KO mice having more leptin than males. In cholangiocytes and LX2 cells in vitro, leptin increased phosphorylated Akt and stimulated cell proliferation. Leptin receptor siRNA and inhibitors of Akt phosphorylation impaired leptin-induced cell proliferation and proinflammatory cytokines. The current data suggest that leptin is abnormally increased in cholestatic mice, and excess leptin increases ductular reaction, hepatic fibrosis, and inflammation via leptin receptor-mediated phosphorylation of Akt in cholangiocytes and HSCs.


Subject(s)
Cholestasis , Receptors, Leptin , Animals , Antibodies, Neutralizing , Cholestasis/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Hepatic Stellate Cells/metabolism , Hyperplasia/pathology , Inflammation/pathology , Leptin/metabolism , Leptin/pharmacology , Liver/metabolism , Liver Cirrhosis/pathology , Male , Mice , Mice, Knockout , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Leptin/metabolism
4.
Front Pharmacol ; 12: 645703, 2021.
Article in English | MEDLINE | ID: mdl-33841164

ABSTRACT

The liver is a major metabolic organ and an immunologically complex organ. It produces and uses many substances such as acute phase proteins, cytokines, chemokines, and complementary components to maintain the balance between immunity and tolerance. Interleukins are important immune control cytokines, that are produced by many body cells. In liver injury, interleukins are produced in large amount by various cell types, and act as pro-inflammatory (e.g. interleukin (IL)-6, IL-13, IL-17, and IL-33) as well as anti-inflammatory (e.g. IL-10) functions in hepatic cells. Recently, interleukins are regarded as interesting therapeutic targets for the treatment of liver fibrosis patients. Hepatic cells such as hepatocytes, hepatic stellate cells, and hepatic macrophages are involved to the initiation, perpetuation, and resolution of fibrosis. The understanding of the role of interleukins in such cells provides opportunity for the development of therapeutic target drugs. This paper aims to understand the functional roles of interleukins in hepatic and immune cells when the liver is damaged, and suggests the possibility of interleukins as a new treatment target in liver fibrosis.

5.
Stem Cell Reports ; 8(5): 1214-1225, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28494937

ABSTRACT

Accumulating evidence shows that extracellular vesicles (EVs) produced by mesenchymal stem/stromal cells (MSCs) exert their therapeutic effects in several disease models. We previously demonstrated that MSCs suppress autoimmunity in models of type 1 diabetes (T1D) and experimental autoimmune uveoretinitis (EAU). Therefore, here, we investigated the therapeutic potential of MSC-derived EVs using our established mouse models for autoimmune diseases affecting the pancreas and the eye: T1D and EAU. The data demonstrate that MSC-derived EVs effectively prevent the onset of disease in both T1D and EAU. In addition, the mixed lymphocyte reaction assay with MSC-derived EVs indicated that EVs inhibit activation of antigen-presenting cells and suppress development of T helper 1 (Th1) and Th17 cells. These results raise the possibility that MSC-derived EVs may be an alternative to cell therapy for autoimmune disease prevention.


Subject(s)
Autoimmunity , Diabetes Mellitus, Type 1/therapy , Extracellular Vesicles/transplantation , Mesenchymal Stem Cells/cytology , Retinitis/therapy , Uveitis/therapy , Animals , Cells, Cultured , Diabetes Mellitus, Type 1/immunology , Extracellular Vesicles/immunology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , Retinitis/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Uveitis/immunology
6.
Proc Natl Acad Sci U S A ; 114(17): E3536-E3545, 2017 04 25.
Article in English | MEDLINE | ID: mdl-28396435

ABSTRACT

Status epilepticus (SE), a medical emergency that is typically terminated through antiepileptic drug treatment, leads to hippocampus dysfunction typified by neurodegeneration, inflammation, altered neurogenesis, as well as cognitive and memory deficits. Here, we examined the effects of intranasal (IN) administration of extracellular vesicles (EVs) secreted from human bone marrow-derived mesenchymal stem cells (MSCs) on SE-induced adverse changes. The EVs used in this study are referred to as A1-exosomes because of their robust antiinflammatory properties. We subjected young mice to pilocarpine-induced SE for 2 h and then administered A1-exosomes or vehicle IN twice over 24 h. The A1-exosomes reached the hippocampus within 6 h of administration, and animals receiving them exhibited diminished loss of glutamatergic and GABAergic neurons and greatly reduced inflammation in the hippocampus. Moreover, the neuroprotective and antiinflammatory effects of A1-exosomes were coupled with long-term preservation of normal hippocampal neurogenesis and cognitive and memory function, in contrast to waned and abnormal neurogenesis, persistent inflammation, and functional deficits in animals receiving vehicle. These results provide evidence that IN administration of A1-exosomes is efficient for minimizing the adverse effects of SE in the hippocampus and preventing SE-induced cognitive and memory impairments.


Subject(s)
Exosomes/transplantation , Memory Disorders/therapy , Mesenchymal Stem Cells/metabolism , Neurogenesis , Status Epilepticus/therapy , Administration, Intranasal , Animals , Cell Line , Exosomes/metabolism , Exosomes/pathology , Humans , Male , Memory Disorders/metabolism , Memory Disorders/pathology , Memory Disorders/physiopathology , Mesenchymal Stem Cells/pathology , Mice , Status Epilepticus/metabolism , Status Epilepticus/pathology , Status Epilepticus/physiopathology
7.
BMB Rep ; 50(2): 58-59, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28115038

ABSTRACT

The beneficial paracrine roles of mesenchymal stem cells (MSCs) in tissue repair have potential in therapeutic strategies against various diseases. However, the key therapeutic factors secreted from MSCs and their exact molecular mechanisms of action remain unclear. In this study, the cell-free secretome of umbilical cord-derived MSCs showed significant anti-fibrotic activity in the mouse models of liver fibrosis. The involved action mechanism was the regulation of hepatic stellate cell activation by direct inhibition of the TGFß/Smad-signaling. Antagonizing the milk fat globule-EGF factor 8 (MFGE8) activity blocked the anti-fibrotic effects of the MSC secretome in vitro and in vivo. Moreover, MFGE8 was secreted by MSCs from the umbilical cord as well as other tissues, including teeth and bone marrow. Administration of recombinant MFGE8 protein alone had a significant anti-fibrotic effect in two different models of liver fibrosis. Additionally, MFGE8 downregulated TGFß type I receptor expression by binding to αvß3 integrin on HSCs. These findings revealed the potential role of MFGE8 in modulating TGFß-signaling. Thus, MFGE8 could serve as a novel therapeutic agent for liver fibrosis. [BMB Reports 2017; 50(2): 58-59].


Subject(s)
Antigens, Surface/isolation & purification , Antigens, Surface/physiology , Liver Cirrhosis/prevention & control , Mesenchymal Stem Cells/metabolism , Milk Proteins/isolation & purification , Milk Proteins/metabolism , Animals , Cells, Cultured , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Mesenchymal Stem Cell Transplantation , Metabolome/physiology , Mice
8.
Gastroenterology ; 152(5): 1174-1186, 2017 04.
Article in English | MEDLINE | ID: mdl-27956229

ABSTRACT

BACKGROUND & AIMS: Mesenchymal stem cells (MSCs) mediate tissue repair and might be used to prevent or reduce liver fibrosis. However, little is known about the anti-fibrotic factors secreted from MSCs or their mechanisms. METHODS: Umbilical cord-derived MSCs (UCMSCs) were differentiated into hepatocyte-like cells (hpUCMSCs), medium was collected, and secretome proteins were identified and quantified using nanochip-liquid chromatography/quadrupole time-of-flight mass spectrometry. Liver fibrosis was induced in mice by intraperitoneal injection of thioacetamide or CCl4; some mice were then given injections of secretomes or proteins. Liver tissues were collected and analyzed by histology or polymerase chain reaction array to analyze changes in gene expression patterns. We analyzed the effects of MSC secretomes and potential anti-fibrotic proteins on transforming growth factor ß 1 (TGFß1)-mediated activation of human hepatic stellate cell (HSC) lines (hTert-HSC and LX2) and human primary HSCs. Liver tissues were collected from 16 patients with liver cirrhosis and 16 individuals without cirrhosis (controls) in Korea and analyzed by immunohistochemistry and immunoblots. RESULTS: In mice with fibrosis, accumulation of extracellular matrix proteins was significantly reduced 3 days after injecting secretomes from UCMSCs, and to a greater extent from hpUCMSCs; numbers of activated HSCs that expressed the myogenic marker α-smooth muscle actin (α-SMA, encoded by ACTA2 [actin, alpha 2, smooth muscle]) were also reduced. Secretomes from UCMSCs, and to a greater extent from hpUCMSCs, reduced liver expression of multiple fibrotic factors, collagens, metalloproteinases, TGFß, and Smad proteins in the TGFß signaling pathways. In HSC cell lines and primary HSCs, TGFß1-stimulated upregulation of α-SMA was significantly inhibited (and SMAD2 phosphorylation reduced) by secretomes from UCMSCs, and to a greater extent from hpUCMSCs. We identified 32 proteins in secretomes of UCMSCs that were more highly concentrated in secretomes from hpUCMSCs and inhibited TGFß-mediated activation of HSCs. One of these, milk fat globule-EGF factor 8 (MFGE8), was a strong inhibitor of activation of human primary HSCs. We found MFGE8 to down-regulate expression of TGFß type I receptor by binding to αvß3 integrin on HSCs and to be secreted by MSCs from umbilical cord, teeth, and bone marrow. In mice, injection of recombinant human MFGE8 had anti-fibrotic effects comparable to those of the hpUCMSC secretome, reducing extracellular matrix deposition and HSC activation. Co-injection of an antibody against MFGE8 reduced the anti-fibrotic effects of the hpUCMSC secretome in mice. Levels of MFGE8 were reduced in cirrhotic liver tissue from patients compared with controls. CONCLUSIONS: MFGE8 is an anti-fibrotic protein in MSC secretomes that strongly inhibits TGFß signaling and reduces extracellular matrix deposition and liver fibrosis in mice.


Subject(s)
Antigens, Surface/metabolism , Liver Cirrhosis/metabolism , Milk Proteins/metabolism , Animals , Carbon Tetrachloride/toxicity , Cell Line , Collagen/metabolism , Extracellular Matrix/metabolism , Hepatic Stellate Cells , Hepatocytes , Humans , Integrin alphaVbeta3/metabolism , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Male , Mesenchymal Stem Cells/metabolism , Metabolome , Metalloproteases/metabolism , Mice , Receptors, Transforming Growth Factor beta/metabolism , Smad2 Protein/metabolism , Thioacetamide/toxicity , Transforming Growth Factor beta1/metabolism
9.
Cell Rep ; 15(4): 814-829, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27149847

ABSTRACT

Recent studies have shown that defined factors could lead to the direct conversion of fibroblasts into induced hepatocyte-like cells (iHeps). However, reported conversion efficiencies are very low, and the underlying mechanism of the direct hepatic reprogramming is largely unknown. Here, we report that direct conversion into iHeps is a stepwise transition involving the erasure of somatic memory, mesenchymal-to-epithelial transition, and induction of hepatic cell fate in a sequential manner. Through screening for additional factors that could potentially enhance the conversion kinetics, we have found that c-Myc and Klf4 (CK) dramatically accelerate conversion kinetics, resulting in remarkably improved iHep generation. Furthermore, we identified small molecules that could lead to the robust generation of iHeps without CK. Finally, we show that Hnf1α supported by small molecules is sufficient to efficiently induce direct hepatic reprogramming. This approach might help to fully elucidate the direct conversion process and also facilitate the translation of iHep into the clinic.

10.
PLoS One ; 11(1): e0147553, 2016.
Article in English | MEDLINE | ID: mdl-26793973

ABSTRACT

TNF-α stimulated gene/protein 6 (TNFAIP6/TSG-6) is a multifunctional protein that has a number of potential therapeutic applications. Experiments and clinical trials with TSG-6, however, have been limited by the technical difficulties of producing the recombinant protein. We prepared stable clones of CHO cells that expressed recombinant human TSG-6 (rhTSG-6) as a secreted glycoprotein. Paradoxically, both cell number and protein production decreased dramatically when the clones were expanded. The decreases occurred because the protein aggregated the synthesizing CHO cells by binding to the brush border of hyaluronan that is found around many cultured cells. In addition, the rhTSG-6 readily self-aggregated. To address these problems, we added to the medium an inhibitor of hyaluronan synthesis and heparin to compete with the binding of TSG-6 to hyaluronan. Also, we optimized the composition of the culture medium, and transferred the CHO cells from a spinner culture system to a bioreactor that controlled pH and thereby decreased pH-dependent binding properties of the protein. With these and other improvements in the culture conditions, we obtained 57.0 mg ± 9.16 S.D. of rhTSG-6 in 5 or 6 liter of medium. The rhTSG-6 accounted for 18.0% ± 3.76 S.D. of the total protein in the medium. We then purified the protein with a Ni-chelate column that bound the His tag engineered into the C-terminus of the protein followed by an anion exchange column. The yield of the purified monomeric rhTSG-6 was 4.1 mg to 5.6 mg per liter of culture medium. After intravenous injection into mice, the protein had a longer plasma half-life than commercially available rhTSG-6 isolated from a mammalian cell lysate, apparently because it was recovered as a secreted glycoprotein. The bioactivity of the rhTSG-6 in suppressing inflammation was demonstrated in a murine model.


Subject(s)
Adult Stem Cells/cytology , Cell Adhesion Molecules/chemistry , Inflammation/prevention & control , Recombinant Proteins/chemistry , Adult Stem Cells/metabolism , Animals , Cell Adhesion Molecules/administration & dosage , Cell Adhesion Molecules/metabolism , Cricetinae , Cricetulus , Half-Life , Humans , Hyaluronic Acid/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Lipopolysaccharides/toxicity , Mice , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism
12.
Proc Natl Acad Sci U S A ; 113(1): 170-5, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26699510

ABSTRACT

Extracellular vesicles (EVs) secreted by cells present an attractive strategy for developing new therapies, but progress in the field is limited by several issues: The quality of the EVs varies with the type and physiological status of the producer cells; protocols used to isolate the EVs are difficult to scale up; and assays for efficacy are difficult to develop. In the present report, we have addressed these issues by using human mesenchymal stem/stromal cells (MSCs) that produce EVs when incubated in a protein-free medium, preselecting the preparations of MSCs with a biomarker for their potency in modulating inflammation, incubating the cells in a chemically defined protein-free medium that provided a stable environment, isolating the EVs with a scalable chromatographic procedure, and developing an in vivo assay for efficacy of the cells in suppressing neuroinflammation after traumatic brain injury (TBI) in mice. In addition, we demonstrate that i.v. infusion of the isolated EVs shortly after induction of TBI rescued pattern separation and spatial learning impairments 1 mo later.


Subject(s)
Brain Injuries/complications , Cognition Disorders/therapy , Encephalitis/therapy , Extracellular Vesicles/chemistry , Mesenchymal Stem Cells/chemistry , Animals , Biomarkers/analysis , Brain Injuries/psychology , Cells, Cultured , Chromatography, Ion Exchange , Cognition Disorders/etiology , Cognition Disorders/psychology , Culture Media, Serum-Free , Encephalitis/etiology , Encephalitis/psychology , Humans , Mesenchymal Stem Cells/ultrastructure , Mice , Spatial Learning , Tetraspanin 28/analysis , Tetraspanin 30/analysis
13.
Exp Mol Med ; 47: e174, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26206421

ABSTRACT

The milk fat globule-EGF-factor 8 protein (MFG-E8) has been identified in various tissues, where it has an important role in intercellular interactions, cellular migration, and neovascularization. Previous studies showed that MFG-E8 is expressed in different cell types under normal and pathophysiological conditions, but its expression in hematopoietic stem cells (HSCs) during hematopoiesis has not been reported. In the present study, we investigated MFG-E8 expression in multiple hematopoietic tissues at different stages of mouse embryogenesis. Using immunohistochemistry, we showed that MFG-E8 was specifically expressed in CD34(+) HSCs at all hematopoietic sites, including the yolk sac, aorta-gonad-mesonephros region, placenta and fetal liver, during embryogenesis. Fluorescence-activated cell sorting and polymerase chain reaction analyses demonstrated that CD34(+) cells, purified from the fetal liver, expressed additional HSC markers, c-Kit and Sca-1, and that these CD34(+) cells, but not CD34(-) cells, highly expressed MFG-E8. We also found that MFG-E8 was not expressed in HSCs in adult mouse bone marrow, and that its expression was confined to F4/80(+) macrophages. Together, this study demonstrates, for the first time, that MFG-8 is expressed in fetal HSC populations, and that MFG-E8 may have a role in embryonic hematopoiesis.


Subject(s)
Antigens, Surface/analysis , Hematopoietic Stem Cells/cytology , Mice/embryology , Milk Proteins/analysis , Animals , Antigens, CD34/analysis , Bone Marrow/ultrastructure , Female , Liver/embryology , Placentation , Pregnancy
14.
Toxicol Sci ; 147(1): 190-206, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26089346

ABSTRACT

Highly homogeneous and functional stem cell-derived hepatocyte-like cells (HLCs) are considered a promising option in the cell-based therapy of liver disease and the development of effective in vitro toxicity screening tools. However, the purity of cells and expression and/or activity of drug metabolizing enzymes in stem cell-derived HLCs are usually too low to be useful for clinical or in vitro applications. Here, we describe a highly optimized hepatic differentiation protocol, which produces >90% (BGO1 and CHA15) albumin-positive HLCs with no purification process from human embryonic stem cell lines. In addition, we show that hepatic enzyme gene expressions and activities were significantly improved by generating 3D spheroidal aggregate of HLCs, compared with 2D HLCs. The 3D differentiation method increased expression of nuclear receptors (NRs) that regulate the proper expression of key hepatic enzymes. Furthermore, significantly increased hepatic functions such as albumin and urea secretion were observed in 3D hepatic spheroids, compared with 2D HLCs. HLCs in the spheroid exhibited morphological and ultrastructural features of normal hepatocytes. Importantly, we show that repeated exposures to xenobiotics facilitated further functional maturation of HLC, as confirmed by increased expression of genes for drug metabolizing enzymes and transcription factors. In conclusion, the 3D culture system with repeated exposures to xenobiotics may be a new strategy for enhancing hepatic metabolizing ability of stem cell-derived HLCs as a cell source for in vitro high-throughput hepatotoxicity models.


Subject(s)
Hepatocytes/drug effects , Hepatocytes/metabolism , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Xenobiotics/toxicity , Apoptosis/drug effects , Cell Adhesion , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Cytochromes/biosynthesis , Cytochromes/genetics , Gene Expression , Hepatocytes/enzymology , Human Embryonic Stem Cells/enzymology , Humans , Liver Function Tests , Mixed Function Oxygenases/biosynthesis , Mixed Function Oxygenases/genetics , Transcription Factors/biosynthesis , Transcription Factors/genetics
15.
Cell Transplant ; 24(10): 2155-68, 2015.
Article in English | MEDLINE | ID: mdl-25397866

ABSTRACT

We previously reported the in vitro differentiation of human embryonic stem cells (hESCs) into pancreatic endoderm. Here we demonstrate that islet-like three-dimensional (3D) aggregates can be derived from the pancreatic endoderm by optimizing our previous protocol. Sequential treatment with Wnt3a, activin A, and noggin induced a transient upregulation of T and MixL1, followed by increased expression of endodermal genes, including FOXA2, SOX17, and CXCR4. Subsequent treatment with retinoic acid highly upregulated PDX1 expression. We also show that inhibition of sonic hedgehog signaling by bFGF/activin ßB and cotreatment with VEGF and FGF7 produced many 3D cellular clusters that express both SOX17 and PDX1. We found for the first time that proteoglycans and vimentin(+) mesenchymal cells were mainly localized in hESC-derived PDX1(+) clusters. Importantly, treatment with chlorate, an inhibitor of proteoglycan sulfation, together with inhibition of Notch signaling significantly increased the expression of Neurog3 and NeuroD1, promoting a transition from PDX1(+) progenitor cells toward mature pancreatic endocrine cells. Purified dithizone(+) 3D aggregates generated by our refined protocol produced pancreatic hormones and released insulin in response to both glucose and pharmacological drugs in vitro. Furthermore, the islet-like 3D aggregates decreased blood glucose levels and continued to exhibit pancreatic features after transplantation into diabetic mice. Generation of islet-like 3D cell aggregates from human pluripotent stem cells may overcome the shortage of cadaveric donor islets for future cases of clinical islet transplantation.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Endoderm/cytology , Human Embryonic Stem Cells/cytology , Hyperglycemia/therapy , Islets of Langerhans/cytology , Animals , Cell Culture Techniques , Cell Differentiation/immunology , Cell Differentiation/physiology , Cell Line , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/transplantation , Humans , Hyperglycemia/immunology , Mice , Streptozocin
16.
Tissue Cell ; 46(2): 127-35, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24472423

ABSTRACT

Mesenchymal stem cells (MSCs) are mesoderm-derived cells that are considered a good source of somatic cells for treatment of many degenerative diseases. Previous studies have reported the differentiation of mesodermal MSCs into endodermal and ectodermal cell types beyond their embryonic lineages, including hepatocytes and neurons. However, the molecular pathways responsible for the direct or indirect cell type conversion and the functional ability of the differentiated cells remain unclear and need further research. In the present study, we demonstrated that valproic acid (VPA), which is a histone deacetylase inhibitor, induced an increase in the expression of endodermal genes including CXCR4, SOX17, FOXA1, FOXA2, GSC, c-MET, EOMES, and HNF-1ß in human umbilical cord derived MSCs (hUCMSCs). In addition, we found that VPA is able to increase these endodermal genes in hUCMSCs by activating signal transduction of AKT and ERK. VPA pretreatment increased hepatic differentiation at the expense of adipogenic differentiation. The effects of VPA on modulating hUCMSCs fate were diminished by blocking AKT and ERK activation using specific signaling inhibitors. Together, our results suggest that VPA contributes to the lineage conversion of hUCMSCs to hepatic cell fate by upregulating the expression of endodermal genes through AKT and ERK activation.


Subject(s)
Cell Differentiation/drug effects , Enzyme Inhibitors/pharmacology , Hepatocytes/metabolism , Mesenchymal Stem Cells/metabolism , Umbilical Cord/metabolism , Valproic Acid/pharmacology , Antigens, Differentiation/biosynthesis , Cells, Cultured , Gene Expression Regulation/drug effects , Hepatocytes/cytology , Humans , Mesenchymal Stem Cells/cytology , Umbilical Cord/cytology
17.
Eur J Cancer ; 50(2): 341-50, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24161763

ABSTRACT

AIM: Interleukin-8 (IL-8) has been suggested as a prognostic biomarker for human hepatocellular carcinoma (HCC), but its roles in HCC progression and drug resistance have not been studied. This study investigates the role and underlying mechanism of IL-8 in the chemoresistance and progressive growth of HCC. METHODS: The change of chemosensitivity and proportion of side population in hepatoma cells was examined by cell growth and flow cytometric analyses after anti-cancer treatments or knockdown of IL-8. Expression of IL-8 and ATP-binding cassette (ABC) transporters in hepatoma cells, xenograft and clinical HCC tissues was determined by Western blot and immunohistochemical analyses. Tumourigenicity of hepatoma cells was evaluated in vivo after silencing IL-8 gene. RESULTS: Treatment of hepatoma cells with anti-cancer drugs increased the production of IL-8 and its receptor, as well as the proportion of side population (SP). Exogenous IL-8 increased the SP fraction and expression of multidrug resistance-1, decreasing the drug sensitivity. Silencing of IL-8 gene decreased the ratio of SP cells and drug resistance properties. Both IL-8 and ABC transporters were highly expressed in xenograft and clinical HCC tissues, and knockdown of IL-8 significantly reduced tumour size in vivo. CONCLUSION: Anti-cancer drug-induced IL-8 secretion increased the expression of ABC transporters and SP cells, promoting the growth of HCC in vitro. Thus IL-8 may be a potential therapeutic target in the treatment of HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Interleukin-8/metabolism , Liver Neoplasms/drug therapy , Xenograft Model Antitumor Assays , ATP-Binding Cassette Transporters/metabolism , Animals , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Flow Cytometry , Hep G2 Cells , Humans , Immunohistochemistry , Interleukin-8/genetics , Interleukin-8/pharmacology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Paclitaxel/pharmacology , RNA Interference , Receptors, Interleukin-8/metabolism , Side-Population Cells/drug effects
18.
Stem Cells Dev ; 22(12): 1818-29, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23373441

ABSTRACT

Transplantation and drug discovery programs for liver diseases are hampered by the shortage of donor tissue. While recent studies have shown that hepatic cells can be derived from human embryonic stem cells (hESCs), few cases have shown selective enrichment of hESC-derived hepatocytes and their integration into host liver tissues. Here we demonstrate that the dissociation and reaggregation procedure after an endodermal differentiation of hESC produces spheroids mainly consisted of cells showing hepatic phenotypes in vitro and in vivo. A combined treatment with Wnt3a and bone morphogenic protein 4 efficiently differentiated hESCs into definitive endoderm in an adherent culture. Dissociation followed by reaggregation of these cells in a nonadherent condition lead to the isolation of spheroid-forming cells that preferentially expressed early hepatic markers from the adherent cell population. Further differentiation of these spheroid cells in the presence of the hepatocyte growth factor, oncostatin M, and dexamethasone produced a highly enriched population of cells exhibiting characteristics of early hepatocytes, including glycogen storage, indocyanine green uptake, and synthesis of urea and albumin. Furthermore, we show that grafted spheroid cells express hepatic features and attenuate the serum aspartate aminotransferase level in a model of acute liver injury. These data suggest that hepatic progenitor cells can be enriched by the spheroid formation of differentiating hESCs and that these cells have engraftment potential to replace damaged liver tissues.


Subject(s)
Chemical and Drug Induced Liver Injury/therapy , Embryonic Stem Cells/cytology , Endoderm/transplantation , Hepatocytes/transplantation , Spheroids, Cellular/transplantation , Albumins/biosynthesis , Animals , Biomarkers , Bone Morphogenetic Protein 4/pharmacology , Carbon Tetrachloride , Cell Culture Techniques , Cell Differentiation , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Dexamethasone/pharmacology , Embryonic Stem Cells/metabolism , Endoderm/cytology , Endoderm/metabolism , Glycogen/biosynthesis , Graft Survival , Hepatocyte Growth Factor/pharmacology , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Mice , Mice, Nude , Oncostatin M/pharmacology , Spheroids, Cellular/cytology , Spheroids, Cellular/metabolism , Transplantation, Heterologous , Urea/metabolism , Wnt3A Protein/pharmacology
19.
Toxicol Mech Methods ; 22(2): 118-30, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22082211

ABSTRACT

We developed and analyzed a new surrogate endpoint of the mouse embryonic stem cell test (EST) for developmental neurotoxicity. To determine the sensitivity, specificity, and transferability of the new endpoint, a pre-validation team from three independent laboratories optimized and standardized the protocol for neuronal differentiation of mouse embryonic stem cells (mESCs) by measuring the neuronal differentiation rates of mESCs under different culture conditions, such as the presence or absence of basic fibroblast growth factor (bFGF) in the growth media and varying lengths of culture. In addition, a component ratio of neuronal cells was measured by using flow cytometry analysis of ß-III tubulin (Tuj1)-positive cells and real-time polymerase chain reaction analysis of microtubule-associated protein 2 (MAP2) mRNA. Our results showed that the best growth was achieved by culturing mESCs for 12 d in N2B27 medium without bFGF or ascorbic acid. Lead (II) acetate and aroclor 1254 were used to test the usefulness of the new endpoint. When we used the known ID(50) values for lead (II) acetate in the EST model, it was classified as non-embryotoxic; however, when we used the new ID(50) values that we determined in this study, it was classified as weakly embryotoxic. Aroclor 1254 and penicillin G were also classified as weakly embryotoxic and non-embryotoxic compounds, respectively, when cardiac and neuronal differentiation ID(50) values were used. Therefore, our new surrogate endpoint for developmental neurotoxicity is not only sensitive and specific but also transferable among laboratories.


Subject(s)
Embryonic Stem Cells/drug effects , Neurons/drug effects , Animals , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Lead/toxicity , Mice , NIH 3T3 Cells
20.
FASEB J ; 24(9): 3310-20, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20466877

ABSTRACT

Myoblast differentiation is indispensable for skeletal muscle formation and is governed by the precisely coordinated regulation of a series of transcription factors, including MyoD and myogenin, and transcriptional coregulators. TAZ (transcriptional coactivator with PDZ-binding motif) has been characterized as a modulator of mesenchymal stem cell differentiation into osteoblasts and adipocytes through its regulation of lineage-specific master transcription factors. In this study, we investigated whether TAZ affects myoblast differentiation, which is one of the differentiated lineages of mesenchymal stem cells. Ectopic overexpression of TAZ in myoblasts increases myogenic gene expression in a MyoD-dependent manner and hastens myofiber formation, whereas TAZ knockdown delays myogenic differentiation. In addition, enforced coexpression of TAZ and MyoD in fibroblasts accelerates MyoD-induced myogenic differentiation. TAZ physically interacts with MyoD through the WW domain and activates MyoD-dependent gene transcription. TAZ additionally enhances the interaction of MyoD with the myogenin gene promoter. These results strongly suggest that TAZ functions as a novel transcriptional modulator of myogenic differentiation by promoting MyoD-mediated myogenic gene expression.


Subject(s)
Cell Differentiation/physiology , MyoD Protein/metabolism , Transcription Factors/metabolism , Acyltransferases , Animals , Cell Differentiation/genetics , Cell Line , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique , Immunoblotting , Immunohistochemistry , Immunoprecipitation , Mice , Mice, Inbred C57BL , MyoD Protein/genetics , Myoblasts/cytology , Myoblasts/metabolism , Myogenin/genetics , Myogenin/metabolism , Polymerase Chain Reaction , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...