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










Publication year range
1.
Patient Relat Outcome Meas ; 11: 137-142, 2020.
Article in English | MEDLINE | ID: mdl-32547281

ABSTRACT

INTRODUCTION: Patient satisfaction has become an essential metric in addition to the type of care they receive. Phone calls, emails, and text to patients after their healthcare visit are the typical way of obtaining the data reflecting patient satisfaction. The purpose of this retrospective quality improvement study is to compare the traditional post-outpatient clinic survey method with an onsite concise two-question survey using a tablet method immediately after the patient visit using Net Promoter Score (NPS) questions. METHODS: Data were collected retrospectively from February to August 2018 from an outpatient subspecialty clinic in southern California using an existing database from two different sources: the traditional method (TM) and the tablet-based tool (TBT), using NPS. The TM data were obtained from a third-party company using two questions via phone, email, and text collected 2-4 weeks after the patient's visit. The TBT has only two questions that were given to patients upon their visit check-out. These two questions assessed both provider and clinic's performance using the NPS method. RESULTS: In total, there were 1708 patients seen from February to August 2018. In the TM, the total outgoing messages during this period were 580 (34.0%) with 156 responses (27%). In the TBT, 648 out of 1708 (37.9%) surveys were collected with a 100% response rate. The NPS score showed that 99.2% of the providers were promoters. The NPS score for the clinic was 96% which reflects a promoter score. CONCLUSION: Our results indicate that when using the TBT immediately after their visit to the clinic, a higher response rate was noted. In addition, both methods had similar outcomes in terms of patient satisfaction NPS scores. Future prospective studies with a larger sample size are warranted to evaluate the effectiveness of the TBT tool in assessing patient satisfaction.

2.
Aging Cell ; 17(6): e12836, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30168649

ABSTRACT

Aging is associated with an inevitable and universal loss of cell homeostasis and restricts an organism's lifespan by an increased susceptibility to diseases and tissue degeneration. The glucose uptake associated with producing energy for cell survival is one of the major causes of ROS production under physiological conditions. However, the overall mechanisms by which glucose uptake results in cellular senescence remain mysterious. In this study, we found that TXNIP deficiency accelerated the senescent phenotypes of MEF cells under high glucose condition. TXNIP-/- MEF cells showed greater induced glucose uptake and ROS levels than wild-type cells, and N-acetylcysteine (NAC) treatment rescued the cellular senescence of TXNIP-/- MEF cells. Interestingly, TXNIP-/- MEF cells showed continuous activation of AKT during long-term subculture, and AKT signaling inhibition completely blocked the cellular senescence of TXNIP-/- MEF cells. In addition, we found that TXNIP interacted with AKT via the PH domain of AKT, and their interaction was increased by high glucose or H2 O2 treatment. The inhibition of AKT activity by TXNIP was confirmed using western blotting and an in vitro kinase assay. TXNIP deficiency in type 1 diabetes mice (Akita) efficiently decreased the blood glucose levels and finally increased mouse survival. However, in normal mice, TXNIP deficiency induced metabolic aging of mice and cellular senescence of kidney cells by inducing AKT activity and aging-associated gene expression. Altogether, these results suggest that TXNIP regulates cellular senescence by inhibiting AKT pathways via a direct interaction under conditions of glucose-derived metabolic stress.


Subject(s)
Carrier Proteins/metabolism , Cellular Senescence , Glucose/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Stress, Physiological , Thioredoxins/metabolism , Animals , Cellular Senescence/drug effects , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Energy Metabolism/drug effects , Enzyme Activation/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Kidney/pathology , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protein Binding/drug effects , Stress, Physiological/drug effects
3.
Sci Rep ; 7: 46153, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28383049

ABSTRACT

Suppressor of cytokine signaling (SOCS) proteins are negative regulators of cytokine responses. Although recent reports have shown regulatory roles for SOCS proteins in innate and adaptive immunity, their roles in natural killer (NK) cell development are largely unknown. Here, we show that SOCS2 is involved in NK cell development. SOCS2-/- mice showed a high frequency of NK cells in the bone marrow and spleen. Knockdown of SOCS2 was associated with enhanced differentiation of NK cells in vitro, and the transplantation of hematopoietic stem cells (HSCs) into congenic mice resulted in enhanced differentiation in SOCS2-/- HSCs. We found that SOCS2 could inhibit Janus kinase 2 (JAK2) activity and JAK2-STAT5 signaling pathways via direct interaction with JAK2. Furthermore, SOCS2-/- mice showed a reduction in lung metastases and an increase in survival following melanoma challenge. Overall, our findings suggest that SOCS2 negatively regulates the development of NK cells by inhibiting JAK2 activity via direct interaction.


Subject(s)
Cell Differentiation , Janus Kinase 2/antagonists & inhibitors , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Biomarkers/metabolism , Gene Expression Regulation , Gene Knockdown Techniques , Hematopoietic Stem Cells/metabolism , Interleukin-15/metabolism , Janus Kinase 2/metabolism , Mice, Inbred C57BL , Protein Binding , STAT5 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/deficiency
4.
Cell Signal ; 34: 110-120, 2017 06.
Article in English | MEDLINE | ID: mdl-28323005

ABSTRACT

The nuclear factor kappa B (NF-κB) pathway is pivotal in controlling survival and apoptosis of cancer cells. Macrophage migration inhibitory factor (MIF), a cytokine that regulates the immune response and tumorigenesis under inflammatory conditions, is upregulated in various tumors. However, the intracellular functions of MIF are unclear. In this study, we found that MIF directly interacted with thioredoxin-interacting protein (TXNIP), a tumor suppressor and known inhibitor of NF-κB activity, and MIF significantly induced NF-κB activation. MIF competed with TXNIP for NF-κB activation, and the intracellular MIF induced NF-κB target genes, including c-IAP2, Bcl-xL, ICAM-1, MMP2 and uPA, by inhibiting the interactions between TXNIP and HDACs or p65. Furthermore, we identified the interaction motifs between MIF and TXNIP via site-directed mutagenesis of their cysteine (Cys) residues. Cys57 and Cys81 of MIF and Cys36 and Cys120 of TXNIP were responsible for the interaction. MIF reversed the TXNIP-induced suppression of cell proliferation and migration. Overall, we suggest that MIF induces NF-κB activity by counter acting the inhibitory effect of TXNIP on the NF-κB pathway via direct interaction with TXNIP. These findings reveal a novel intracellular function of MIF in the progression of cancer.


Subject(s)
Carrier Proteins/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , NF-kappa B/metabolism , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Proliferation/drug effects , Gene Expression/drug effects , HEK293 Cells , HeLa Cells , Humans , Intercellular Adhesion Molecule-1/metabolism , Lipopolysaccharides/toxicity , Macrophage Migration-Inhibitory Factors/antagonists & inhibitors , Macrophage Migration-Inhibitory Factors/genetics , Matrix Metalloproteinase 2/metabolism , Mutagenesis, Site-Directed , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology , bcl-X Protein/metabolism
5.
Nat Commun ; 7: 13674, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27929088

ABSTRACT

Ageing is a natural process in living organisms throughout their lifetime, and most elderly people suffer from ageing-associated diseases. One suggested way to tackle such diseases is to rejuvenate stem cells, which also undergo ageing. Here we report that the thioredoxin-interacting protein (TXNIP)-p38 mitogen-activated protein kinase (p38) axis regulates the ageing of haematopoietic stem cells (HSCs), by causing a higher frequency of long-term HSCs, lineage skewing, a decrease in engraftment, an increase in reactive oxygen species and loss of Cdc42 polarity. TXNIP inhibits p38 activity via direct interaction in HSCs. Furthermore, cell-penetrating peptide (CPP)-conjugated peptide derived from the TXNIP-p38 interaction motif inhibits p38 activity via this docking interaction. This peptide dramatically rejuvenates aged HSCs in vitro and in vivo. Our findings suggest that the TXNIP-p38 axis acts as a regulatory mechanism in HSC ageing and indicate the potent therapeutic potential of using CPP-conjugated peptide to rejuvenate aged HSCs.


Subject(s)
Carrier Proteins/physiology , Cellular Senescence , Hematopoietic Stem Cells/physiology , MAP Kinase Signaling System , Thioredoxins/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Female , Male , Mice
6.
PLoS One ; 9(10): e108913, 2014.
Article in English | MEDLINE | ID: mdl-25313504

ABSTRACT

Natural killer (NK) cells are innate immune effector cells that protect against cancer and some viral infections. Until recently, most studies have investigated the molecular signatures of human or mouse NK cells to identify genes that are specifically expressed during NK cell development. However, the mechanism regulating NK cell development remains unclear. Here, we report a regulatory network of potential interactions during in vitro differentiation of human NK cells, identified using genome-wide mRNA and miRNA databases through hierarchical clustering analysis, gene ontology analysis and a miRNA target prediction program. The microRNA (miR)-583, which demonstrated the largest ratio change in mature NK cells, was highly correlated with IL2 receptor gamma (IL2Rγ) expression. The overexpression of miR-583 had an inhibitory effect on NK cell differentiation. In a reporter assay, the suppressive effect of miR-583 was ablated by mutating the putative miR-583 binding site of the IL2Rγ 3' UTR. Therefore, we show that miR-583 acts as a negative regulator of NK cell differentiation by silencing IL2Rγ. Additionally, we provide a comprehensive database of genome-wide mRNA and miRNA expression during human NK cell differentiation, offering a better understanding of basic human NK cell biology for the application of human NK cells in immunotherapy.


Subject(s)
Cell Differentiation , Down-Regulation , Interleukin Receptor Common gamma Subunit/metabolism , Killer Cells, Natural/metabolism , MicroRNAs/metabolism , RNA, Messenger/metabolism , 3' Untranslated Regions , Base Sequence , Binding Sites , Cells, Cultured , Cluster Analysis , Databases, Genetic , Fetal Blood/cytology , Gene Expression Profiling , Gene Regulatory Networks , Humans , Interleukin Receptor Common gamma Subunit/antagonists & inhibitors , Interleukin Receptor Common gamma Subunit/genetics , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , MicroRNAs/genetics , RNA Interference , Sequence Alignment
7.
Cell Immunol ; 280(1): 1-9, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23246829

ABSTRACT

Vitamin-D3 upregulated protein-1 (VDUP1) is a stress response protein. Pseudomonas aeruginosa (P. aeruginosa) infection is a leading cause of death. Mice infected with live P. aeruginosa exhibit significantly decreased VDUP1 expression. However, the function of VDUP1 during P. aeruginosa-induced mouse bacteremic shock is unknown. To address the function of VDUP1 in P. aeruginosa-infected mice, we constructed a bacteremic shock model wherein both wild-type and VDUP1-deficient mice were infected intra-peritoneally with live P. aeruginosa. We found that VDUP1-deficient mice were more resistant to P. aeruginosa-induced bacteremic shock than wild-type mice, as shown by the increased survival, accelerated bacterial clearance and suppression of cytokine overproduction of the VDUP1-deficient mice. VDUP1 promoted the recruitment of neutrophils into the peritoneal cavities of infected mice. VDUP1 impeded the phagocytosis of non-opsonized P. aeruginosa via phosphatidylinositide 3-kinase (PI3K) pathway in macrophages. P. aeruginosa infection induced the generation of reactive oxygen species (ROS), and the increased production of ROS by the peritoneal cells of VDUP1-deficient mice was advantageous in clearing the bacteria. Overall, VDUP1 aggravates bacteremic shock; thus, VDUP1 can be considered a target molecule for the inhibition of P. aeruginosa-induced bacteremic shock.


Subject(s)
Carrier Proteins/physiology , Peritonitis/physiopathology , Pseudomonas Infections/physiopathology , Shock, Septic/physiopathology , Thioredoxins/physiology , Animals , Apoptosis/physiology , Carrier Proteins/genetics , Chemotaxis, Leukocyte/physiology , Colony Count, Microbial , Cytokines/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/pathology , Peritoneal Cavity/microbiology , Peritoneal Cavity/pathology , Peritonitis/microbiology , Phagocytosis/physiology , Phosphatidylinositol 3-Kinases/physiology , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/isolation & purification , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Specific Pathogen-Free Organisms , Spleen/microbiology , Thioredoxins/genetics
8.
Mol Cell Biochem ; 352(1-2): 109-15, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21312053

ABSTRACT

Platelet-activating factor (PAF; 1-O-alkyl-2-acetyl-sn-glycero-3-phosphorylcholine) and PAF-like oxidized phospholipids including 1-palmitoyl-2-oxovaleroyl-sn-glycero-3-phosphorylcholine (POVPC) are generated upon LDL oxidation. The aim of this study was to evaluate the question of whether POVPC can regulate migration of human bone marrow-derived stem cells (hBMSCs) and to characterize signaling mechanisms involved in the POVPC-induced cell migration. POVPC treatment resulted in dose- and time-dependent increase of hBMSCs migration. Treatment of cells with BN52021, a specific antagonist of PAF receptor, completely blocked cell migration induced by not only PAF but also POVPC. Silencing of endogenous PAF receptor expression using PAF receptor-specific small interfering RNA resulted in significant attenuation of cell migration induced by PAF or POVPC. Both PAF and POVPC induced expression of Krüppel-like factor 4 (KLF4) in hBMSCs. POVPC- or PAF-induced cell migration was abrogated by small interfering RNA-mediated depletion of endogenous KLF4. These results suggest that PAF receptor plays a pivotal role in POVPC-induced migration of human BMSCs through PAF receptor-mediated expression of KLF4.


Subject(s)
Bone Marrow Cells/cytology , Cell Movement/physiology , Kruppel-Like Transcription Factors/physiology , Mesenchymal Stem Cells/cytology , Phosphatidylcholines/physiology , Base Sequence , Cells, Cultured , DNA Primers , Humans , Kruppel-Like Factor 4 , Oxidation-Reduction , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction
9.
Cell Physiol Biochem ; 26(4-5): 689-98, 2010.
Article in English | MEDLINE | ID: mdl-21063106

ABSTRACT

BACKGROUND: Oxidized low density lipoprotein (oxLDL) is involved in the development of vascular diseases. Platelet-activating factor (PAF; 1-O-alkyl-2-acetyl-sn-glyero-3-phosphorylcholine) is a key component of oxLDL. METHODS: In the present study, we evaluate whether oxLDL can regulate migration of human bone-marrow derived stem cells (hBMSCs) and characterize the role of PAF in the oxLDL-induced migration. RESULTS AND CONCLUSIONS: oxLDL stimulated chemotaxis of hBMSCs in vitro. Treatment of the cells with BN52021, a specific antagonist of PAF receptor (PAF-R), completely blocked the cell migration induced by PAF, but not platelet-derived growth factor (PDGF-BB). Using PAF-R-specific small interfering RNA, it was demonstrated that silencing of endogenous PAF-R expression significantly attenuated cell migration induced by PAF, but not PDGF-BB, suggesting the specific involvement of PAF-R in the oxLDL-induced cell migration. In addition, PAF-induced migration of hBMSCs was abrogated by pretreating cells with mitogen-activated protein kinase (MAPK) inhibitors, including the MEK inhibitor U0126, the p38 MAPK inhibitor SB202190, and the JNK inhibitor SP600125. Moreover, adenoviral overexpression of a dominant negative mutant of p38 MAPK blocked PAF-stimulated migration. Taken together, these results suggest that PAF plays a pivotal role in the oxLDL-induced recruitment of hBMSCs through mechanisms involving PAF-R-dependent activation of MAPKs.


Subject(s)
Bone Marrow Cells/cytology , Lipoproteins, LDL/metabolism , Mesenchymal Stem Cells/cytology , Platelet Membrane Glycoproteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Anthracenes/pharmacology , Becaplermin , Butadienes/pharmacology , Cell Movement , Humans , Imidazoles/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Nitriles/pharmacology , Platelet Activating Factor/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Platelet Membrane Glycoproteins/genetics , Platelet-Derived Growth Factor/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-sis , Pyridines/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Exp Mol Med ; 42(8): 565-73, 2010 Aug 31.
Article in English | MEDLINE | ID: mdl-20628268

ABSTRACT

Rheumatoid arthritis (RA) is a chronic, inflammatory autoimmune disorder that causes the immune system to attack the joints. Transforming growth factor-beta1 (TGF-beta1) is a secreted protein that promotes differentiation of synovial fibroblasts to alpha-smooth muscle actin (alpha-SMA)-positive myofibroblasts to repair the damaged joints. Synovial fluid from patients with RA (RA-SF) induced expression of alpha-SMA in human adipose tissue-derived mesenchymal stem cells (hASCs). RA-SF-induced alpha-SMA expression was abrogated by immunodepletion of TGF-beta1 from RA-SF with anti-TGF-beta1 antibody. Furthermore, pretreatment of hASCs with the TGF-beta type I receptor inhibitor SB431542 or lentiviral small hairpin RNA-mediated silencing of TGF-beta type I receptor expression in hASCs blocked RA-SF-induced alpha-SMA expression. Small interfering RNA-mediated silencing of Smad2 or adenoviral overexpression of Smad7 (an inhibitory Smad isoform) completely inhibited RA-SF-stimulated alpha-SMA expression. These results suggest that TGF-beta1 plays a pivotal role in RA-SF-induced differentiation of hASCs to alpha-SMA-positive cells.


Subject(s)
Actins/metabolism , Adipose Tissue/cytology , Arthritis, Rheumatoid/metabolism , Mesenchymal Stem Cells/metabolism , Synovial Fluid/metabolism , Transforming Growth Factor beta1/metabolism , Humans , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction , Smad2 Protein/metabolism , Stress Fibers/metabolism
11.
Biochim Biophys Acta ; 1801(1): 23-30, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19733258

ABSTRACT

Migration of mesenchymal stem cells plays a key role in regeneration of injured tissues. Rheumatoid arthritis (RA) is a chronic inflammatory disease and synovial fluid (SF) reportedly contains a variety of chemotactic factors. This study was undertaken to investigate the role of SF in migration of human bone marrow-derived mesenchymal stem cells (hBMSCs) and the molecular mechanism of SF-induced cell migration. SF from RA patients greatly stimulated migration of hBMSCs and the SF-induced migration was completely abrogated by pretreatment of the cells with the lysophosphatidic acid (LPA) receptor antagonist Ki16425 and by small interfering RNA- or lentiviral small hairpin RNA-mediated silencing of endogenous LPA(1)/Edg2. Moreover, SF from RA patients contains higher concentrations of LPA and an LPA-producing enzyme autotoxin than normal SF. In addition, SF from RA patients increased the intracellular concentration of calcium through a Ki16425-sensitive mechanism and pretreatment of the cells with the calmodulin inhibitor W7 or calmodulin-dependent protein kinase II inhibitor KN93 abrogated the SF-induced cell migration. These results suggest that LPA-LPA(1) plays a key role in the migration of hBMSCs induced by SF from RA patients through LPA(1)-dependent activation of calmodulin-dependent protein kinase II.


Subject(s)
Arthritis, Rheumatoid/metabolism , Cell Movement/physiology , Lysophospholipids/metabolism , Mesenchymal Stem Cells/cytology , Synovial Fluid/chemistry , Bone Marrow Cells/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Humans , Mesenchymal Stem Cells/metabolism , Receptors, Lysophosphatidic Acid/metabolism , Synovial Fluid/metabolism
12.
Exp Mol Med ; 41(1): 17-24, 2009 Jan 31.
Article in English | MEDLINE | ID: mdl-19287196

ABSTRACT

Prostanoid metabolites are key mediators in inflammatory responses, and accumulating evidence suggests that mesenchymal stem cells (MSCs) can be recruited to injured or inflamed tissues. In the present study, we investigated whether prostanoid metabolites can regulate migration, proliferation, and differentiation potentials of MSCs. We demonstrated herein that the stable thromboxane A(2) (Tx(2)) mimetic U46619 strongly stimulated migration and proliferation of human adipose tissue-derived MSCs (hADSCs). Furthermore, U46619 treatment increased expression of alpha-smooth muscle actin (alpha-SMA), a smooth muscle marker, in hADSCs, suggesting differentiation of hADSCs into smooth muscle-like cells. U46619 activated ERK and p38 MAPK, and pretreatment of the cells with the MEK inhibitor U0126 or the p38 MAPK inhibitor SB202190 abrogated the U46619-induced migration, proliferation, and alpha-SMA expression. These results suggest that TxA2 plays a key role in the migration, proliferation, and differentiation of hADSCs into smooth muscle-like cells through signaling mechanisms involving ERK and p38 MAPK.


Subject(s)
Adipose Tissue/cytology , Cell Physiological Phenomena , Mesenchymal Stem Cells/cytology , Thromboxane A2/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Cell Physiological Phenomena/drug effects , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Stem Cells ; 26(3): 789-97, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18065393

ABSTRACT

Lysophosphatidic acid (LPA) is enriched in ascites of ovarian cancer patients and is involved in growth and invasion of ovarian cancer cells. Accumulating evidence suggests cancer-associated myofibroblasts play a pivotal role in tumorigenesis through secreting stromal cell-derived factor-1 (SDF-1). In the present study, we demonstrate that LPA induces expression of alpha-smooth muscle actin (alpha-SMA), a marker for myofibroblasts, in human adipose tissue-derived mesenchymal stem cells (hADSCs). The LPA-induced expression of alpha-SMA was completely abrogated by pretreatment of the cells with Ki16425, an antagonist of LPA receptors, or by silencing LPA(1) or LPA(2) isoform expression with small interference RNA (siRNA). LPA elicited phosphorylation of Smad2/3, and siRNA-mediated depletion of endogenous Smad2/3 or adenoviral expression of Smad7, an inhibitory Smad, abrogated the LPA induced expression of alpha-SMA and phosphorylation of Smad2/3. LPA-induced secretion of transforming growth factor (TGF)-beta1 in hADSCs, and pretreatment of the cells with SB431542, a TGF-beta type I receptor kinase inhibitor, or anti-TGF-beta1 neutralizing antibody inhibited the LPA-induced expression of alpha-SMA and phosphorylation of Smad2. Furthermore, ascites from ovarian cancer patients or conditioned medium from ovarian cancer cells induced expression of alpha-SMA and phosphorylation of Smad2, and pretreatment of the cells with Ki16425 or SB431542 abrogated the expression of alpha-SMA and phosphorylation of Smad2. In addition, LPA increased the expression of SDF-1 in hADSCs, and pretreatment of the cells with Ki16425 or SB431562 attenuated the LPA-stimulated expression of SDF-1. These results suggest that cancer-derived LPA stimulates differentiation of hADSCs to myofibroblast-like cells and increases SDF-1 expression through activating autocrine TGF-beta1-Smad signaling pathway.


Subject(s)
Cell Differentiation/drug effects , Fibroblasts/cytology , Fibroblasts/drug effects , Lysophospholipids/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Ovarian Neoplasms/pathology , Actins/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Adult , Aged , Animals , Ascites , Autocrine Communication/drug effects , Chemokine CXCL12/biosynthesis , Chemokine CXCL12/genetics , Female , Humans , Middle Aged , Rats , Receptors, Lysophosphatidic Acid/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Transforming Growth Factor beta1/pharmacology
14.
J Cell Biochem ; 101(5): 1238-51, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17226768

ABSTRACT

Oncostatin M (OSM) is a multifunctional cytokine of the interleukin-6 family and has been implicated in embryonic development, differentiation, inflammation, and regeneration of liver and bone. In the present study, we demonstrated that treatment of human adipose mesenchymal stem cells (hADSCs) with OSM-attenuated adipogenic differentiation, as indicated by decreased accumulation of intracellular lipid droplets and down-regulated expression of adipocytic markers, such as lipoprotein lipase and PPARgamma. However, OSM treatment stimulated osteogenic differentiation, as demonstrated by the increase in matrix mineralization and expression levels of osteogenic differentiation markers, including alkaline phosphatase, Runx2, and osteocalcin. OSM treatment induced activation of JAK2, JAK3, and ERK in hADSCs, and pre-treatment of hADSCs with the JAK2 inhibitor, AG490, significantly restored the OSM-induced inhibition of adipogenic differentiation. Whereas, the JAK3 inhibitor, WHI-P131, and the MEK inhibitor, U0126, had no effects on the anti-adipogenic activity of OSM. On the other hand, the pro-osteogenic activity of OSM was prevented by treatment of the cells with WHI-P131 or U0126, but not with AG490. These results indicate that distinct signaling pathways, including JAK2, JAK3, and MEK-ERK, play specific roles in the OSM-induced anti-adipogenic and pro-osteogenic differentiation of hADSCs.


Subject(s)
Adipogenesis/drug effects , Adipose Tissue/cytology , Adipose Tissue/drug effects , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Oncostatin M/pharmacology , Osteogenesis/drug effects , Adipogenesis/genetics , Adipose Tissue/enzymology , Biomarkers/metabolism , Cell Differentiation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Humans , Interleukin-6/pharmacology , Janus Kinase 2/metabolism , Janus Kinase 3/metabolism , Mesenchymal Stem Cells/enzymology , Mitogen-Activated Protein Kinase Kinases/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Time Factors
15.
Int J Biochem Cell Biol ; 39(2): 439-49, 2007.
Article in English | MEDLINE | ID: mdl-17081797

ABSTRACT

Adiponectin, an adipokine secreted from adipocytes, plays a crucial role in the regulation of glucose and lipid metabolism. In the present study, we examine the role of the IL-6 family of cytokines in the expression of adiponectin in human adipocytes derived from human adipose tissue-derived stromal cells. Oncostatin M (OSM), but not IL-6, attenuated the expression level of adiponectin dose- and time-dependently, and the inhibitory effect of OSM on adiponectin expression was as potent as that of TNF-alpha. The OSM-induced down-regulation of adiponectin expression was correlated with the down-regulation of PPARgamma2 and lipoprotein lipase, markers for adipogenic differentiation, and depletion of intracellular lipid droplets, suggesting dedifferentiation of adipocytes in response to OSM. OSM induced phosphorylation of STAT1, and treatment of adipocytes with JAK3 inhibitor WHI-P131 or MEK inhibitor U0126, but not with JAK2 inhibitor AG490, prevented the activation of STAT1. Furthermore, the OSM-induced suppression of adiponectin expression and dedifferentiation of adipocytes were ameliorated by WHI-P131 or U0126, but not by AG490. These results suggest that OSM inhibits adiponectin expression by inducing dedifferentiation of adipocytes through signaling pathways involving JAK3 and MEK, but not JAK2.


Subject(s)
Adipocytes/cytology , Adiponectin/genetics , Janus Kinase 3/metabolism , MAP Kinase Kinase Kinases/metabolism , Oncostatin M/metabolism , Adipocytes/metabolism , Cell Differentiation , Down-Regulation , Humans , Recombinant Proteins/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
16.
Int J Biochem Cell Biol ; 39(3): 650-9, 2007.
Article in English | MEDLINE | ID: mdl-17169599

ABSTRACT

Stromal-derived factor-1 (SDF-1) is a CXC chemokine that attracts leukocytes and endothelial progenitor cells. In the present study, we demonstrated that oncostatin M (OSM) stimulates expression and secretion of SDF-1 in both human adipose tissue-derived mesenchymal stem cells (hATSCs) and bone marrow-derived mesenchymal stem cells. The OSM-stimulated expression of SDF-1 in hATSCs was completely abrogated by pretreatment of the cells with U0126, an MEK-specific inhibitor, but not with AG490, a JAK2 inhibitor, or WHI-P131, a JAK3 inhibitor, suggesting that ERK, but not JAK2 and JAK3, is involved in the OSM-induced expression of SDF-1. Pretreatment of hATSCs with anti-VEGF neutralizing antibody or VEGF receptor inhibitors, SU5416 and KRN633, had no significant impact on the OSM induction of SDF-1. Furthermore, treatment of hATSCs with recombinant human VEGF165 or adenoviral overexpression of VEGF did not increase the expression of SDF-1. These results suggest that OSM induces secretion of SDF-1 through ERK-, but not VEGF-, dependent signaling pathways in mesenchymal stem cells.


Subject(s)
Chemokines, CXC/biosynthesis , Chemokines, CXC/genetics , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Oncostatin M/pharmacology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Base Sequence , Butadienes/pharmacology , Cells, Cultured , Chemokine CXCL12 , DNA Primers/genetics , Gene Expression/drug effects , Humans , MAP Kinase Signaling System/drug effects , Nitriles/pharmacology , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Vascular Endothelial Growth Factor A/biosynthesis
17.
J Cell Sci ; 119(Pt 23): 4994-5005, 2006 Dec 01.
Article in English | MEDLINE | ID: mdl-17105765

ABSTRACT

Mesenchymal stem cells (MSCs) can differentiate into diverse cell types including adipogenic, osteogenic, chondrogenic and myogenic lineages. In the present study, we demonstrated for the first time that sphingosylphosphorylcholine (SPC) induces differentiation of human adipose-tissue-derived mesenchymal stem cells (hATSCs) to smooth-muscle-like cell types. SPC increased the expression levels of several smooth-muscle-specific genes, such as those for alpha-smooth-muscle actin (alpha-SMA), h1-calponin and SM22alpha, as effectively as transforming growth factor beta (TGF-beta1) and TGF-beta3. SPC elicited delayed phosphorylation of Smad2 after 24 hours exposure, in contrast to rapid phosphorylation of Smad2 induced by TGF-beta treatment for 10 minutes. Pretreatment of the cells with pertussis toxin or U0126, an MEK inhibitor, markedly attenuated the SPC-induced expression of beta-SMA and delayed phosphorylation of Smad2, suggesting that the Gi/o-ERK pathway is involved in the increased expression of alpha-SMA through induction of delayed Smad2 activation. In addition, SPC increased secretion of TGF-beta1 through an ERK-dependent pathway, and the SPC-induced expression of alpha-SMA and delayed phosphorylation of Smad2 were blocked by SB-431542, a TGF-beta type I receptor kinase inhibitor, or anti-TGF-beta1 neutralizing antibody. Silencing of Smad2 expression with small interfering RNA (siRNA) abrogated the SPC-induced expression of alpha-SMA. These results suggest that SPC-stimulated secretion of TGF-beta1 plays a crucial role in SPC-induced smooth muscle cell (SMC) differentiation through a Smad2-dependent pathway. Both SPC and TGF-beta increased the expression levels of serum-response factor (SRF) and myocardin, transcription factors involved in smooth muscle differentiation. siRNA-mediated depletion of SRF or myocardin abolished the alpha-SMA expression induced by SPC or TGF-beta. These results suggest that SPC induces differentiation of hATSCs to smooth-muscle-like cell types through G(i/o)-ERK-dependent autocrine secretion of TGF-beta, which activates a Smad2-SRF/myocardin-dependent pathway.


Subject(s)
Mesenchymal Stem Cells/cytology , Myocytes, Smooth Muscle/cytology , Phosphorylcholine/analogs & derivatives , Sphingosine/analogs & derivatives , Transforming Growth Factor beta/physiology , Actins/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Humans , Mesenchymal Stem Cells/drug effects , Muscle, Smooth/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/physiology , Phosphorylation/drug effects , Phosphorylcholine/pharmacology , Serum Response Factor/physiology , Signal Transduction/drug effects , Smad2 Protein/metabolism , Sphingosine/pharmacology , Stress Fibers/drug effects , Stress Fibers/metabolism , Subcutaneous Fat/cytology , Trans-Activators/physiology , Transforming Growth Factor beta1/metabolism
18.
J Lipid Res ; 47(3): 653-64, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16339111

ABSTRACT

Sphingosylphosphorylcholine (SPC) has been implicated in a variety of cellular responses, including proliferation and differentiation. In this study, we demonstrate that d-erythro-SPC, but not l-threo-SPC, stereoselectively stimulated the proliferation of human adipose tissue-derived mesenchymal stem cells (hADSCs), with a maximal increase at 5 microM, and increased the intracellular concentration of Ca(2+) ([Ca(2+)](i)) in hADSCs, which do not express known SPC receptors (i.e., OGR1, GPR4, G2A, and GPR12). The SPC-induced proliferation and increase in [Ca(2+)](i) were sensitive to pertussis toxin (PTX) and the phospholipase C (PLC) inhibitor U73122, suggesting that PTX-sensitive G proteins, Gi or Go, and PLC are involved in SPC-induced proliferation. In addition, SPC treatment induced the phosphorylation of c-Jun and extracellular signal-regulated kinase, and SPC-induced proliferation was completely prevented by pretreatment with the c-Jun N-terminal kinase (JNK)-specific inhibitor SP600125 but not with the MEK-specific inhibitor U0126. Furthermore, the SPC-induced proliferation and JNK activation were completely attenuated by overexpression of a dominant negative mutant of JNK2, and the SPC-induced activation of JNK was inhibited by pretreatment with PTX or U73122. Treatment of hADSCs with lysophosphatidic acid (LPA) receptor antagonist, Ki16425, had no impact on the SPC-induced increase in [Ca(2+)](i). However, SPC-induced proliferation was partially, but significantly, attenuated by pretreatment of the cells with Ki16425.These results indicate that SPC stimulates the proliferation of hADSCs through the Gi/Go-PLC-JNK pathway and that LPA receptors may be responsible in part for the SPC-induced proliferation.


Subject(s)
Adipose Tissue/metabolism , Cell Proliferation/drug effects , MAP Kinase Kinase 4/metabolism , Mesenchymal Stem Cells/metabolism , Phosphorylcholine/analogs & derivatives , Sphingosine/analogs & derivatives , Calcium/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , Humans , Lysophosphatidylcholines/metabolism , Mitogen-Activated Protein Kinase 9/genetics , Mitogen-Activated Protein Kinase 9/metabolism , Models, Biological , Pertussis Toxin/metabolism , Pertussis Toxin/pharmacology , Phosphorylcholine/metabolism , Phosphorylcholine/pharmacology , Receptors, Lysophosphatidic Acid/metabolism , Signal Transduction , Sphingosine/metabolism , Sphingosine/pharmacology , Type C Phospholipases/metabolism
19.
Int J Biochem Cell Biol ; 37(11): 2357-65, 2005 Nov.
Article in English | MEDLINE | ID: mdl-15979922

ABSTRACT

Interleukin-6 (IL-6) subfamily of cytokines, including oncostatin M (OSM), leukemia inhibitory factor (LIF), and IL-6, has been implicated in a variety of physiological responses, such as cell growth, differentiation, and inflammation. In the present study, we demonstrated that both OSM and LIF stimulated the proliferation of human adipose tissue-derived mesenchymal stem cells (hATSCs), however, IL-6 had no effect on cell proliferation. OSM treatment induced phosphorylation of ERK, and pretreatment with U0126, a MEK inhibitor, prevented the OSM-stimulated proliferation of hATSCs, suggesting that the MEK/ERK pathway is involved in the OSM-induced proliferation. Treatment with OSM also induced phosphorylation of JAK2 and JAK3, and pretreatment of the cells with WHI-P131, a JAK3 inhibitor, but not with AG490, a JAK2 inhibitor, attenuated the OSM-induced proliferation of hATSCs. Furthermore, OSM treatment elicited phosphorylation of STAT1 and STAT3, and pretreatment with WHI-P131 specifically prevented the OSM-induced phosphorylation of STAT1, without affecting the OSM-induced phosphorylation of ERK and STAT3. These results suggest that two separate signaling pathways, such as MEK/ERK and JAK3/STAT1, are independently involved in the OSM-stimulated proliferation of hATSCs.


Subject(s)
Adipose Tissue/cytology , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Mesoderm/cytology , Peptides/pharmacology , Stem Cells/drug effects , Stem Cells/physiology , Butadienes/metabolism , Cells, Cultured , Enzyme Inhibitors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Interleukin-6/pharmacology , Janus Kinase 3 , Leukemia Inhibitory Factor , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Nitriles/metabolism , Oncostatin M , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Stem Cells/cytology
20.
J Cell Biochem ; 95(6): 1135-45, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-15962287

ABSTRACT

Platelet-derived growth factor (PDGF) is a critical regulator of proliferation and migration for mesenchymal type cells. In this study, we examined the role of mitogen-activated protein (MAP) kinases in the PDGF-BB-induced proliferation and migration of human adipose tissue-derived mesenchymal stem cells (hATSCs). The PDGF-induced proliferation was prevented by a pretreatment with the c-Jun N-terminal kinase (JNK) inhibitor, SP600125. However, it was not prevented by a pretreatment with a p38 MAP kinase inhibitor, SB202190, and a specific inhibitor of the upstream kinase of extracellular signal-regulated kinase (ERK1/2), U0126. Treatment with PDGF induced the activation of JNK and ERK in hATSCs, and pretreatment with SP600125 specifically inhibited the PDGF-induced activation of JNK. Treatment with PDGF induced the cell cycle transition from the G0/G1 phase to the S phase, the elevated expression of cyclin D1, and the phosphorylation of Rb, which were prevented by a pretreatment with SP600125. In addition, the PDGF-induced migration of hATSCs was completely blocked by a pretreatment with SP600125, but not with U0126 and SB202190. These results suggest that JNK protein kinase plays a key role in the PDGF-induced proliferation and migration of mesenchymal stem cells.


Subject(s)
Adipose Tissue/cytology , Adipose Tissue/drug effects , Cell Movement/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Platelet-Derived Growth Factor/pharmacology , Adipose Tissue/enzymology , Becaplermin , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclin D1/metabolism , Humans , Mesenchymal Stem Cells/cytology , Osteogenesis/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-sis , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...