Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters











Publication year range
1.
Int J Biol Sci ; 8(10): 1375-84, 2012.
Article in English | MEDLINE | ID: mdl-23139635

ABSTRACT

Liver X receptors (LXRs) play important roles in regulating cholesterol homeostasis, and lipid and energy metabolism. Therefore, LXR ligands could be used for the management of metabolic disorders. We evaluated rhein, a natural compound from Rheum palmatum L., as an antagonist for LXRs and investigated its anti-obesity mechanism in high-fat diet-fed mice. Surface plasmon resonance assays were performed to examine the direct binding of rhein to LXRs. LXR target gene expression was assessed in 3T3-L1 adipocytes and HepG2 hepatic cells in vitro. C57BL/6J mice fed a high-fat diet were orally administered with rhein for 4 weeks, and then the expression levels of LXR-related genes were analyzed. Rhein bound directly to LXRs. The expression levels of LXR target genes were suppressed by rhein in 3T3-L1 and HepG2 cells. In white adipose tissue, muscle and liver, rhein reprogrammed the expression of LXR target genes related to adipogenesis and cholesterol metabolism. Rhein activated uncoupling protein 1 (UCP1) expression in brown adipose tissue (BAT) in wild-type mice, but did not affect UCP1 expression in LXR knockout mice. In HIB-1B brown adipocytes, rhein activated the UCP1 gene by antagonizing the repressive effect of LXR on UCP1 expression. This study suggests that rhein may protect against obesity and related metabolic disorders through LXR antagonism and regulation of UCP1 expression in BAT.


Subject(s)
Adipose Tissue, Brown/drug effects , Anthraquinones/pharmacology , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Obesity/drug therapy , Orphan Nuclear Receptors/antagonists & inhibitors , Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Administration, Oral , Animals , Anthraquinones/chemistry , Anthraquinones/therapeutic use , Anti-Obesity Agents/chemistry , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Diet, High-Fat , Gene Expression/drug effects , Gene Knockout Techniques , Ion Channels/genetics , Liver X Receptors , Metabolic Diseases/drug therapy , Metabolic Diseases/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/genetics , Molecular Sequence Data , NIH 3T3 Cells , Obesity/metabolism , Orphan Nuclear Receptors/genetics , Rheum/chemistry , Surface Plasmon Resonance , Uncoupling Protein 1
2.
Clin Exp Med ; 12(3): 173-80, 2012 Sep.
Article in English | MEDLINE | ID: mdl-21932124

ABSTRACT

The objectives of this study were to assess the toxicity and immunological response induced by the intra-dermal (i.d.) administration of MUC1-peptide-pulsed dendritic cells (DCs) in advanced pancreatic cancer patients. Patients with recurrent lesions or metastasis after surgery, and immunohistochemistry positive for MUC1 were treated in cohorts that received 3-6 × 10(6) DCs i.d. for three or four vaccines. Each vaccine was composed of autologus DCs pulsed with MUC1-peptide. Peripheral blood mononuclear cells (PBMCs) that harvested 2 weeks after the second immunization were compared with PBMCs obtained before treatment for immunological response. Serial ELISPOT assays of PBMCs for antitumor reactivity were performed. Three patients received all four vaccines, and four patients received three vaccines. These patients were evaluable for toxicity and immunological monitoring. There were no grade 3 or 4 toxicities associated with the vaccines or major evidence of autoimmunity. Interferon-γ and granzyme B ELISPOT assay reactivity increased significantly in 2 of 7 patients (P < 0.05). The administration of MUC1-peptide-pulsed DCs is non-toxic and capable of inducing immunological response to tumor antigen MUC1 in advanced pancreatic cancer patients. Additional studies are necessary to improve tumor rejection responses.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Mucin-1/immunology , Pancreatic Neoplasms/therapy , Amino Acid Sequence , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/metabolism , Cell Proliferation , Cell Survival , Dendritic Cells/metabolism , Enzyme-Linked Immunospot Assay , Female , Granzymes/immunology , Humans , Immunity, Cellular , Immunohistochemistry , Injections, Intradermal , Interferon-gamma/immunology , Male , Middle Aged , Molecular Sequence Data , Mucin-1/administration & dosage , Mucin-1/metabolism , Neoplasm Staging , Pancreatic Neoplasms/immunology , Peptides/administration & dosage , Peptides/immunology , Peptides/metabolism , Phenotype , Pilot Projects , Vaccination
3.
Cell Res ; 21(9): 1358-69, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21519350

ABSTRACT

Immunization with inactivated autoreactive T cells is an effective therapeutic approach to ameliorating autoimmune diseases, while the underlying mechanisms that regulate autoreactive T cells are not completely understood. This study tested the hypothesis that T-cell vaccination (TCV) inhibits autoimmune diabetes in mice through the suppression of Th17 cells. The results showed that TCV treatment decreased hyperglycemia in type 1 diabetes (T1D) induced by multiple low-dose streptozotocin (MLD-STZ) as compared with the controls, preserved the number of healthy pancreatic islets and increased the production of insulin in the islets. Further study revealed that TCV significantly decreased the production of both interleukin (IL)-17 and IL-23 in intrapancreatic infiltrating lymphocytes (IPL) through marked inhibition of mRNA level of retinoic acid-related orphan receptor γt (RORγt) and signal transducer and activator of transcription 3 (Stat3) phosphorylation. The role of TCV-induced Th17 suppression was further validated in adoptive transfer experiments with polarized Th17 cells in sub-diabetogenic mice, which was similar to the effect of anti-IL-17 antibody treatment. Collectively our study shows that intrapancreatic Th17 cell suppression and healthy islet preservation play an important role in the treatment of T1D by TCV.


Subject(s)
Diabetes Mellitus, Experimental/prevention & control , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , T-Lymphocytes/immunology , Th17 Cells/physiology , Animals , Cell Line , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Experimental/metabolism , Insulin/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-23/metabolism , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phosphorylation , RNA, Messenger/metabolism , Signal Transduction , Th17 Cells/immunology , Th17 Cells/metabolism , Vaccination
4.
Am J Physiol Endocrinol Metab ; 300(5): E886-93, 2011 May.
Article in English | MEDLINE | ID: mdl-21364120

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is associated with obesity, insulin resistance, and inflammatory disorders. In this study, we tested the effect of rhein, a lipophilic anthraquinone derived from a traditional Chinese herbal medicine Rheum palmatum L., on NAFLD-associated hepatic steatosis, insulin resistance, and the T helper (Th)1/Th2 cytokine imbalance in high-fat diet-induced obese (DIO) mice. We found that oral administration of rhein for 40 days significantly increased energy expenditure, reduced body weight, particularly body fat content, improved insulin resistance, and lowered circulating cholesterol levels in DIO mice without affecting food intake. Rhein treatment also reduced liver triglyceride levels, reversed hepatic steatosis, and normalized alanine aminotransferase (ALT) levels in these mice. Gene analysis and Western blot showed that rhein markedly suppressed the expression of the lipogenic enzyme sterol regulatory element-binding protein-1c (SREBP-1c) and its target genes in the liver. Luciferase reporter assay revealed that rhein suppressed the transcriptional activity of SREBP-1c through its upstream regulator, liver X receptor (LXR). This suggests that rhein exerts its effects by targeting LXR, which is also supported by its inability to reduce body weight in LXR knockout mice. Moreover, multiplex ELISA displayed a downregulated Th1 response after rhein treatment. Rhein shifted the Th1/Th2 responses by inhibiting T-box expressed in T-cells (T-bet) expression and enhancing GATA-binding protein-3 (GATA-3) expression through increased signal transducer and activator of transcription 6 (STAT6) phosphorylation. These data indicate that rhein ameliorated NAFLD and associated disorders through LXR-mediated negative energy balance, metabolic regulatory pathways, and immunomodulatory activities involved in hepatic steatosis. The combined effects of rhein to target hepatic metabolic and immune pathways may be beneficial for complex metabolic diseases such as NAFLD.


Subject(s)
Anthraquinones/therapeutic use , Energy Metabolism/drug effects , Enzyme Inhibitors/therapeutic use , Fatty Liver/drug therapy , Fatty Liver/metabolism , Lipogenesis/drug effects , Liver/metabolism , Obesity/immunology , Obesity/metabolism , Adiposity/drug effects , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Cell Line , Chemokines/metabolism , Cytokines/metabolism , Diet , Female , Gene Expression/drug effects , Glucose Tolerance Test , Lipids/blood , Liver/drug effects , Liver X Receptors , Luciferases/metabolism , Mice , Mice, Inbred C57BL , Orphan Nuclear Receptors/metabolism , Oxygen Consumption/drug effects , Transfection
5.
J Clin Invest ; 121(2): 658-70, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21266776

ABSTRACT

Th17 cells are a subset of CD4+ T cells with an important role in clearing certain bacterial and fungal pathogens. However, they have also been implicated in autoimmune diseases such as multiple sclerosis. Exposure of naive CD4+ T cells to IL-6 and TGF-ß leads to Th17 cell differentiation through a process in which many proteins have been implicated. We report here that ectopic expression of liver X receptor (LXR) inhibits Th17 polarization of mouse CD4+ T cells, while LXR deficiency promotes Th17 differentiation in vitro. LXR activation in mice ameliorated disease in the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis, whereas LXR deficiency exacerbated disease. Further analysis revealed that Srebp-1, which is encoded by an LXR target gene, mediated the suppression of Th17 differentiation by binding to the E-box element on the Il17 promoter, physically interacting with aryl hydrocarbon receptor (Ahr) and inhibiting Ahr-controlled Il17 transcription. The putative active site (PAS) domain of Ahr and the N-terminal acidic region of Srebp-1 were essential for this interaction. Additional analyses suggested that similar LXR-dependent mechanisms were operational during human Th17 differentiation in vitro. This study reports what we believe to be a novel signaling pathway underlying LXR-mediated regulation of Th17 cell differentiation and autoimmunity.


Subject(s)
Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/immunology , Orphan Nuclear Receptors/immunology , Th17 Cells/immunology , Animals , Autoimmunity/immunology , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Liver X Receptors , Mice , Mice, Knockout , Orphan Nuclear Receptors/agonists , Orphan Nuclear Receptors/genetics , Signal Transduction/immunology , Sterol Regulatory Element Binding Protein 1/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Th17 Cells/cytology
6.
Diabetes ; 59(12): 3041-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20855545

ABSTRACT

OBJECTIVE: Toll-like receptor 4 (TLR4) has been reported to induce insulin resistance through inflammation in high-fat-fed mice. However, the physiological role of TLR4 in metabolism is unknown. Here, we investigated the involvement of TLR4 in fasting metabolism. RESEARCH DESIGN AND METHODS: Wild-type and TLR4 deficient (TLR4(-/-)) mice were either fed or fasted for 24 h. Glucose and lipid levels in circulation and tissues were measured. Glucose and lipid metabolism in tissues, as well as the expression of related enzymes, was examined. RESULTS: Mice lacking TLR4 displayed aggravated fasting hypoglycemia, along with normal hepatic gluconeogenesis, but reversed activity of pyruvate dehydrogenase complex (PDC) in skeletal muscle, which might account for the fasting hypoglycemia. TLR4(-/-) mice also exhibited higher lipid levels in circulation and skeletal muscle after fasting and reversed expression of lipogenic enzymes in skeletal muscle but not liver and adipose tissue. Adipose tissue lipolysis is normal and muscle fatty acid oxidation is increased in TLR4(-/-) mice after fasting. Inhibition of fatty acid synthesis in TLR4(-/-) mice abolished hyperlipidemia, hypoglycemia, and PDC activity increase, suggesting that TLR4-dependent inhibition of muscle lipogenesis may contribute to glucose and lipid homeostasis during fasting. Further studies showed that TLR4 deficiency had no effect on insulin signaling and muscle proinflammatory cytokine production in response to fasting. CONCLUSIONS: These data suggest that TLR4 plays a critical role in glucose and lipid metabolism independent of insulin during fasting and identify a novel physiological role for TLR4 in fuel homeostasis.


Subject(s)
Fasting/physiology , Toll-Like Receptor 4/physiology , Adipose Tissue/physiology , Animals , Carbon Dioxide/analysis , DNA, Complementary/genetics , Fatty Acids/metabolism , Homeostasis , Hypoglycemia/genetics , Lipolysis , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Oxygen Consumption , Polymerase Chain Reaction , RNA/drug effects , RNA/isolation & purification , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
7.
J Immunol ; 185(3): 1855-63, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20622114

ABSTRACT

Berberine (BBR), an isoquinoline alkaloid derived from plants, is widely used as an anti-inflammatory remedy in traditional Chinese medicine. In this study, we showed that BBR was efficacious in the amelioration of experimental autoimmune encephalomyelitis (EAE) through novel regulatory mechanisms involving pathogenic Th1 and Th17 cells. BBR inhibited differentiation of Th17 cells and, to a lesser degree, Th1 cells through direct actions on the JAK/STAT pathway, whereas it had no effect on the relative number of CD4(+)Foxp3(+) regulatory T cells. In addition, BBR indirectly influenced Th17 and Th1 cell functions through its effect on the expression and function of costimulatory molecules and the production of IL-6, which was attributable to the inhibition of NF-kappaB activity in CD11b(+) APCs. BBR treatment completely abolished the encephalitogenicity of MOG(35-55)-reactive Th17 cells in an adoptive transfer EAE model, and the same treatment significantly inhibited the ability of MOG(35-55)-reactive Th1 cells to induce EAE. This study provides new evidence that natural compounds, such as BBR, are of great value in the search for novel anti-inflammatory agents and therapeutic targets for autoimmune diseases.


Subject(s)
Berberine/therapeutic use , Cell Differentiation/drug effects , Cell Differentiation/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Interleukin-17/biosynthesis , Th1 Cells/drug effects , Th1 Cells/immunology , Amino Acid Sequence , Animals , Cells, Cultured , Drugs, Chinese Herbal/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , T-Lymphocytes, Helper-Inducer/cytology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/cytology
8.
J Immunol ; 185(4): 2563-9, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20631308

ABSTRACT

IL-17 is a recently identified proinflammatory cytokine that plays pivotal roles in several chronic inflammatory disease models. Its expression was also found to be elevated in the serum of patients with chronic diseases. However, whether elevated systemic IL-17 expression can induce pathophysiological tissue inflammation is unknown. In this study, we demonstrated that systemic overexpression of IL-17 using an adenoviral vector could induce multiple tissue inflammation and wasting in mice. We also found that the expression of TLR4 was increased in tissues of IL-17-overexpressing mice. Moreover, TLR4 activation is required for IL-17-induced tissue inflammation and wasting, as evidenced by the absence of aggressive atrophy in gastrocnemius muscle, neutrophil accumulation, and expression of proinflammatory cytokines downstream of TLR4 in multiple tissues of TLR4-deficient mice. Further investigation revealed that TLR4 endogenous ligands high-mobility group box 1 and heat shock protein 22, were systemically upregulated and might be involved in the IL-17-induced TLR4 activation. Our results suggest that IL-17 may induce disease-associated tissue inflammation and wasting through TLR4 signaling. The study indicates a novel interaction between IL-17 and TLR4 activation and may have implications in the pathogenesis and treatment of chronic diseases.


Subject(s)
Inflammation/metabolism , Interleukin-17/metabolism , Toll-Like Receptor 4/metabolism , Wasting Syndrome/metabolism , Adenoviridae/genetics , Animals , Blotting, Western , Body Weight/genetics , Body Weight/physiology , Chemokines/genetics , Chemokines/metabolism , Cytokines/genetics , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Inflammation/blood , Inflammation/genetics , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Interleukin-17/blood , Interleukin-17/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Toll-Like Receptor 4/genetics , Transduction, Genetic , Wasting Syndrome/blood , Wasting Syndrome/genetics
9.
PLoS One ; 5(4): e10149, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20405019

ABSTRACT

BACKGROUND: TGF-beta has been postulated to play an important role in the maintenance of epithelial homeostasis and the development of epithelium-derived cancers. However, most of previous studies are mainly focused on the function of TGF-beta in immune cells to the development of allergic asthma and how TGF-beta signaling in airway epithelium itself in allergic inflammation is largely unknown. Furthermore, the in vivo TGF-beta function specifically in the airway epithelium during lung cancer development has been largely elusive. METHODOLOGY/PRINCIPAL FINDINGS: To evaluate the in vivo contribution of TGF-beta signaling in lung epithelium to the development of allergic disease and lung cancer, we generated a transgenic mouse model with Smad7, an intracellular inhibitor of TGF-beta signaling, constitutively expressed in mouse airway Clara cells using a mouse CC10 promoter. The mice were subjected to the development of OVA-induced allergic asthma and urethane-induced lung cancer. The Smad7 transgenic animals significantly protected from OVA-induced asthma, with reduced airway inflammation, airway mucus production, extracellular matrix deposition, and production of OVA-specific IgE. Further analysis of cytokine profiles in lung homogenates revealed that the Th2 cytokines including IL-4, IL-5 and IL-13, as well as other cytokines including IL-17, IL-1, IL-6, IP10, G-CSF, and GM-CSF were significantly reduced in the transgenic mice upon OVA induction. In contrast, the Smad7 transgenic animals had an increased incidence of lung carcinogenesis when subjected to urethane treatment. CONCLUSION/SIGNIFICANCE: These studies, therefore, demonstrate for the first time the in vivo function of TGF-beta signaling specifically in airway epithelium during the development of allergic asthma and lung cancer.


Subject(s)
Asthma/etiology , Lung Neoplasms/etiology , Respiratory Mucosa/metabolism , Signal Transduction , Smad7 Protein/pharmacology , Transforming Growth Factor beta/physiology , Animals , Asthma/chemically induced , Asthma/therapy , Cytokines/analysis , Disease Models, Animal , Genetic Therapy , Inflammation/prevention & control , Lung Neoplasms/chemically induced , Lung Neoplasms/therapy , Mice , Mice, Transgenic , Ovalbumin , Signal Transduction/drug effects , Smad7 Protein/genetics , Th2 Cells , Transforming Growth Factor beta/antagonists & inhibitors , Urethane
10.
Cell Res ; 20(4): 480-91, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20084083

ABSTRACT

Alpha-galactosylceramide (alpha-GC) is widely known to activate invariant natural killer T (iNKT) cells to suppress myelin antigen-specific Th1 responses, protecting susceptible mice against experimental autoimmune encephalomyelitis (EAE). Here, we demonstrate an unexpected finding that high doses of alpha-GC exacerbated, rather than ameliorated, EAE. Similar results were observed when MOG(35-55)-specific T cells treated with high-dose alpha-GC were transferred into naïve syngeneic recipient mice. Further study showed that high doses of alpha-GC directly enhance the Th17 and Th1 response by activation of CD4(+)CD44(+) memory T cells through phosphorylation of STAT3 and activation of NF-kappaB. Unlike the activation of iNKT cells by low doses of alpha-GC, high doses of alpha-GC directly interacted with CD1d expressed on T cells and activated Th17 and Th1 cells. Furthermore, antigen-presenting cells (APCs) predominantly express CD1d1, whereas the majority of CD4(+) T cells express CD1d2. Knockdown of CD1d1 or CD1d2 gene expression by RNAi interfered with the activation of iNKT or Th17/Th1 cells, respectively. Therefore, alpha-GC treatment could improve or worsen EAE by engaging either APCs or Th17/Th1 cells depending on the dose used.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Galactosylceramides/toxicity , Interleukin-17/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens, CD1/metabolism , Antigens, CD1d/genetics , Antigens, CD1d/metabolism , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/metabolism , Gene Knockdown Techniques , Mice , NF-kappa B/metabolism , Natural Killer T-Cells/immunology , RNA Interference , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Helper-Inducer/cytology , Th1 Cells/immunology
11.
J Biol Chem ; 284(41): 28420-28429, 2009 Oct 09.
Article in English | MEDLINE | ID: mdl-19661066

ABSTRACT

Berberine, an alkaloid derivative from Berberis vulgaris L., has been used extensively in traditional Chinese medicine to treat diarrhea and diabetes, but the underlying mechanisms for treating diabetes are not fully understood. Recent studies suggested that berberine has many beneficial biological effects, including anti-inflammation. Because type 1 diabetes is caused by T cell-mediated destruction of beta cells and severe islet inflammation, we hypothesized that berberine could ameliorate type 1 diabetes through its immune regulation properties. Here we reported that 2 weeks of oral administration of berberine prevented the progression of type 1 diabetes in half of the NOD mice and decreased Th17 and Th1 cytokine secretion. Berberine suppressed Th17 and Th1 differentiation by reducing the expression of lineage markers. We found that berberine inhibited Th17 differentiation by activating ERK1/2 and inhibited Th1 differentiation by inhibiting p38 MAPK and JNK activation. Berberine down-regulated the activity of STAT1 and STAT4 through the suppression of p38 MAPK and JNK activation, and it controlled the stability of STAT4 through the ubiquitin-proteasome pathway. Our findings indicate that berberine targets MAPK to suppress Th17 and Th1 differentiation in type 1 diabetic NOD mice. This study revealed a novel role of ERK in Th17 differentiation through down-regulation of STAT3 phosphorylation and RORgamma t expression.


Subject(s)
Berberine/therapeutic use , Cell Differentiation/physiology , Diabetes Mellitus, Type 1/drug therapy , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-17/immunology , JNK Mitogen-Activated Protein Kinases/metabolism , Th1 Cells/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Berberine/immunology , Cytokines/immunology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Disease Models, Animal , Disease Progression , Female , Humans , Lymph Nodes/cytology , Lymph Nodes/immunology , Mice , Mice, Inbred NOD , Nuclear Receptor Subfamily 1, Group F, Member 3 , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/genetics , Receptors, Thyroid Hormone/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Spleen/cytology , Spleen/immunology , Th1 Cells/cytology , Th1 Cells/immunology
12.
Int J Mol Med ; 23(4): 449-54, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19288019

ABSTRACT

Lecithin is an essential biological component and widely used as a nutritional supplement for protecting cells from oxidation, increase fat burning and preventing cardiovascular disease. Lecithin contains fatty acids identified as the peroxisome proliferator-activated receptor (PPAR) agonists. However, the role of lecithin in adipogenesis and lipogenesis remains elusive. 3T3-L1 cells and mouse primary preadipocytes were used to characterize the properties of lecithin related to adipogenesis and lipogenesis. We found that lecithin promoted adipocyte differentiation and differentiation-specific gene expression, and increased triglycerides and free fatty acid levels in the adipocytes. These effects are independent of the clonal expansion of 3T3-L1 cells and the upstream PPARgamma regulator, CCAAT-enhancer-binding protein beta. Furthermore, lecithin induced lipid accumulation in human hepatoma HepG2 cells. Our data suggest that lecithin is involved in adipogenesis, lipogenesis and hepatic lipid accumulation and it is implicated in obesity and hepatic steatosis.


Subject(s)
Adipocytes/drug effects , Cell Differentiation/drug effects , Fatty Acids, Nonesterified/metabolism , Lecithins/pharmacology , Triglycerides/metabolism , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/metabolism , Animals , Biological Transport/drug effects , Blotting, Western , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Gene Expression/drug effects , Glucose/pharmacokinetics , Humans , Insulin/pharmacology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , Reverse Transcriptase Polymerase Chain Reaction
13.
Cell Res ; 19(5): 574-83, 2009 May.
Article in English | MEDLINE | ID: mdl-19188932

ABSTRACT

Glatiramer acetate (GA) is an immunomodulatory peptide drug used to treat multiple sclerosis. Its treatment effect has been expanded to other autoimmune conditions such as uveoretinitis, inflammatory bowel disease, graft rejection and hepatic fibrosis. Here, we report that GA was effective in altering the clinical course of diabetes in cyclophosphamide (CY)-potentiated non-obese diabetic (CY-NOD) mice. Treatment with GA significantly reduced the diabetic rate in the mice and ameliorated insulitis, which coincided with increased CD4+CD25+Foxp3+ T cell response in treated mice. GA treatment led to increased expression of transcription factor Foxp3 and elevated production of interleukin-4 (IL-4) both in vivo and in vitro. It was evident that the effect of GA on up-regulation of Foxp3 was mediated partially through IL-4. IL-4 was found to maintain Foxp3 expression and regulatory function of CD4+CD25+ regulatory T cells (Tregs). This study provides new evidence that GA has treatment potential for type 1 diabetes through the induction of Tregs and that increased IL-4 production is partially responsible for the enhanced Treg's function in GA treatment.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Hypoglycemic Agents/pharmacology , Peptides/pharmacology , T-Lymphocytes, Regulatory/immunology , Animals , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Glatiramer Acetate , Interleukin-4/metabolism , Mice , Mice, Inbred NOD , T-Lymphocytes, Regulatory/metabolism , Up-Regulation
14.
Endocrinology ; 150(1): 104-13, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18818299

ABSTRACT

Peroxisome proliferator-activated receptor (PPAR) gamma is a nuclear receptor that coordinates carbohydrate and lipid metabolism, and is a therapeutic target for type 2 diabetes. Tanshinone IIA (Tan) is a lipophilic diterpene that is widely used to treat cardiovascular diseases in traditional Chinese medicine, and has recently been found to reduce body weight and lower blood lipids. However, its underlying mechanism of antiadipogenic effects remains unknown. Here, we report that Tan inhibits 3T3-L1 preadipocyte differentiation and transcriptional activities of full-length PPARgamma and PPARgamma ligand-binding domains. The effects of Tan are mediated through its property as a natural antagonist of PPARgamma (dissociation constant of an inhibitor value, 2.562 +/- 0.711 microm). Tan treatment reduced adipose mass and body weight, improved glucose tolerance, and lowered the low-density lipoprotein to high-density lipoprotein ratio without changing the food intake in a high-fat diet-induced obese animal model. Our results suggest that the combined properties of Tan in adipogenesis, glucose tolerance, lipogenesis, and cardiovascular protection are beneficial for treating diabetic patients with complex metabolic conditions, in which modulating a single target is often not sufficient to achieve the desired effect.


Subject(s)
3T3 Cells/physiology , Anti-Obesity Agents/pharmacology , Obesity/prevention & control , PPAR gamma/antagonists & inhibitors , Phenanthrenes/pharmacology , 3T3 Cells/drug effects , Abietanes , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , DNA, Complementary/genetics , Female , Flow Cytometry , Genes, Reporter , Glucose Tolerance Test , Lipids/blood , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , RNA/genetics , RNA/isolation & purification , Transfection
15.
PPAR Res ; 2008: 581348, 2008.
Article in English | MEDLINE | ID: mdl-19096709

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are transcriptional factors involved in the regulation of insulin resistance and adipogenesis. Cinnamon, a widely used spice in food preparation and traditional antidiabetic remedy, is found to activate PPARgamma and alpha, resulting in improved insulin resistance, reduced fasted glucose, FFA, LDL-c, and AST levels in high-caloric diet-induced obesity (DIO) and db/db mice in its water extract form. In vitro studies demonstrate that cinnamon increases the expression of peroxisome proliferator-activated receptors gamma and alpha (PPARgamma/alpha) and their target genes such as LPL, CD36, GLUT4, and ACO in 3T3-L1 adipocyte. The transactivities of both full length and ligand-binding domain (LBD) of PPARgamma and PPARalpha are activated by cinnamon as evidenced by reporter gene assays. These data suggest that cinnamon in its water extract form can act as a dual activator of PPARgamma and alpha, and may be an alternative to PPARgamma activator in managing obesity-related diabetes and hyperlipidemia.

16.
Cell Biol Int ; 32(4): 456-61, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18272410

ABSTRACT

Type I diabetes is characterized by the deficiency of endocrine beta cells in the pancreatic islets of Langerhans and transplantation of islet cells can be an effective therapeutic approach. Embryonic stem cells can be differentiated into any cell type, and therefore represent an unlimited source of islet cells for the transplantation and treatment for type I diabetes. We have adopted an easy and reproducible in vitro differentiation system with a reduced serum concentration plus nicotinamide to generate early pancreatic progenitor cells from embryonic stem cells. Gene expression analysis indicated that the differentiated cells expressed not only endoderm markers such as GATA-4, HNF-3beta, but also early markers of pancreatic development including key transcription factors PDX-1 and IAPP. Some pancreatic specific markers, such as insulin I, insulin II, Glu-2 and glucagon, were also expressed to some extent at the mRNA level. Differentiated ES cells showed low level immunoreactivity for insulin. However, transplantation of these early pancreatic progenitor clusters into STZ-induced diabetic mice failed to reverse the hyperglycemic state of the disease as reported previously. The results suggest that culture manipulation can direct ES cells to differentiate into early pancreatic progenitor cells committing to pancreatic islet cell fate, but these cells cannot function normally to reduce blood glucose of diabetic mice at this stage.


Subject(s)
Cell Differentiation , Diabetes Mellitus, Experimental/therapy , Embryonic Stem Cells/cytology , Islets of Langerhans Transplantation , Pancreas/cytology , Stem Cells/cytology , Animals , Blood Glucose/metabolism , Cell Differentiation/genetics , Cell Shape , Gene Expression Regulation , Immunohistochemistry , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Stem Cell Transplantation , Streptozocin
17.
Mol Med Rep ; 1(1): 61-70, 2008.
Article in English | MEDLINE | ID: mdl-21479379

ABSTRACT

Dendritic cells (DCs) play a pivotal role in T cell-mediated immunity and have been shown to induce strong anti-tumor immune responses. As of yet, only a limited number of objective tumor regressions have been observed in clinical studies using a DC vaccine. Suppressor of cytokine signaling-1 (SOCS1) is a key negative regulator of the JAK/STAT signal pathway and plays an essential role in suppressing systemic autoimmunity that is mediated by DCs. The aim of this study was to investigate whether SOCS1-silenced DCs can break the vaccine-induced immune tolerance stimulated by high-dose DC, thereby enhancing anti-tumor activity. In the mouse melanoma model, we found that a 2x106 TRP2-pulsed DC vaccine was able to induce immune tolerance, while a 2x106 SOCS1-silenced DC/TRP2 vaccine prevented immune tolerance. Further experiments revealed that activation-induced T cell death (AICD) through the Fas/Fas-L pathway may play a crucial role in immune tolerance induced by 2x106 TRP2-pulsed DC. SOCS1-silencing in DCs could prevent immune tolerance by inhibiting Fas and Fas-L expression, induced by an increase in IL-12p70 and IL-6 production. In addition, in 2x106 SOCS1-silenced DC/TRP2 immunized mice, higher levels of IL-12p70 and IFN-γ and lower IL-17 production may inhibit tumor angiogenesis and therefore assist in breaking immune tolerance. In conclusion, high-doses of DCs can inhibit the vaccine-induced AICD of T cells and cytokine regulation in tumor angiogenesis. These results indicate that SOCS1-silenced DC vaccines may greatly enhance anti-tumor activity by breaking self-tolerance.

SELECTION OF CITATIONS
SEARCH DETAIL