Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Immunity ; 30(1): 80-91, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19144316

ABSTRACT

Foxp3(+) regulatory T (Treg) cells limit pathogenic immune responses to self-antigens and foreign antigens. An essential role for microRNA (miRNA) in the maintenance and function of Treg cells, revealed by the Treg cell-specific Dicer ablation, raised a question as to a specific miRNA contribution. We found that Foxp3 controlled the elevated miR155 expression required for maintaining Treg cell proliferative activity and numbers under nonlymphopenic conditions. Moreover, miR155 deficiency in Treg cells resulted in increased suppressor of cytokine signaling 1 (SOCS1) expression accompanied by impaired activation of signal transducer and activator of transcription 5 (STAT5) transcription factor in response to limiting amounts of interleukin-2. Our studies suggest that Foxp3-dependent regulation of miR155 maintains competitive fitness of Treg cell subsets by targeting SOCS1, and they provide experimental support for a proposed role for miRNAs in ensuring the robustness of cellular phenotypes.


Subject(s)
Forkhead Transcription Factors/physiology , MicroRNAs , Suppressor of Cytokine Signaling Proteins/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Base Sequence , Flow Cytometry , Forkhead Transcription Factors/pharmacology , Humans , Immunohistochemistry , Mice , Mice, Knockout , Molecular Sequence Data , Polymerase Chain Reaction , Sequence Alignment , Suppressor of Cytokine Signaling 1 Protein
2.
BMC Pulm Med ; 15: 13, 2015 Feb 18.
Article in English | MEDLINE | ID: mdl-25888222

ABSTRACT

BACKGROUND: Interleukin-27 (IL-27) is a multifunctional cytokine with both pro-inflammatory and immunoregulatory functions. At present, the role of IL-27 in pulmonary fibrosis remains unknown. METHODS: In this study, we observed the expression of IL-27/IL-27R in a mouse model of bleomycin (BLM)-induced pulmonary fibrosis. We verified the role of IL-27 using hematoxylin and eosin as well as Masson's staining methods and measuring the content of hydroxyproline as well as collagen I and III. We assessed the differentiation of T lymphocytes in the spleen and measured the concentration of cytokines in bronchoalveolar lavage fluid (BALF) and the expression level of relevant proteins in the JAK/STAT and TGF-ß/Smad signaling pathways in lung tissue. RESULTS: Increased IL-27 expression in BLM-induced pulmonary fibrosis was noted. IL-27 treatment may alleviate pulmonary fibrosis and increase the survival of mice. IL-27 inhibited the development of CD4(+) IL-17(+), CD4(+) IL-4(+) T, and CD4(+) Foxp3(+) cells and the secretion of IL-17, IL-4, IL-6, and TGF-ß. IL-27 induced the production of CD4(+) IL-10(+) and CD4(+) INF-γ(+) T cells. IL-27 decreased the levels of phosphorylated STAT1, STAT3, STAT5, Smad1, and Smad3 but increased the level of SOCS3. CONCLUSIONS: This study demonstrates that IL-27 potentially attenuates BLM-induced pulmonary fibrosis by regulating Th17 differentiation and cytokine secretion.


Subject(s)
Cell Differentiation/immunology , Cytokines/immunology , Interleukins/genetics , Lung/immunology , Pulmonary Fibrosis/immunology , Receptors, Cytokine/genetics , Th17 Cells/immunology , Animals , Antibiotics, Antineoplastic/toxicity , Bleomycin/toxicity , Blotting, Western , Bronchoalveolar Lavage Fluid/immunology , Cell Differentiation/drug effects , Cytokines/drug effects , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interleukins/immunology , Interleukins/pharmacology , Janus Kinases/drug effects , Janus Kinases/immunology , Lung/drug effects , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Pulmonary Fibrosis/chemically induced , Real-Time Polymerase Chain Reaction , Receptors, Cytokine/immunology , Receptors, Interleukin , Reverse Transcriptase Polymerase Chain Reaction , STAT Transcription Factors/drug effects , STAT Transcription Factors/immunology , Signal Transduction , Smad Proteins/drug effects , Smad Proteins/immunology , Spleen/cytology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Suppressor of Cytokine Signaling Proteins/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Th17 Cells/drug effects , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/immunology
3.
Br J Dermatol ; 171(3): 512-23, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24602010

ABSTRACT

BACKGROUND: Inflammatory chemokines, such as macrophage-derived chemokine (MDC/CCL22), are elevated in the serum and lesioned skin of patients with atopic dermatitis (AD), and are ligands for C-C chemokine receptor 4, which is predominantly expressed on T helper 2 lymphocytes, basophils and natural killer cells. We have previously reported that quercetagetin has an inhibitory activity on inflammatory chemokines, which is induced by interferon (IFN)-γ and tumour necrosis factor (TNF)-α, occurring via inhibition of the signal transducer and activator of transcription 1 (STAT1) signal. OBJECTIVES: To investigate the specific mechanisms of quercetagetin on the STAT1 signal. METHODS: We confirmed the inhibitory activity of quercetagetin on MDC and STAT1 in HaCaT keratinocytes. The interaction between STAT1 and IFN-γR1 was investigated using immunoprecipitation. The small interfering RNA approach was used to investigate the role of suppressor of cytokine signalling 1 (SOCS1) and transforming growth factor (TGF)-ß1 induced by quercetagetin. RESULTS: Quercetagetin inhibited the expression of MDC at both the protein and mRNA levels in IFN-γ- and TNF-α-stimulated HaCaT human keratinocytes. Moreover, quercetagetin inhibited the phosphorylation of STAT1 through upregulation of SOCS1. Increased expression of SOCS1 disrupted the binding of STAT1 to IFN-γR1. Furthermore, quercetagetin augmented the expression of TGF-ß1, which is known to modulate the immune response and inflammation. CONCLUSIONS: These results suggest that quercetagetin may be a potent inhibitor of the STAT1 signal, which could be a new molecular target for anti-inflammatory treatment, and may thus have therapeutic applications as an immune modulator in inflammatory diseases such as AD.


Subject(s)
Chemokine CCL22/antagonists & inhibitors , Chromones/pharmacology , Keratinocytes/drug effects , STAT1 Transcription Factor/drug effects , Transforming Growth Factor beta1/drug effects , Flavones , Humans , Interferon-gamma/drug effects , Janus Kinases/drug effects , Receptors, Interferon/drug effects , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Tumor Necrosis Factor-alpha/drug effects , Interferon gamma Receptor
4.
Acta Neurochir (Wien) ; 156(5): 941-9; discussion 949, 2014 May.
Article in English | MEDLINE | ID: mdl-24463741

ABSTRACT

BACKGROUND: There are complex interactions between acetylcholine (ACh), the suppressor of cytokine signaling-3 (SOCS-3), and cytokines, however, little is known about their dynamic expression or their effects on cerebral vasospasm (CVS) after subarachnoid hemorrhage (SAH). Therefore, we aimed to describe and clarify the dynamic expression of SOCS-3 and cytokines after SAH, as well as the relationships between the levels of SOCS-3, cytokines, and ACh. METHODS: The rat model of single cisterna magna injection was used to mimic acute SAH. The degree of CVS was indicated by lumen diameter and artery wall thickness under H&E staining. A semi-quantitative immunohistochemical analysis method was used to clarify the role of SOCS-3 in the CVS after SAH. We also measured the content of IL-6 and IL-10 in cerebrospinal fluid. RESULTS: We found that SOCS-3 expression levels increased rapidly within 12 h after SAH, more slowly after 12 h, and did not reach a peak within 48 h. Interleukin 6 (IL-6) levels rapidly increased within 24 h after SAH, reached a peak 24 h after SAH, and decreased slightly at 48 h. IL-10 levels increased during the first 6 h after SAH, after which this increase tapered off. ACh treatment reduced IL-6 levels and resulted in elevated levels of SOCS-3, but had no effect on IL-10 expression. Furthermore, ACh treatment relieved basilar arterial vasospasm, whereas mecamylamine pretreatment counteracted the activity of ACh. CONCLUSIONS: Taken together, these data indicate that SOCS-3 was involved in vasospasm via an IL-6- and IL-10-related mechanism, and that CVS following SAH could be reversed by the intraventricular injection of ACh.


Subject(s)
Basilar Artery/metabolism , Interleukin-10/cerebrospinal fluid , Interleukin-6/cerebrospinal fluid , Subarachnoid Hemorrhage/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Vasospasm, Intracranial/metabolism , Acetylcholine/pharmacology , Animals , Basilar Artery/drug effects , Cisterna Magna , Cytokines , Immunohistochemistry , Injections, Intraventricular , Interleukin-10/metabolism , Interleukin-6/metabolism , Male , Rats , Subarachnoid Hemorrhage/complications , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Vasoconstriction/drug effects , Vasodilator Agents/pharmacology , Vasospasm, Intracranial/etiology
5.
Mol Cell Neurosci ; 46(2): 507-15, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21145973

ABSTRACT

Intravitreal injections of recombinant ciliary neurotrophic factor (rCNTF) protect adult rat retinal ganglion cells (RGCs) after injury and stimulate regeneration, an effect enhanced by co-injection with a cAMP analogue (CPT-cAMP). This effect is partly mediated by PKA and associated signaling pathways, but CPT-cAMP also moderates upregulation of suppressor of cytokine signaling (SOCS) pathways after rCNTF injection, which will also enhance the responsiveness of RGCs to this and perhaps other cytokines. We now report that intravitreal injections of CPT-cAMP do not potentiate RGC axonal regeneration when CNTF is expressed via an adeno-associated viral vector (rAAV2), and concomitantly we show that increases in retinal SOCS mRNA expression are less when CNTF is delivered using the vector. We also directly tested the impact of elevated SOCS3 expression on the survival and regeneration of injured adult RGCs by injecting a bicistronic rAAV2-SOCS3-GFP vector into the vitreous of eyes in rats with a peripheral nerve graft sutured onto the cut optic nerve. Overexpression of SOCS3 resulted in an overall reduction in axonal regrowth and almost complete regeneration failure of RGCs transduced with the rAAV2-SOCS3-GFP vector. Furthermore, rAAV2-mediated expression of SOCS3 abolished the normally neurotrophic effects elicited by intravitreal rCNTF injections. In summary, CNTF delivery to the retina using viral vectors may be more effective than bolus rCNTF injections because the gene therapy approach has a less pronounced effect on neuron-intrinsic SOCS repressor pathways. Our new gain of function data using rAAV2-SOCS3-GFP demonstrate the negative impact of enhanced SOCS3 expression on the regenerative potential of mature CNS neurons.


Subject(s)
Axons/metabolism , Ciliary Neurotrophic Factor/administration & dosage , Genetic Therapy/methods , Nerve Regeneration/physiology , Retinal Ganglion Cells/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Adenoviridae/genetics , Animals , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Cyclic AMP/administration & dosage , Cyclic AMP/analogs & derivatives , Female , Gene Expression , Genetic Vectors/genetics , Immunohistochemistry , Intravitreal Injections , Microscopy, Confocal , Nerve Regeneration/drug effects , Neuroprotective Agents/administration & dosage , Optic Nerve Injuries/physiopathology , Optic Nerve Injuries/therapy , Polymerase Chain Reaction , RNA, Messenger/analysis , Rats , Rats, Wistar , Recombinant Proteins/administration & dosage , Retinal Ganglion Cells/drug effects , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Transduction, Genetic
6.
Immunology ; 131(1): 118-27, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20406299

ABSTRACT

While it is known that the anti-inflammatory effects of interleukin (IL)-4 require new protein synthesis, the exact mechanisms by which IL-4 suppresses the production of pro-inflammatory cytokines by human monocytes and macrophages is unclear. IL-4 rapidly induced suppressor of cytokine signalling-1 (SOCS1) mRNA and protein, which peaked at 60 min, much earlier than lipopolysaccharide (LPS)-induced SOCS1 mRNA and protein which were consistently maximal 4 hr post-exposure. SOCS1 is a molecule generally considered to be induced for negative feedback of inflammatory processes. We investigated whether the early induction of SOCS1 by IL-4 was responsible for the suppression of LPS-induced tumour necrosis factor (TNF)-alpha production by IL-4. IL-4 suppressed LPS-induced TNF-alpha in freshly isolated monocytes at the level of transcription but acted by a different, possibly translational, mechanism in monocytes cultured overnight in macrophage colony-stimulating factor (M-CSF). Despite different modes of regulation by IL-4, the kinetics and magnitude of induction of SOCS1 mRNA and protein by IL-4 in the two cell types were identical. There was no significant difference in the suppression by IL-4 of LPS-induced TNF-alpha production by bone-marrow derived macrophages from wild-type mice, Ifngamma(-/-) mice and mice lacking SOCS1 (Socs1(-/-)Ifngamma(-/-)). These data suggest that SOCS1 is not involved in the suppression of LPS-induced TNF-alpha production by IL-4.


Subject(s)
Anti-Inflammatory Agents , Interleukin-4 , Monocytes , Signal Transduction , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Cells, Cultured , Humans , Interferon-gamma/pharmacology , Interleukin-4/metabolism , Interleukin-4/pharmacology , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Suppressor of Cytokine Signaling Proteins/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
Am J Physiol Cell Physiol ; 297(5): C1294-306, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19741199

ABSTRACT

The cholinergic anti-inflammatory pathway is a physiological mechanism that inhibits cytokine production and minimizes tissue injury during inflammation. Previous investigations revealed that cholinergic stimulation (via cholinergic agonists and vagus nerve stimulation) suppresses endothelial cell activation and leukocyte recruitment. The purpose of this study was to investigate the mechanisms by which cholinergic agonists (e.g., nicotine and GTS-21) regulate endothelial cell activation. Specifically, we examined the effects of cholinergic agonists on IL-6-mediated endothelial cell activation through the JAK2/STAT3 signaling pathway. Treatment of macrovascular human umbilical vein endothelial cells (HUVECs) and microvascular endothelial cells (MVECs) with the cholinergic agonists nicotine and GTS-21 significantly reduced IL-6-mediated monocyte chemoattractant protein-1 (MCP-1) production and ICAM-1 expression which are regulated through the JAK2/STAT3 pathway. We found that treatment of endothelial cells with cholinergic agonists significantly reduced STAT3 activation by phosphorylation and DNA binding. The inhibition of STAT3 phosphorylation was reversed by sodium orthovanadate, an inhibitor of tyrosine phosphatases, as well as by NSC-87877 suggesting a SHP1/2-dependent mechanism. Further investigations showed that cholinergic agonists reduced the phosphorylation of JAK2, an upstream component of the JAK2/STAT3 pathway. Finally, we observed that nicotine and GTS-21 treatment decreased levels of SOCS3 (suppressor of cytokine signaling; a regulator of the inflammatory activity of IL-6) in activated endothelial cells. These data demonstrate that cholinergic agonists suppress IL-6-mediated endothelial cell activation through the JAK2/STAT3 pathway. Our results have significant implications for better understanding the therapeutic potential of cholinergic agonists for treating IL-6 mediated inflammatory conditions.


Subject(s)
Endothelial Cells/metabolism , Janus Kinase 2/metabolism , Nicotinic Agonists/pharmacology , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Benzylidene Compounds/pharmacology , Blotting, Western , Cells, Cultured , Chemokine CCL2/drug effects , Chemokine CCL2/metabolism , Endothelial Cells/drug effects , Humans , Intercellular Adhesion Molecule-1/drug effects , Intercellular Adhesion Molecule-1/metabolism , Interleukin-6/metabolism , Janus Kinase 2/drug effects , Nicotine/pharmacology , Phosphorylation/drug effects , Pyridines/pharmacology , STAT3 Transcription Factor/drug effects , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Suppressor of Cytokine Signaling Proteins/metabolism
8.
J Leukoc Biol ; 83(3): 672-9, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18156187

ABSTRACT

LPS of Porphyromonas gingivalis (P. gingivalis) is suggested to be a virulence factor in periodontitis, stimulating host cells to produce proinflammatory mediators. However, P. gingivalis LPS has been reported to show lower biological activity compared with Escherichia coli (E. coli) LPS. Although differences in the chemical structure of lipid A and the receptor conferring LPS signaling are thought to account for these characteristics, the precise reason is unknown. Here, we demonstrate that P. gingivalis LPS up-regulates IL-1R-associated kinase (IRAK)-M, a negative regulator of the TLR signaling pathway, in a THP-1-derived macrophage more robustly than E. coli LPS. Although down-regulation of IRAK-M by small interfering (si)RNA augmented transcription and translation of TNF-, IL-6, and IL-12 p40 in LPS-stimulated macrophages, the effect of siRNA was more prominent in P. gingivalis LPS-stimulated cells. Degradation of IRAK-1 was more obvious in E. coli LPS-stimulated macrophages than the cells stimulated with P. gingivalis LPS, suggesting that P. gingivalis LPS-induced IRAK-M suppressed dissociation of IRAK-1 from the receptor complex, resulting in escape from subsequent degradation. This activity may be involved in the chronic infection of this bacterium in periodontal tissue by serving as an escape mechanism from immune surveillance.


Subject(s)
Enzyme Induction/drug effects , Interleukin-1 Receptor-Associated Kinases/genetics , Lipopolysaccharides/pharmacology , Macrophages/enzymology , Monocytes/physiology , Porphyromonas gingivalis , Cell Line , Escherichia coli , Gene Expression Regulation, Enzymologic/drug effects , Humans , Interleukin-1 Receptor-Associated Kinases/biosynthesis , Lipopolysaccharides/isolation & purification , Macrophages/drug effects , NF-kappa B/physiology , RNA, Small Interfering , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Suppressor of Cytokine Signaling Proteins/genetics , Transfection , Tumor Necrosis Factor-alpha/genetics
9.
Kidney Int ; 74(11): 1434-43, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19008912

ABSTRACT

Renal hypertrophy and deposition of extracellular matrix proteins are consistent findings in diabetic nephropathy and these processes can be halted or reversed by euglycemic control. Using DNA microarray analysis of glomerular RNA from control and diabetic rats we found that the expression levels of insulin-like growth factor 1 receptor (IGF-1R) were increased while those of suppressor of cytokine signaling 2 (SOCS2) and STAT5 were decreased. All of these changes were normalized by islet cell transplantation. Overexpression of SOCS2 in rat mesangial cells inhibited IGF-1-induced activation of extracellular signal-regulated kinase, which subsequently reduced type IV collagen and DNA synthesis, an effect due to interaction of SOCS2 with IGF-1R. Inhibition of SOCS2 overexpression by small interfering RNA suppressed IGF-1R-mediated actions by preventing phosphorylation of tyrosine 317 in the p66Shc adaptor protein; however, overexpression of either SOCS1 or SOCS3 did not affect IGF-1R signaling. Insulin directly increased STAT5 and SOCS2 expression in mesangial cells. This study shows that insulin can inhibit the mitogenic action of IGF-1 in mesangial cells by regulating STAT5/SOCS2 expression. Insulin deficiency may contribute to the mesangial expansion found in diabetes through reduced STAT5/SOCS2 expression.


Subject(s)
Gene Expression Regulation/drug effects , Insulin-Like Growth Factor I/physiology , Insulin/pharmacology , Mesangial Cells/metabolism , Receptor, IGF Type 1/genetics , STAT5 Transcription Factor/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Animals , Gene Expression Profiling , Islets of Langerhans Transplantation , Kidney Glomerulus , Oligonucleotide Array Sequence Analysis , Rats , Suppressor of Cytokine Signaling Proteins/drug effects
10.
BMC Cancer ; 8: 150, 2008 May 28.
Article in English | MEDLINE | ID: mdl-18507841

ABSTRACT

BACKGROUND: Suppressor of cytokine signaling 3 (SOCS3) is considered to inhibit cytokine responses and play a negative role in migration of various cells. Proline-rich tyrosine kinase 2 (PYK2) is a non-receptor kinase and has been found crucial to cell motility. However, little is known about whether SOCS3 could regulate PYK2 pro-migratory function in lung cancer. METHODS: The methylation status of SOCS3 was investigated in HBE and A549 cell lines by methylation-specific PCR. A549 cells were either treated with a demethylation agent 5-aza-2'-deoxycytidine or transfected with three SOCS3 mutants with various functional domains deleted. Besides, cells were pretreated with a proteasome inhibitor beta-lactacystin where indicated. The effects of SOCS3 up-regulation on PYK2 expression, PYK2 and ERK1/2 phosphorylations were assessed by western blot using indicated antibodies. RT-PCR was used to estimate PYK2 mRNA levels. Transwell experiments were performed to evaluate cell migration. RESULTS: SOCS3 expression was found impaired in A549 cells and higher PYK2 activity was correlated with enhanced cell migration. We identified that SOCS3 was aberrantly methylated in the exon 2, and 5-aza-2'-deoxycytidine restored SOCS3 expression. Reactivation of SOCS3 attenuated PYK2 expression and phosphorylation, cell migration was inhibited as well. Transfection studies indicated that exogenous SOCS3 interacted with PYK2, and both the Src homology 2 (SH2) and the kinase inhibitory region (KIR) domains of SOCS3 contributed to PYK2 binding. Furthermore, SOCS3 was found to inhibit PYK2-associated ERK1/2 activity in A549 cells. SOCS3 possibly promoted degradation of PYK2 in a SOCS-box-dependent manner and interfered with PYK2-related signaling events, such as cell migration. CONCLUSION: These data indicate that SOCS3 negatively regulates cell motility and decreased SOCS3 induced by methylation may confer a migration advantage to A549 cells. These results also suggest a negative role of SOCS3 in PYK2 signaling, and a previously unidentified regulatory mechanism for PYK2 function.


Subject(s)
Cell Movement/physiology , DNA Methylation , Focal Adhesion Kinase 2/metabolism , Signal Transduction/physiology , Suppressor of Cytokine Signaling Proteins/metabolism , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Blotting, Western , Cell Movement/drug effects , Deoxycytidine/pharmacology , Down-Regulation , Focal Adhesion Kinase 2/drug effects , Humans , In Vitro Techniques , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Phosphorylation , Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Sensitivity and Specificity , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Tumor Cells, Cultured , Up-Regulation
11.
Alcohol Clin Exp Res ; 32(9): 1565-73, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18616672

ABSTRACT

BACKGROUND: Acute alcohol consumption is associated with induction of immuno-inhibitory cytokines and down-regulation of pro-inflammatory responses to various pathogens. We previously reported that alcohol activates janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling leading to IL-10 induction. The JAK-STAT pathway also activates its own negative regulators, suppressors of cytokine signaling (SOCS) 1 and SOCS3. SOCS proteins are inducible inhibitors that negatively regulate STAT3/STAT1 signaling pathways induced by cytokines, IL-6 or IFNs. Here we aimed to explore the effect of acute alcohol on induction of SOCS1/SOCS3 and regulation of STAT3/STAT1 pathways induced by IL-6 or IFNs in human monocytes. METHODS: Blood samples from normal volunteers were collected before and 24 hours after consumption of 2 ml vodka/kg body weight. For in vitro experiments human monocytes were pretreated with ethanol (EtOH) followed by stimulation with cytokines; proteins were analyzed by Western blot, nuclear protein binding to DNA by EMSA, and RNA by real time PCR. RESULTS: Acute in vivo or in vitro alcohol treatment increased both SOCS1 and SOCS3 RNA expression in monocytes. Alcohol treatment resulted in increased STAT3 and STAT1 DNA binding capacity. Activation of both STAT1 and STAT3 has been shown to induce SOCS1/3. We hypothesized that induction of SOCS proteins by alcohol in turn may lead to modulation of cytokine signaling through STAT1 and STAT3. Indeed, we observed significant down-regulation of IL-6-, IFNalpha- and IFNgamma-induced STAT1 DNA binding as well as inhibition of IL-6- and IFNgamma-induced STAT3 when alcohol was added to monocytes 3 hours prior to the cytokine stimulation. Consistent with inhibition of IL-6-induced STAT3 DNA binding in alcohol-pretreated cells, the levels of IL-6-dependent genes, MCP-1 and ICAM-1, was reduced after IL-6 stimulation. Similar to EtOH alone, combined EtOH+IL-6 simulation resulted in increased expression of both SOCS3 and SOCS1 genes. CONCLUSION: While acute alcohol treatment alone activates STAT1/3 signaling pathways and induces SOCS3 and SOCS1 levels in monocytes, alcohol also leads to down-regulation of IL-6-, IFNalpha-, and IFNgamma-induced signaling via STAT1/STAT3 pathways, likely through excessive SOCS activation.


Subject(s)
Ethanol/pharmacology , Monocytes/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Suppressor of Cytokine Signaling Proteins/metabolism , Case-Control Studies , Central Nervous System Depressants/pharmacology , Chemokine CCL2/metabolism , DNA/metabolism , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Female , Humans , Intercellular Adhesion Molecule-1/metabolism , Interferon-alpha/pharmacology , Interferon-gamma/pharmacology , Interleukin-6/pharmacology , Male , Monocytes/cytology , Monocytes/drug effects , RNA, Messenger/metabolism , STAT1 Transcription Factor/drug effects , STAT3 Transcription Factor/drug effects , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/drug effects
12.
Exp Hematol ; 35(3): 454-464, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17309826

ABSTRACT

OBJECTIVE: Interleukin 15 (IL)-15 controls proliferation and survival of T cells, but its effects and the underlying cellular regulation are not well understood. Previous studies have focused on its effects on short-term T-cell cultures. In view of the potential problems associated with using IL-2 alone in adoptive immunotherapy protocols, we investigated the impact of IL-15 on T-cell cultures and the global transcriptional effects it elicits in such cultures. MATERIALS AND METHODS: DNA microarrays and flow cytometry were used to examine the differential effect of 20 ng/mL IL-15 on primary serum-free T-cell cultures activated and cultured in the presence of IL-2. Quantitative reverse transcriptase polymerase chain reaction confirmed select microarray data. RESULTS: IL-15 significantly increased ex vivo expansion of primary human T cells over the entire 11-day expansions without affecting viability. The 1133 genes were consistently differentially expressed among three donor samples. Ontological analysis demonstrated that IL-15 increases expression of genes involved in inflammatory response (e.g., tumor necrosis factor [TNF]-alpha, Oncostatin M, CD40L, and CD33) and apoptosis (e.g., TNF-related apoptosis-inducing ligand). IL-15 also induced expression of four suppressors of cytokine signaling (SOCS) family genes (SOCS1-3, cytokine-inducible SH2-containing protein), which are classical negative regulators of cytokine signaling. IL-15 strongly suppressed the expression of inhibitory natural killer cell receptor genes, including three C-type lectins (KLRB1, KLRC1, and KLRD1), as well as IL-7Ra and Granzyme H. Finally, IL-15 induced differential expression of TNF receptor superfamily members (CD27 and CD30). CONCLUSION: These findings suggest that exogenous IL-15 may have a potential role in adoptive immunotherapy by both enhancing proliferation and modulating functionality during ex vivo T-cell expansion.


Subject(s)
Gene Expression Profiling , Inflammation/immunology , Interleukin-15/immunology , Suppressor of Cytokine Signaling Proteins/genetics , T-Lymphocytes/immunology , Antigens, CD/drug effects , Antigens, CD/genetics , Apoptosis/immunology , Cell Proliferation/drug effects , Cells, Cultured , Cluster Analysis , Flow Cytometry/methods , Granzymes/drug effects , Granzymes/genetics , Humans , Interleukin-15/pharmacology , Interleukin-2/pharmacology , Oligonucleotide Array Sequence Analysis , Receptors, Immunologic/drug effects , Receptors, Immunologic/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction/drug effects , Signal Transduction/immunology , Suppressor of Cytokine Signaling Proteins/drug effects , Suppressor of Cytokine Signaling Proteins/immunology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , Transcription, Genetic
13.
J Leukoc Biol ; 80(5): 1136-44, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16943387

ABSTRACT

Mycobacterium avium is an opportunistic pathogen that commonly infects individuals colonized with HIV-1, although it is less frequent in the post-HAART era. These microorganisms invade macrophages after interacting with TLR2 and/or CD14 co-receptors, but signaling pathways promoting survival in macrophages are not well defined. Although IFN-gamma plays an important role in protective immunity against bacterial infections, IFN-gamma responses are compromised in AIDS patients and evidence suggests that exogenous IFN-gamma is inadequate to clear the mycobacteria. To determine the mechanism by which M. avium survives intracellularly, even in the presence of IFN-gamma, we studied the effect of mycobacteria infection in macrophages during early IFN-gamma signaling events. M. avium infected cells exhibited a reduced response to IFN-gamma, with suppressed phosphorylation of STAT-1 compared with uninfected cells. Interaction of M. avium with macrophage receptors increased gene expression of the suppressors of cytokine signaling (SOCS) to diminish IFN responsiveness. Specifically, we observed an increase in mRNA for both SOCS-3 and SOCS-1, which correlates with elevated levels of SOCS protein and positive immunostaining in M. avium/HIV-1 co-infected tissues. We also linked the p38 MAPK signaling pathway to mycobacterial-induced SOCS gene transcription. The induction of SOCS may be part of the strategy that allows the invader to render the macrophages unresponsive to IFN-gamma, which otherwise promotes clearance of the infection. Our data provide new insights into the manipulation of the host response by this opportunistic pathogen and the potential for modulating SOCS to influence the outcome of M. avium infection in immunocompromised hosts.


Subject(s)
Interferon-gamma/immunology , Macrophages/immunology , Mycobacterium avium/immunology , Suppressor of Cytokine Signaling Proteins/immunology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Expression Regulation/immunology , Humans , Imidazoles/pharmacology , Interferon-gamma/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/microbiology , Mycobacterium avium/drug effects , Pyridines/pharmacology , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Signal Transduction/drug effects , Signal Transduction/immunology , Structure-Activity Relationship , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Suppressor of Cytokine Signaling Proteins/genetics , Transcription, Genetic , Up-Regulation/drug effects
14.
Arch Dermatol Res ; 298(4): 163-73, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16845523

ABSTRACT

Bacterial stimulation may serve to control atopic disorders such as atopic dermatitis (AD) through inducement of Th1 cell-mediated immune response. The lipoteichoic acid (LTA)-related molecule (okLTA) from streptococcal preparation, OK-432, has been shown to be a potent Th1 inducer through the action of IL-12. Examination was made of the therapeutic effects of this okLTA injected intra- and/or subcutaneously into AD-like lesions in NC/Nga mice, particularly in the vicinity of the suppressor of cytokine signaling (SOCS) regulatory pathways. Using immunohistochemical staining with IL-4/IL-12p40 and phosphorylated STAT6/p-STAT4 and RT-PCR for IL-4/IL-12p40, STAT6/STAT4 and mRNA expression and in situ hybridization of SOCS3 and 5, evaluation was made of the immunoregulatory effects of this okLTA in the treatment of spontaneous AD-like lesions in NC/Nga mice. Following the injection of okLTA, remarkable improvement in the lesions of NC/Nga mice was noted. In okLTA-treated skin, IL-12p40/p-STAT4 positive cellular infiltration was extensive while IL-4/p-STAT6 positive cell infiltration was seen to diminish considerably, compared to untreated NC mice. SOCS3 in situ expression in okLTA-treated mice was noted to be significantly less compared to untreated NC mice, in which the expression was prominent. SOCS5 in situ expression was rather, though not significantly, strong in okLTA-treated mice. okLTA treatment is clearly shown to induce Th1 cellular response and down-regulate immune response in the Th2 pathway through SOCS3 reduction in AD-like lesions of NC/Nga mice. The present results demonstrate that bacterial wall components such as okLTA should serve as an effective new therapeutic approach for treating AD.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dermatitis, Atopic/drug therapy , Lipopolysaccharides/pharmacology , Picibanil/pharmacology , STAT6 Transcription Factor/drug effects , Teichoic Acids/pharmacology , Th1 Cells/drug effects , Th2 Cells/drug effects , Animals , Dermatitis, Atopic/pathology , Down-Regulation/drug effects , Immunohistochemistry , In Situ Hybridization , Lipopolysaccharides/chemistry , Mice , Mice, Inbred BALB C , Models, Animal , Monocytes/metabolism , Picibanil/chemistry , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Teichoic Acids/chemistry , Th1 Cells/immunology , Th2 Cells/immunology , Up-Regulation/drug effects
15.
J Dairy Sci ; 89(12): 4640-8, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17106096

ABSTRACT

Frequent milking of dairy cows during early lactation results in a persistent increase in milk yield; however, the mechanism underlying this effect is unknown. We hypothesized that increased exposure of the mammary gland to prolactin (PRL) mediates the milk yield response. Fifteen multiparous Holstein cows were assigned to 3 treatments for the first 3 wk of lactation: twice daily milking with (2x + PRL) or without (2x) supplemental exogenous PRL, or 4 times daily milking (4x). Mammary biopsies were obtained at 7 DIM, and rates of [(3)H]thymidine incorporation into DNA in vitro were determined. Mammary expression of suppressors of cytokine signaling (SOCS)-1, -2, and -3; the long form of PRL-receptor; and alpha-lactalbumin mRNA was measured by real-time reverse-transcription PCR. Incorporation of [(3)H]thymidine into DNA was not affected by frequent milking or PRL treatment; however, analysis of autoradiograms revealed that stromal cell proliferation was greater in 4x cows. Mammary expression of SOCS-1 was not affected by milking frequency or PRL treatment. Expression of SOCS-2 mRNA was increased with frequent milking or PRL treatment, whereas expression of SOCS-3 mRNA was reduced by frequent milking or exogenous PRL. Abundance of PRL-receptor mRNA was reduced, whereas alpha-lactalbumin mRNA was increased with PRL treatment. These results demonstrate that the bovine mammary gland is responsive to exogenous PRL during early lactation. In addition, differences in the response to frequent milking or exogenous PRL during early lactation indicate distinct effects of PRL and milk removal on the mammary function of dairy cows.


Subject(s)
Cattle/physiology , Dairying/methods , Lactation/physiology , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/physiology , Prolactin/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Biopsy , Cell Proliferation/drug effects , Female , Gene Expression/drug effects , Gene Expression/physiology , Lactation/drug effects , Prolactin/administration & dosage , Prolactin/blood , Prolactin/physiology , Radioisotopes/analysis , Random Allocation , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Suppressor of Cytokine Signaling Proteins/drug effects , Suppressor of Cytokine Signaling Proteins/physiology , Thymidine/analysis , Thymidine/metabolism , Time Factors , Tritium/analysis
16.
J Drug Target ; 23(1): 59-66, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25185784

ABSTRACT

BACKGROUND: miR-155 acts as a ubiquitous oncogene in major classes of human cancers and is a potential target for therapeutic intervention. However, the role of miR-155 in multiple myeloma is poorly understood. METHODS: To explore the role of miR-155 in multiple myeloma, we assessed the influence of tiny seed-targeting anti-miR-155 (t-anti-miR-155) on multiple myeloma cell line (RPMI-8266) viability and apoptosis in vitro. RESULTS: t-anti-miR-155 significantly inhibited multiple myeloma cell proliferation, migration, and colony formation. Additionally, t-anti-miR-155 significantly increased CD19 positive cell numbers, which are novel biomarkers for multiple myeloma and suppressor of cytokine signaling 1(SOCS1) was shown to be a target gene for miR-155 in multiple myeloma. Finally, the miR-155 signaling pathway was investigated by KEGG assay. CONCLUSION: miR-155 in RPMI-8266 cells is a critical oncomiR in multiple myeloma and seed-targeting t-anti-miR-155 might be a novel strategy for miR-155-based therapeutics.


Subject(s)
Disease Progression , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/antagonists & inhibitors , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Oligonucleotides, Antisense/pharmacology , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Migration Assays , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , MicroRNAs/biosynthesis , MicroRNAs/genetics , Multiple Myeloma/drug therapy , Signal Transduction/drug effects , Signal Transduction/genetics , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/biosynthesis , Suppressor of Cytokine Signaling Proteins/drug effects , Tumor Stem Cell Assay
17.
Eur J Endocrinol ; 173(3): 283-90, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26034073

ABSTRACT

OBJECTIVE: Fasting and exercise stimulates, whereas glucose suppresses GH secretion, but it is uncertain how these conditions impact GH signaling in peripheral tissues. To test the original 'feast and famine hypothesis' by Rabinowitz and Zierler, according to which the metabolic effects of GH are predominant during fasting, we specifically hypothesized that fasting and exercise act in synergy to increase STAT-5b target gene expression. DESIGN AND METHODS: Eight healthy men were studied on two occasions in relation to a 1 h exercise bout: i) with a concomitant i.v. glucose infusion ('feast') and ii) after a 36 h fast ('famine'). Muscle and fat biopsy specimens were obtained before, immediately after, and 30 min after exercise. RESULTS: GH increased during exercise on both examination days and this effect was amplified by fasting, and free fatty acid (FFA) levels increased after fasting. STAT-5b phosphorylation increased similarly following exercise on both occasions. In adipose tissue, suppressors of cytokine signaling 1 (SOCS1) and SOCS2 were increased after exercise on the fasting day and both fasting and exercise increased cytokine inducible SH2-containing protein (CISH). In muscle, SOCS2 and CISH mRNA were persistently increased after fasting. Muscle SOCS1, SOCS3, and CISH mRNA expression increased, whereas SOCS2 decreased after exercise on both examination days. CONCLUSIONS: This study demonstrates that fasting and exercise act in tandem to amplify STAT-5b target gene expression (SOCS and CISH) in adipose and muscle tissue in accordance with the 'feast and famine hypothesis'; the adipose tissue signaling responses, which hitherto have not been scrutinized, may play a particular role in promoting FFA mobilization.


Subject(s)
Adipose Tissue/metabolism , Exercise/physiology , Fasting/metabolism , Glucose/pharmacology , Human Growth Hormone/metabolism , Muscle, Skeletal/metabolism , RNA, Messenger/metabolism , Sweetening Agents/pharmacology , Adipose Tissue/drug effects , Adult , Fatty Acids, Nonesterified/metabolism , Human Growth Hormone/drug effects , Humans , Male , Muscle, Skeletal/drug effects , Phosphorylation/drug effects , Phosphorylation/genetics , Phosphorylation/physiology , RNA, Messenger/drug effects , STAT5 Transcription Factor/drug effects , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Young Adult
18.
Clinics (Sao Paulo) ; 69(1): 68-74, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24473562

ABSTRACT

OBJECTIVE: It has been shown that SOCS-1 plays an important role in the proper control of cytokine/growth factor responses and acts as a tumor suppressor in acute myeloid leukemias. Therefore, the objective of the present study was to evaluate the in vitro effect of treatment with Nutlin-3, a small molecule inhibitor of the MDM2/p53 interaction, on the expression of the suppressor of cytokine signaling 1 in primary acute myeloid leukemia cells and in myeloid cell lines with differential p53 status. METHOD: The expression of the suppressor of cytokine signaling 1 was quantitatively analyzed by real-time PCR in myeloid p53wild-type (OCI and MOLM) and p53null HL-60, leukemic cell lines, in patient-derived acute myeloid leukemia blasts, and in primary normal cell types, such as macrophages, endothelial cells, and bone marrow mesenchymal stem cells. The p53-dependence of the suppressor of cytokine signaling 1 upregulation that is induced by Nutlin-3 was analyzed in experiments performed using siRNA for p53, while the functional upregulation of the suppressor of cytokine signaling 1 was analyzed by assessing the levels of phosphorylated STAT-3. RESULTS: Nutlin-3 significantly upregulated the transcription of the suppressor of cytokine signaling 1 in p53wild-type OCI and MOLM but not in p53deleted p53null HL60, myeloid leukemic cell lines, as well as in primary acute myeloid leukemia blasts. Conversely, and somewhat unexpectedly, Nutlin-3 did not modulate the suppressor of cytokine signaling 1 expression in primary normal macrophages, endothelial cells, and bone marrow mesenchymal stem cells. The p53-dependent upregulation of the suppressor of cytokine signaling 1 by Nutlin-3 was associated with the downregulation of phosphorylated STAT-3, a major molecular target of the suppressor of cytokine signaling 1. CONCLUSION: Overall, our data suggest a potential role for the suppressor of cytokine signaling 1 as a therapeutic target of Nutlin-3 in p53 wild-type acute myeloid leukemias.


Subject(s)
Imidazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Piperazines/pharmacology , Suppressor of Cytokine Signaling Proteins/drug effects , Up-Regulation/drug effects , Blotting, Western , Cell Line, Tumor , Cell Survival , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Humans , Imidazoles/metabolism , Leukemia, Myeloid, Acute/metabolism , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Piperazines/metabolism , Real-Time Polymerase Chain Reaction , Reproducibility of Results , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Time Factors , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism
19.
Clinics (Sao Paulo) ; 68(10): 1325-32, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24212839

ABSTRACT

OBJECTIVES: Suppressor of cytokine signaling 3, myxovirus resistance protein and osteopontin gene polymorphisms may influence the therapeutic response in patients with chronic hepatitis C, and an association with IL28 might increase the power to predict sustained virologic response. Our aims were to evaluate the association between myxovirus resistance protein, osteopontin and suppressor of cytokine signaling 3 gene polymorphisms in combination with IL28B and to assess the therapy response in hepatitis C patients treated with pegylated-interferon plus ribavirin. METHOD: Myxovirus resistance protein, osteopontin, suppressor of cytokine signaling 3 and IL28B polymorphisms were analyzed by PCR-restriction fragment length polymorphism, direct sequencing and real-time PCR. Ancestry was determined using genetic markers. RESULTS: We analyzed 181 individuals, including 52 who were sustained virologic responders. The protective genotype frequencies among the sustained virologic response group were as follows: the G/G suppressor of cytokine signaling 3 (rs4969170) (62.2%); T/T osteopontin (rs2853744) (60%); T/T osteopontin (rs11730582) (64.3%); and the G/T myxovirus resistance protein (rs2071430) genotype (54%). The patients who had ≥3 of the protective genotypes from the myxovirus resistance protein, the suppressor of cytokine signaling 3 and osteopontin had a greater than 90% probability of achieving a sustained response (p<0.0001). The C/C IL28B genotype was present in 58.8% of the subjects in this group. The sustained virological response rates increased to 85.7% and 91.7% by analyzing C/C IL28B with the T/T osteopontin genotype at rs11730582 and the G/G suppressor of cytokine signaling 3 genotype, respectively. Genetic ancestry analysis revealed an admixed population. CONCLUSION: Hepatitis C genotype 1 patients who were responders to interferon-based therapy had a high frequency of multiple protective polymorphisms in the myxovirus resistance protein, osteopontin and suppressor of cytokine signaling 3 genes. The combined analysis of the suppressor of cytokine signaling 3 and IL28B genotypes more effectively predicted sustained virologic response than IL28B analysis alone.


Subject(s)
Hepatitis C, Chronic/drug therapy , Interleukins/genetics , Myxovirus Resistance Proteins/genetics , Osteopontin/genetics , Polymorphism, Genetic/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Adult , Antiviral Agents/therapeutic use , Female , Gene Frequency , Genetic Markers , Genotype , Hepacivirus/drug effects , Humans , Interferon alpha-2 , Interferon-alpha/therapeutic use , Interferons , Male , Middle Aged , Myxovirus Resistance Proteins/drug effects , Osteopontin/drug effects , Polyethylene Glycols/therapeutic use , Predictive Value of Tests , Real-Time Polymerase Chain Reaction , Recombinant Proteins/therapeutic use , Ribavirin/therapeutic use , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/drug effects , Treatment Outcome
20.
Curr Diabetes Rev ; 9(6): 472-7, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24111619

ABSTRACT

UNLABELLED: It has been known that chronic liver treatments interfere with blood glucose metabolism. It was recognized that diabetes mellitus among chronic hepatitis C was greater in other types of chronic liver diseases. Hepatitis C directly promotes insulin resistance through the proteosomal degradation of insulin resistance substrate. It suppressed hepatocyte glucose uptake through down-regulation of surface expression of glucose transporter. Long-term exposure to cytokine over expression seems to be cytotoxic to both beta cells of the pancreas and to hepatocytes. Elevated tumor necrosis factor-a, or its neutralization, increased insulin sensitivity. Interferon-a may also elevate the serum level of interleukin-1 which is cytotoxic to pancreatic islet cells. Both diabetes mellitus and resistance to interferon-a therapy are abnormally mediated by over-expression of suppressor of cytokine signaling-1 in hepatocytes of chronic hepatitis C patients. CONCLUSION: These data suggest that interferon-a therapy should be administered with caution in patients showing any predisposition to Diabetes mellitus. Anti inflammatory therapy is critically recommended as a protector against disease development due to cytokine mediated Diabetes mellitus during hepatitis C therapy, since inflammation seems to be a main candidate to interferon suspected diabetogenesis.


Subject(s)
Antiviral Agents/adverse effects , Diabetes Mellitus, Type 2/immunology , Hepatitis C, Chronic/immunology , Inflammation/immunology , Interferon-alpha/adverse effects , Antiviral Agents/administration & dosage , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/physiopathology , Down-Regulation , Female , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/physiopathology , Humans , Inflammation/chemically induced , Inflammation/physiopathology , Insulin Resistance , Interferon-alpha/administration & dosage , Interleukin-1/blood , Interleukin-1/metabolism , Male , Patient Selection , Signal Transduction , Suppressor of Cytokine Signaling Proteins/biosynthesis , Suppressor of Cytokine Signaling Proteins/drug effects , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL