Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Commun Biol ; 4(1): 775, 2021 06 23.
Article in English | MEDLINE | ID: mdl-34163008

ABSTRACT

Patients with long-standing diabetes have a high risk for cardiac complications that is exacerbated by increased reactive oxygen species (ROS) production. We found that feeding cyanocobalamin (B12), a scavenger of superoxide, not only prevented but reversed signs of cardiomyopathy in type 1 diabetic Elmo1H/H Ins2Akita/+ mice. ROS reductions in plasma and hearts were comparable to those in mice treated with other antioxidants, N-acetyl-L-cysteine or tempol, but B12 produced better cardioprotective effects. Diabetes markedly decreased plasma insulin-like growth factor (IGF)-1 levels, while B12, but not N-acetyl-L-cysteine nor tempol, restored them. B12 activated hepatic IGF-1 production via normalization of S-adenosylmethionine levels, DNA methyltransferase (DNMT)-1/3a/3b mRNA, and DNA methylation of promoters for suppressor of cytokine signaling (SOCS)-1/3. Reductions of cardiac IGF-1 mRNA and phosphorylated IGF-1 receptors were also restored. Thus, B12 is a promising option for preventing diabetic cardiomyopathy via ROS reduction and IGF-1 retrieval through DNMT-SOCS1/3 signaling.


Subject(s)
Cardiomyopathies/prevention & control , DNA-Cytosine Methylases/physiology , Diabetes Mellitus, Type 1/complications , Insulin-Like Growth Factor I/physiology , Oxidative Stress/drug effects , Suppressor of Cytokine Signaling 1 Protein/physiology , Suppressor of Cytokine Signaling 3 Protein/physiology , Vitamin B 12/pharmacology , Adaptor Proteins, Signal Transducing/physiology , Animals , Diabetes Mellitus, Type 1/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects
2.
J Diabetes ; 13(9): 701-712, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33576141

ABSTRACT

BACKGROUND: Previous studies reported that testosterone and DNA methylation of suppressor of cytokine signaling-3 (SOCS3) were associated with type 2 diabetes (T2D). Testosterone affects SOCS3 gene expression. Therefore, we aimed to investigate how the SOCS3 methylation mediates the relationship between testosterone and T2D among Chinese rural adults. METHODS: A case-control study comprised 365 T2D patients and 651 controls was conducted. Liquid chromatography-tandem mass spectrometry and MethylTarget were used to determine the levels of serum testosterone and DNA methylation of SOCS3 gene, respectively. The odds ratio (OR) of testosterone or SOCS3 methylation for T2D was calculated using logistic regression models, and ß value of testosterone for SOCS3 methylation was evaluated by linear regression models. Furthermore, through mediation analysis the mediating effect of SOCS3 methylation on the association of testosterone with T2D was estimated. RESULTS: After adjusting for multiple variables, the protective effect of testosterone on T2D was found in men (OR = 0.61, 95% confidence interval [CI]: 0.47-0.80), and the methylation of Chr17:76356190 or Chr17:76356199 was negatively related to T2D in both men and women. Moreover, testosterone was positively associated with Chr17:76356190 methylation in men and Chr17:76356199 methylation in women (both P < .05). The mediation analysis showed that the Chr17:76356190 methylation partly mediated effect of testosterone on T2D in men was approximately 8.2%. CONCLUSIONS: High levels of serum testosterone in men and Chr17:76356190 and Chr17:76356199 (SOCS3) methylation were related to a lower prevalent T2D. In addition, Chr17:76356190 methylation partially mediated the effect of testosterone on T2D in Chinese rural men.


Subject(s)
DNA Methylation , Diabetes Mellitus, Type 2/blood , Suppressor of Cytokine Signaling 3 Protein/physiology , Testosterone/blood , Aged , China , Chromatography, Liquid/methods , Female , Humans , Male , Middle Aged , Rural Population , Tandem Mass Spectrometry/methods
3.
Neurosci Bull ; 36(7): 778-792, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32306216

ABSTRACT

SOCS3, a feedback inhibitor of the JAK/STAT signal pathway, negatively regulates axonal regrowth and inflammation in the central nervous system (CNS). Here, we demonstrated a distinct role of SOCS3 in the injured spinal cord of the gecko following tail amputation. Severing the gecko spinal cord did not evoke an inflammatory cascade except for an injury-stimulated elevation of the granulocyte/macrophage colony-stimulating factor (GM-CSF) and interferon gamma (IFN-γ) cytokines. Simultaneously, the expression of SOCS3 was upregulated in microglia, and unexpectedly not in neurons. Enforced expression of SOCS3 was sufficient to suppress the GM-CSF/IFN-γ-driven inflammatory responses through its KIR domain by attenuating the activities of JAK1 and JAK2. SOCS3 was also linked to GM-CSF/IFN-γ-induced cross-tolerance. Transfection of adenovirus overexpressing SOCS3 in the injured cord resulted in a significant decrease of inflammatory cytokines. These results reveal a distinct role of SOCS3 in the regenerating spinal cord, and provide new hints for CNS repair in mammals.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor , Interferon-gamma , Spinal Cord Regeneration , Suppressor of Cytokine Signaling 3 Protein/physiology , Animals , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Inflammation , Interferon-gamma/physiology , Lizards , Microglia , Neurons
4.
Eur Rev Med Pharmacol Sci ; 23(21): 9286-9294, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31773696

ABSTRACT

OBJECTIVE: Cytokine signal transduction inhibitor 3 (SOCS3) negatively regulates Janus kinases (JAK) - signal transducer and activator of transcription (STAT) pathway. Bioinformatics analysis revealed a targeted relationship between miR-203 and SOCS3 mRNA. This study investigated the role of miR-203 in ovarian cancer cell proliferation and apoptosis. PATIENTS AND METHODS: Ovarian cancer tissues and adjacent tissues were collected to detect the expression of miR-203 and SOCS3. Ovarian cancer HO8910 cells were divided into miR-NC group, miR-203 inhibitor group, and miR-203 mimic group followed by the analysis of the expression of miR-203 and SOCS3 mRNA by quantitative Reverse Transcription-PCR (qRT-PCR), protein expression of p-JAK2 and p-STAT3 by Western blot, cell apoptosis by flow cytometry, and proliferation by 5-Ethynyl-2'-deoxyuridine (EdU) staining chronologically. RESULTS: Compared with adjacent tissues, miR-203 expression was significantly increased in tumor tissues and SCOS3 mRNA expression was decreased. Compared with those with lower miR-203 expression, the prognosis of patients with higher expression of miR-203 was significantly worse. There was a targeted regulatory relationship between miR-203 and SOCS3 mRNA. Compared with IOSE80 cells, miR-203 expression in HO8910 and SKOV3 cells was increased, and its expressions of SOCS3 mRNA and protein were decreased. Compared with miR-NC group, the transfection of miR-203 inhibitor significantly increased SOCS3 expression, and decreased the expression of p-JAK2 and p-STAT3 protein. We draw the conclusion that miR-203 increased cell apoptosis and decreased cell proliferation. However, opposite results were observed after the transfection of miR-203 mimic. CONCLUSIONS: Abnormal miR-203 and SOCS3 expression are related to the pathogenesis of ovarian cancer. MiR-203 affects the proliferation of JAK-STAT pathway and regulates the proliferation and apoptosis of ovarian cancer cells by targeting the inhibition of SOCS3 expression.


Subject(s)
Apoptosis/physiology , Carcinoma, Ovarian Epithelial/physiopathology , Cell Proliferation/physiology , MicroRNAs/physiology , Ovarian Neoplasms/physiopathology , Suppressor of Cytokine Signaling 3 Protein/physiology , Carcinoma, Ovarian Epithelial/metabolism , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Janus Kinase 2/biosynthesis , MicroRNAs/agonists , MicroRNAs/antagonists & inhibitors , MicroRNAs/biosynthesis , Molecular Mimicry/physiology , Ovarian Neoplasms/metabolism , STAT3 Transcription Factor/biosynthesis , Suppressor of Cytokine Signaling 3 Protein/biosynthesis , Transfection
5.
Immunity ; 50(4): 1007-1023, 2019 04 16.
Article in English | MEDLINE | ID: mdl-30995492

ABSTRACT

Interleukin-6 (IL-6) is a pleiotropic cytokine with roles in immunity, tissue regeneration, and metabolism. Rapid production of IL-6 contributes to host defense during infection and tissue injury, but excessive synthesis of IL-6 and dysregulation of IL-6 receptor signaling is involved in disease pathology. Therapeutic agents targeting the IL-6 axis are effective in rheumatoid arthritis, and applications are being extended to other settings of acute and chronic inflammation. Recent studies reveal that selective blockade of different modes of IL-6 receptor signaling has different outcomes on disease pathology, suggesting novel strategies for therapeutic intervention. However, some inflammatory diseases do not seem to respond to IL-6 blockade. Here, we review the current state of IL-6-targeting approaches in the clinic and discuss how to apply the growing understanding of the immunobiology of IL-6 to clinical decisions.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Inflammation/drug therapy , Interleukin-6/antagonists & inhibitors , Molecular Targeted Therapy , Signal Transduction/drug effects , Animals , Antibodies, Monoclonal/immunology , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/immunology , Clinical Trials as Topic , Cytokine Receptor gp130/antagonists & inhibitors , Cytokine Receptor gp130/immunology , Humans , Inflammation/immunology , Interleukin-6/biosynthesis , Interleukin-6/deficiency , Interleukin-6/immunology , Janus Kinases/antagonists & inhibitors , Janus Kinases/physiology , MAP Kinase Signaling System/drug effects , Mice , Mice, Knockout , Receptors, Interleukin-6/immunology , Ribonucleases/deficiency , STAT3 Transcription Factor/physiology , Suppressor of Cytokine Signaling 1 Protein/physiology , Suppressor of Cytokine Signaling 3 Protein/physiology
6.
Am J Physiol Regul Integr Comp Physiol ; 316(4): R338-R351, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30673296

ABSTRACT

Suppressor of cytokine signaling 3 (SOCS3) is a negative regulator of leptin signaling. We previously showed that the chronic effects of leptin on blood pressure (BP) and glucose regulation are mediated by stimulation of proopiomelanocortin (POMC) neurons. In this study we examined the importance of endogenous SOCS3 in POMC neurons in control of metabolic and cardiovascular function and potential sex differences. Male and female SOCS3flox/flox/POMC-Cre mice in which SOCS3 was selectively deleted in POMC neurons and control SOCS3flox/flox mice were studied during a control diet (CD) or a high-fat diet (HFD) and during chronic leptin infusion. Body weight was lower in male and female SOCS3flox/flox/POMC-Cre than control mice fed the CD, despite similar food intake. Male SOCS3flox/flox/POMC-Cre mice exhibited increased energy expenditure. BP and heart rate were similar in male and female SOCS3flox/flox/POMC-Cre and control mice fed the CD. HFD-fed male and female SOCS3flox/flox/POMC-Cre mice showed attenuated weight gain. HFD-induced elevations in baseline BP and BP responses to an air-jet stress test were greater in female SOCS3flox/flox/POMC-Cre than control mice. Chronic leptin infusion produced similar responses for food intake, body weight, oxygen consumption, blood glucose, BP, and heart rate in all groups. Thus SOCS3 deficiency in POMC neurons influences body weight regulation in the setting of CD and HFD and differentially affects BP and energy balance in a sex-specific manner but does not amplify the dietary, glycemic, or cardiovascular effects of leptin.


Subject(s)
Cardiovascular Physiological Phenomena , Neurons/physiology , Pro-Opiomelanocortin/physiology , Suppressor of Cytokine Signaling 3 Protein/physiology , Animals , Animals, Genetically Modified , Diet , Diet, High-Fat , Eating , Female , Leptin/pharmacology , Male , Mice , Mice, Knockout , Neurons/metabolism , Oxygen Consumption/genetics , Suppressor of Cytokine Signaling 3 Protein/deficiency , Suppressor of Cytokine Signaling 3 Protein/genetics , Weight Gain/genetics
7.
Hormones (Athens) ; 18(2): 127-136, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30414080

ABSTRACT

The suppressors of cytokine signaling (SOCS) are a group of eight proteins responsible for preventing excessive cytokine signaling. Among this protein family, SOCS3 has received special attention. SOCS3 expression is important to control certain allergy autoimmune diseases. Furthermore, SOCS3 expression is elevated in obesity and it is involved in the inhibition of leptin and insulin signaling, two important hormones involved in the control of energy metabolism. Therefore, increased SOCS3 expression in obese individuals is associated with several metabolic disorders, including reduced energy expenditure, increased food intake and adiposity, and insulin and leptin resistance. In addition, recent studies found that SOCS3 expression regulates energy and glucose homeostasis in several metabolic conditions, such as pregnancy, caloric restriction, and refeeding. Importantly, attenuation of SOCS3 expression in most cases improves leptin and insulin sensitivity, leading to beneficial metabolic effects. This review aims to discuss the role of SOCS3 in the control of blood glucose levels as well as in energy homeostasis. The development of pharmacological compounds to inhibit SOCS3 activity and/or expression may represent a promising therapeutic approach to treat type 2 diabetes mellitus, obesity, and other metabolic imbalances.


Subject(s)
Metabolic Diseases/drug therapy , Molecular Targeted Therapy/trends , Suppressor of Cytokine Signaling 3 Protein/antagonists & inhibitors , Suppressor of Cytokine Signaling 3 Protein/physiology , Diabetes Mellitus, Type 2/drug therapy , Female , Humans , Molecular Targeted Therapy/methods , Obesity/drug therapy , Pregnancy , Pregnancy Complications/drug therapy
8.
Brain Behav Immun ; 68: 111-122, 2018 02.
Article in English | MEDLINE | ID: mdl-29017971

ABSTRACT

Postoperative pain is a common form of acute pain that, if not managed effectively, can become chronic pain. Evidence has shown that glia, especially microglia, mediate neuroinflammation, which plays a vital role in pain sensitization. Moreover, toll-like receptor 4 (TLR4), the tumor necrosis factor receptor (TNF-R), the interleukin-1 receptor (IL-1R), and the interleukin-6 receptor (IL-6R) have been considered key components in central pain sensitization and neuroinflammation. Therefore, we hypothesized that activation of the body's endogenous "immune brakes" will inhibit these receptors and achieve inflammation tolerance as well as relieve postoperative pain. After searching for potential candidates to serve as this immune brake, we identified and focused on the suppressor of cytokine signaling 3 (SOCS3) gene. To regulate SOCS3 expression, we used paeoniflorin to induce heat shock protein 70 (HSP70)/TLR4 signaling. We found that paeoniflorin significantly induced SOCS3 expression both in vitro and in vivo and promoted the efflux of HSP70 from the cytoplasm to the extracellular environment. Furthermore, paeoniflorin markedly attenuated incision-induced mechanical allodynia, and this effect was abolished by small interfering RNAs targeting SOCS3. These findings demonstrated an effective and safe strategy to alleviate postoperative pain.


Subject(s)
Pain, Postoperative/immunology , Pain, Postoperative/metabolism , Suppressor of Cytokine Signaling 3 Protein/physiology , Animals , Cytokines/metabolism , Drug Tolerance/physiology , Glucosides/pharmacology , HSP70 Heat-Shock Proteins/metabolism , Hyperalgesia/metabolism , Inflammation/metabolism , Mice , Microglia/physiology , Monoterpenes/pharmacology , Neuralgia/metabolism , Neuroglia/physiology , Neuroimmunomodulation/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Toll-Like Receptor 4/metabolism
9.
J Exp Med ; 214(9): 2523-2533, 2017 Sep 04.
Article in English | MEDLINE | ID: mdl-28710273

ABSTRACT

Patients with hypomorphic mutations in STAT3 and patients with hypermorphic mutations in STAT1 share several clinical and cellular phenotypes suggesting overlapping pathophysiologic mechanisms. We, therefore, examined cytokine signaling and CD4+ T cell differentiation in these cohorts to characterize common pathways. As expected, differentiation of Th17 cells was impaired in both cohorts. We found that STAT1 was hyperphosphorylated in response to cytokine stimulation in both cohorts and that STAT1-dependent PD-L1 up-regulation-known to inhibit Th17 differentiation in mouse models-was markedly enhanced as well. Overexpression of SOCS3 strongly inhibited phosphorylation of STAT1 and PD-L1 up-regulation, suggesting that diminished SOCS3 expression may lead to the observed effects. Defects in Th17 differentiation could be partially overcome in vitro via PD-L1 inhibition and in a mouse model of STAT3 loss-of-function by crossing them with PD-1 knockout mice. PD-L1 may be a potential therapeutic target in several genetic diseases of immune deficiency affecting cytokine signaling.


Subject(s)
B7-H1 Antigen/physiology , Cell Differentiation/physiology , STAT1 Transcription Factor/physiology , STAT3 Transcription Factor/physiology , Th17 Cells/physiology , Adolescent , Adult , Animals , Child , Cytokines/physiology , Female , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Immunologic Deficiency Syndromes/physiopathology , Interleukins/physiology , Male , Mice , Mice, Knockout , Middle Aged , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/physiology , Suppressor of Cytokine Signaling 3 Protein/physiology , Up-Regulation , Young Adult
10.
Cytokine ; 96: 274-278, 2017 08.
Article in English | MEDLINE | ID: mdl-28554144

ABSTRACT

Excessive inflammation reduces skeletal muscle protein synthesis leading to wasting and weakness. The janus kinase/signal transducers and activators of transcription-3 (JAK/STAT3) pathway is important for the regulation of inflammatory signaling. As such, suppressor of cytokine signaling-3 (SOCS3), the negative regulator of JAK/STAT signaling, is thought to be important in the control of muscle homeostasis. We hypothesized that muscle-specific deletion of SOCS3 would impair the anabolic response to leucine during an inflammatory insult. Twelve week old (n=8 per group) SOCS3 muscle-specific knockout mice (SOCS3-MKO) and littermate controls (WT) were injected with lipopolysaccharide (LPS, 1mg/kg) or saline and were studied during fasted conditions or after receiving 0.5g/kg leucine 3h after the injection of LPS. Markers of inflammation, anabolic signaling, and protein synthesis were measured 4h after LPS injection. LPS injection robustly increased mRNA expression of inflammatory molecules (Socs3, Socs1, Il-6, Ccl2, Tnfα and Cd68). In muscles from SOCS3-MKO mice, the Socs3 mRNA response to LPS was significantly blunted (∼6-fold) while STAT3 Tyr705 phosphorylation was exacerbated (18-fold). Leucine administration increased protein synthesis in both WT (∼1.6-fold) and SOCS3-MKO mice (∼1.5-fold) compared to basal levels. LPS administration blunted this effect, but there were no differences between WT and SOCS3-MKO mice. Muscle-specific SOCS3 deletion did not alter the response of AKT, mTOR, S6 or 4EBP1 under any treatment conditions. Therefore, SOCS3 does not appear to mediate the early inflammatory or leucine-induced changes in protein synthesis in skeletal muscle.


Subject(s)
Anabolic Agents , Inflammation/metabolism , Leucine/administration & dosage , Muscle, Skeletal/metabolism , Protein Biosynthesis , Suppressor of Cytokine Signaling 3 Protein/physiology , Animals , Chemokine CCL2/genetics , Disease Models, Animal , Interleukin-6/genetics , Leucine/metabolism , Lipopolysaccharides/administration & dosage , Male , Mice , Mice, Knockout , Phosphorylation , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/deficiency , Suppressor of Cytokine Signaling 3 Protein/genetics , Tumor Necrosis Factor-alpha/genetics
11.
Inflammation ; 40(1): 58-67, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27704310

ABSTRACT

Obesity provokes an imbalance in the immune system, including an aberrant type I interferon response during some viral infections and after TLR stimulation. SOCS3 overexpression and altered systemic leptin levels could be responsible for the reduced type I interferon production in people with obesity and, eventually, significantly increase the risk of viral infection. The aim of this study was to determine whether SOCS3- and leptin-induced tolerance are responsible for the reduced type I interferon production in people with obesity. SOCS3 overexpression in PBMCs from people with obesity was inhibited with the small interfering RNA (siRNA) assay, and leptin-induced tolerance was evaluated in PBMCs from non-obese volunte\ers and U937 cells treated with TLR ligands. SOCS3, but not SOCS1, gene silencing via siRNA increased the type I interferon response in PBMCs obtained from people with obesity. On the other hand, leptin induced SOCS3 expression and inhibited type I interferons in PBMCs from healthy donors and in U937 monocytes stimulated with TLR ligands. Taken together, these results demonstrate that reduced type I interferon production in obesity is caused by SOCS3 overexpression as well as tolerance induced by leptin. Here, we demonstrate a key role of leptin and SOCS3 in inhibiting the type I interferon response during obesity.


Subject(s)
Interferon Type I/antagonists & inhibitors , Leptin/physiology , Obesity/metabolism , Suppressor of Cytokine Signaling 3 Protein/physiology , Cells, Cultured , Humans , Ligands , Monocytes , Toll-Like Receptors , U937 Cells
12.
PLoS One ; 11(11): e0166015, 2016.
Article in English | MEDLINE | ID: mdl-27824951

ABSTRACT

BACKGROUND: Elevated levels of C-reactive protein (CRP, determined by a high-sensitivity assay) indicate low-grade inflammation which is implicated in many age-related disorders. Epigenetic studies on CRP might discover molecular mechanisms underlying CRP regulation. We aimed to identify DNA methylation sites related to CRP concentrations in cells and tissues regulating low-grade inflammation. RESULTS: Genome-wide DNA methylation was measured in peripheral blood in 1,741 participants of the KORA F4 study using Illumina HumanMethylation450 BeadChip arrays. Four CpG sites (located at BCL3, AQP3, SOCS3, and cg19821297 intergenic at chromosome 19p13.2, P ≤ 1.01E-07) were significantly hypomethylated at high CRP concentrations independent of various confounders including age, sex, BMI, smoking, and white blood cell composition. Findings were not sex-specific. CRP-related top genes were enriched in JAK/STAT pathways (Benjamini-Hochberg corrected P < 0.05). Results were followed-up in three studies using DNA from peripheral blood (EPICOR, n = 503) and adipose tissue (TwinsUK, n = 368) measured as described above and from liver tissue (LMU liver cohort, n = 286) measured by MALDI-TOF mass spectrometry using EpiTYPER. CpG sites at the AQP3 locus (significant p-values in peripheral blood = 1.72E-03 and liver tissue = 1.51E-03) and the SOCS3 locus (p-values in liver < 2.82E-05) were associated with CRP in the validation panels. CONCLUSIONS: Epigenetic modifications seem to engage in low-grade inflammation, possibly via JAK/STAT mediated pathways. Results suggest a shared relevance across different tissues at the AQP3 locus and highlight a role of DNA methylation for CRP regulation at the SOCS3 locus.


Subject(s)
Aquaporin 3/physiology , Epigenesis, Genetic/physiology , Inflammation/genetics , Suppressor of Cytokine Signaling 3 Protein/physiology , Adipose Tissue/physiopathology , Aquaporin 3/genetics , C-Reactive Protein/physiology , DNA Methylation/physiology , Epigenesis, Genetic/genetics , Female , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Genome-Wide Association Study , Humans , Inflammation/physiopathology , Janus Kinases/physiology , Liver/physiopathology , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , STAT Transcription Factors/physiology , Signal Transduction/physiology , Suppressor of Cytokine Signaling 3 Protein/genetics
13.
Skelet Muscle ; 6: 36, 2016.
Article in English | MEDLINE | ID: mdl-27800152

ABSTRACT

BACKGROUND: Muscles of old animals are injured more easily and regenerate poorly, attributed in part to increased levels of circulating pro-inflammatory cytokines. The Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling cascade is a key mediator of inflammatory cytokine action, and signaling via this pathway is increased in muscles with aging. As a negative regulator of JAK/STAT signaling, a key mediator of myogenic proliferation and differentiation, altered expression of suppressor of cytokine signaling (SOCS3) is likely to have important consequences for muscle regeneration. To model this scenario, we investigated the effect of SOCS3 deletion within mature muscle fibers on injury and repair. We tested the hypothesis that reduced SOCS3 function would alter the inflammatory response and impair muscle regeneration after myotoxic injury. METHODS: Mice with a specific deletion of SOCS3 within mature skeletal muscle fibers were used to assess the effect of SOCS3 deletion on muscle injury and repair. Twelve-week-old or 24-month-old SOCS3 muscle-specific knockout (SOCS3 MKO) mice and littermate controls were either left uninjured or injured with a single injection of notexin (10 µg/ml) into the right tibialis anterior (TA) muscle. At 1, 2, 3, 5, 7, or 14 days post-injury, the right TA muscle was excised and subjected to histological, western immunoblotting, and gene expression analyses. Force production and fatigue were assessed in uninjured muscles and at 7 days post-notexin injury. RESULTS: In uninjured muscles, SOCS3 deletion decreased force production during fatigue but had no effect on the gross or histological appearance of the TA muscles. After notexin injury, deletion of SOCS3 increased STAT3 phosphorylation at day 1 and increased the mRNA expression of the inflammatory cytokine TNF-α, and the inflammatory cell markers F4/80 and CD68 at day 2. Gene expression analysis of the regeneration markers Pax7, MyoD, and Myogenin indicated SOCS3 deletion had no effect on the progression of muscle repair after notexin injury. Inflammation and regeneration were also unchanged in the muscles of 24-month-old SOCS3 MKO mice compared with control. CONCLUSIONS: Loss of SOCS3 expression in mature muscle fibers increased the inflammatory response to myotoxic injury but did not impair muscle regeneration in either adult or old mice. Therefore, reduced SOCS3 expression in muscle fibers is unlikely to underlie impaired muscle regeneration. Further investigation into the role of SOCS3 in other cell types involved in muscle repair is warranted.


Subject(s)
Muscle Fibers, Skeletal/physiology , Myositis/metabolism , Regeneration , Suppressor of Cytokine Signaling 3 Protein/physiology , Animals , Cells, Cultured , Cytokines/metabolism , Elapid Venoms , Female , Janus Kinases/metabolism , Male , Mice , Mice, Knockout , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Myositis/chemically induced , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism
14.
J Hematol Oncol ; 9(1): 69, 2016 08 12.
Article in English | MEDLINE | ID: mdl-27516205

ABSTRACT

BACKGROUND: Abnormal expression of SOCS3 has been implicated in myeloproliferative neoplasms, but the role of SOCS3 in the pathogenesis of leukemia remains largely unknown. Here, we examined the function of SOCS3 in the growth and chemo-sensitivity of chronic myeloid leukemia (CML) and explored the involved mechanisms. METHODS: Expression levels of SOCS3 in several leukemia cell lines and bone marrow mononuclear cells (BMNCs) from CML patients were determined using quantitative real-time PCR (qPCR) and Western blotting (WB). The roles of SOCS3 in the proliferation, apoptosis, and drug resistance of CML cells were examined by clonogenic progenitor cell assay, flow cytometry, and CCK-8 assay. A detailed analysis of the underlying mechanism of SOCS3 in K562 cells was performed using the Human HT-12 v4 Expression BeadChip, which has more than 48000 gene probes including 600 microRNAs (miRNA) probes. The correlation between the mRNA expression of SOCS3 and miR-124-3p in BMNCs from 30 CML patients was tested by qPCR and analyzed by Pearson correlation and linear regression analysis. The potential target of miR-124-3p in CML cells was explored using the luciferase reporter assay, qPCR, and WB. The effect of SOCS3 on the miR-124-3p/B4GALT1 axis was investigated by qPCR, WB, CCK-8 assay, and tumorigenicity assays in nude mice. RESULTS: SOCS3 was down-regulated in CML cell lines and most of BMNCs from CML patients, and the expression level of SOCS3 was associated with the inhibition of cell proliferation and drug resistance of CML cells. Over-expression of SOCS3 in K562 cells inhibited the expression of leukemia-specific genes and promoted the expression of some miRNAs, among which miR-124-3p was the highest. SOCS3 over-expression enhanced the expression of miR-124-3p and vice versa. The mRNA expression of miR-124-3p and SOCS3 in BMNCs from 30 CML patients was positively correlated. Consistently, the tumor suppressing effects of SOCS3 were partially neutralized by the miR-124-3p inhibitor. B4GALT1 was downstream of miR-124-3p and regulated by SOCS3/miR-124-3p in vitro. Furthermore, SOCS3 over-expression could inhibit the growth and B4GALT expression of K562 cells in vivo. CONCLUSIONS: SOCS3/miR-124-3p/B4GALT1 axis plays an important role in the pathogenesis of CML.


Subject(s)
Galactosyltransferases/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , MicroRNAs/metabolism , Suppressor of Cytokine Signaling 3 Protein/physiology , Animals , Bone Marrow Cells/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Drug Resistance, Neoplasm/drug effects , Galactosyltransferases/drug effects , Gene Expression Regulation, Leukemic , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukocytes, Mononuclear/metabolism , Mice, Nude , MicroRNAs/drug effects , RNA, Messenger/analysis , Suppressor of Cytokine Signaling 3 Protein/analysis , Suppressor of Cytokine Signaling 3 Protein/genetics
15.
J Leukoc Biol ; 100(4): 771-780, 2016 10.
Article in English | MEDLINE | ID: mdl-27106674

ABSTRACT

Suppressor of cytokine signaling (SOCS) proteins are recognized as key feedback inhibitors modulating the inflammatory activities of macrophages, but comparatively little is known about whether and how they affect phagocytosis. Here, we evaluated the role of SOCS3 in driving the inflammatory phenotype and phagocytic uptake of apoptotic cells by human macrophages and the signaling pathways that are necessary for efficient phagocytosis. In M1-activated human monocyte-derived macrophages, SOCS3 silencing, using short interfering RNA technology, resulted in a decreased expression of proinflammatory markers and an increased expression of M2 macrophage markers. Strikingly, we demonstrated for the first time that SOCS3 knockdown significantly enhances the phagocytic capacity of M1 macrophages for carboxylate-modified beads and apoptotic neutrophils. With the use of live-cell video microscopy, we showed that SOCS3 knockdown radically affects the temporal dynamics of particle engulfment, enabling more rapid uptake of a second target and delaying postengulfment processing, as evidenced by deferred acquisition of phagosome maturation markers. SOCS3 knockdown impacts on phagocytosis through increased PI3K and Ras-related C3 botulinum toxin substrate 1 (Rac1) activity, pathways essential for engulfment and clearance of apoptotic cells. Enhanced phagocytosis in SOCS3-silenced cells was reversed by pharmacological PI3K inhibition. Furthermore, we revealed that actin polymerization, downstream of PI3K/Rac1 activation, was significantly altered in SOCS3-silenced cells, providing a mechanism for their greater phagocytic activity. The findings support a new model, whereby SOCS3 not only plays an important role in driving macrophage inflammatory responses but modulates key signaling pathways organizing the actin cytoskeleton to regulate the efficiency of phagocytic processes.


Subject(s)
Macrophages/physiology , Phagocytosis/physiology , Suppressor of Cytokine Signaling 3 Protein/physiology , Actin Cytoskeleton/ultrastructure , Apoptosis , Cells, Cultured , Humans , Inflammation , Macrophages/classification , Macrophages/ultrastructure , Microscopy, Video , Microspheres , Phagosomes/physiology , Phagosomes/ultrastructure , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/physiology , Suppressor of Cytokine Signaling 3 Protein/deficiency , Suppressor of Cytokine Signaling 3 Protein/genetics , Time-Lapse Imaging , rac1 GTP-Binding Protein/physiology
16.
Biomarkers ; 21(5): 409-15, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27099071

ABSTRACT

We aimed to verify the levels of IGFBP2 and SOCS3 in cartilage and chondrocytes of Kashin-Beck disease (KBD) patients and the effects of different selenium concentrations on the protein expression levels. Chondrocytes were cultured with sodium selenite in vitro. Immunohistochemistry and western blotting were used to verify the protein expressions. IGFBP2 and SOCS3 were up-regulated in KBD chondrocytes and decreased with increasing selenium concentrations. IGFBP2 expressed highest in the middle zone of KBD cartilage, SOCS3 expressed higher in the middle and deep zone. IGFBP2 and SOCS3 may be the biomarkers for KBD diagnosis and evaluating the effect of selenium supplement.


Subject(s)
Insulin-Like Growth Factor Binding Protein 2/physiology , Kashin-Beck Disease/pathology , Selenium/pharmacology , Suppressor of Cytokine Signaling 3 Protein/physiology , Biomarkers, Pharmacological/analysis , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cells, Cultured , Chondrocytes/metabolism , Chondrocytes/pathology , Gene Expression Regulation/drug effects , Humans , Insulin-Like Growth Factor Binding Protein 2/analysis , Kashin-Beck Disease/drug therapy , Kashin-Beck Disease/etiology , Selenium/therapeutic use , Suppressor of Cytokine Signaling 3 Protein/analysis
17.
J Dent Res ; 95(9): 1018-25, 2016 08.
Article in English | MEDLINE | ID: mdl-27126447

ABSTRACT

The host immune response plays a key role in bacteria-induced alveolar bone resorption. Endogenous control of the magnitude and duration of inflammatory signaling is considered an important determinant of the extent of periodontal pathology. Suppressor of cytokine signaling (SOCS) proteins are inhibitors of cytokine signaling pathways and may play a role in restraining periodontal inflammation. We hypothesized that SOCS-3 regulates alveolar bone loss in experimental periodontitis. Periodontal bone loss was induced in 16-wk-old myeloid-specific SOCS-3-knockout and wild-type (WT) C57Bl6-B.129 mice by oral inoculation 9 times with 10(9) colony-forming units of Porphyromonas gingivalis A7436 through an oral gavage model for periodontitis. Sham controls for both types of mice received vehicle without bacteria. The mice were euthanized 6 wk after the last oral inoculation. Increased bone loss was demonstrated in P. gingivalis-infected SOCS-3-knockout mice as compared with P. gingivalis-infected WT mice by direct morphologic measurements, micro-computed tomography analyses, and quantitative histology. Loss of SOCS-3 function resulted in an increased number of alveolar bone osteoclasts and increased RANKL expression after P. gingivalis infection. SOCS-3 deficiency in myeloid cells also promotes a higher P. gingivalis lipopolysaccharide-induced inflammatory response with higher secretion of IL-1ß, IL-6, and KC (IL-8) by peritoneal macrophages as compared with WT controls. Our data implicate SOCS-3 as a critical negative regulator of alveolar bone loss in periodontitis.


Subject(s)
Alveolar Bone Loss/physiopathology , Periodontitis/physiopathology , Suppressor of Cytokine Signaling 3 Protein/physiology , Suppressor of Cytokine Signaling Proteins/physiology , Alveolar Bone Loss/diagnostic imaging , Alveolar Bone Loss/pathology , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/physiology , Periodontitis/pathology , Porphyromonas gingivalis , X-Ray Microtomography
18.
Oncotarget ; 7(15): 20621-35, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26967393

ABSTRACT

In glioma, microglia and macrophages are the largest population of tumor-infiltrating cells, referred to as glioma associated macrophages (GAMs). Herein, we sought to determine the role of Suppressor of Cytokine Signaling 3 (SOCS3), a negative regulator of Signal Transducer and Activator of Transcription 3 (STAT3), in GAM functionality in glioma. We utilized a conditional model in which SOCS3 deletion is restricted to the myeloid cell population. We found that SOCS3-deficient bone marrow-derived macrophages display enhanced and prolonged expression of pro-inflammatory M1 cytokines when exposed to glioma tumor cell conditioned medium in vitro. Moreover, we found that deletion of SOCS3 in the myeloid cell population delays intracranial tumor growth and increases survival of mice bearing orthotopic glioma tumors in vivo. Although intracranial tumors from mice with SOCS3-deficient myeloid cells appear histologically similar to control mice, we observed that loss of SOCS3 in myeloid cells results in decreased M2 polarized macrophage infiltration in the tumors. Furthermore, loss of SOCS3 in myeloid cells results in increased CD8+ T-cell and decreased regulatory T-cell infiltration in the tumors. These findings demonstrate a beneficial effect of M1 polarized macrophages on suppressing glioma tumor growth, and highlight the importance of immune cells in the tumor microenvironment.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Myeloid Cells/pathology , Suppressor of Cytokine Signaling 3 Protein/physiology , T-Lymphocytes, Regulatory/pathology , Animals , Apoptosis , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Cell Proliferation , Glioma/genetics , Glioma/immunology , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Cells, Cultured , Tumor Microenvironment/immunology
19.
Neurobiol Dis ; 89: 10-22, 2016 May.
Article in English | MEDLINE | ID: mdl-26804026

ABSTRACT

NG2 cells, also known as oligodendrocyte progenitors or polydendrocytes, are a major component of the glial scar that forms after spinal cord injury. NG2 cells react to injury by proliferating around the lesion site and differentiating into oligodendrocytes and astrocytes, but the molecular mechanism is poorly understood. In this study, we tested the role of the transcription factor STAT3, and its suppressor SOCS3, in NG2 cell proliferation and differentiation after spinal cord injury. Using knockout mice in which STAT3 or SOCS3 are genetically deleted specifically in NG2 cells, we found that deletion of STAT3 led to a reduction in oligodendrogenesis, while deletion of SOCS3 led to enhanced proliferation of NG2 cells within the glial scar after spinal cord injury. Additionally, STAT3 and SOCS3 were not required for astrogliogenesis from NG2 cells after spinal cord injury. Interestingly, genetic deletion of STAT3 and SOCS3 did not have opposing effects, suggesting that SOCS3 may have targets other than the STAT3 pathway in NG2 cells after spinal cord injury. Altogether, our data show that both STAT3 and SOCS3 play important, yet unexpected, roles in NG2 cell proliferation and differentiation after spinal cord injury.


Subject(s)
Cell Differentiation , Cell Proliferation , Oligodendroglia/physiology , STAT3 Transcription Factor/physiology , Spinal Cord Injuries/physiopathology , Suppressor of Cytokine Signaling 3 Protein/physiology , Animals , Astrocytes/physiology , Cell Count , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligodendroglia/metabolism , STAT3 Transcription Factor/genetics , Stem Cells/physiology , Suppressor of Cytokine Signaling 3 Protein/genetics
20.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 45(5): 469-476, 2016 05 25.
Article in Chinese | MEDLINE | ID: mdl-28087906

ABSTRACT

Objective: To investigate the effects and the underlying molecular mechanisms of curcumin on pulmonary artery smooth muscle cells in rat model with chronic obstructive pulmonary disease (COPD). Methods: A total of 75 male Wistar rats were randomly divided into control group (group CN), model group (group M), low-dose curcumin group (group CL), medium-dose curcumin group (group CM) and high-dose curcumin group (group CH). HE staining was used to observe the morphology of pulmonary artery. Proliferating cell nuclear antigen (PCNA), apoptosis-related protein Bcl-2 and Bax were detected by immunohistochemical staining. TUNEL kit was used to analyze the effects of curcumin on apoptosis of smooth muscle cells, and the protein expressions of SOCS-3/JAK2/STAT pathway in lung tissues were determined by western blot. Results: Right ventricular systolic pressure (RVSP) and right ventricular hypertrophy index (RVMI) in group M were significantly higher than those in group CN, group CH and group CM (all P<0.05). HE staining and TUNEL kit test showed that the number of pulmonary artery smooth muscle cells had a significant increase in group M, while the pulmonary artery tube became thin, and the smooth muscle cells shrinked in group CM and group CH. Immunohistochemistry showed that PCNA and Bcl-2 in group M were significantly higher than those in group CN (all P<0.05), while Bax expression was significantly lower than that in group CN (P<0.05). PCNA in group CM and group CH were significantly lower than that in group M (all P<0.05), while Bax expression was significantly higher than that in group M (P<0.05). Western blot showed that SOCS-3 protein was significantly decreased in group M, while the p-JAK2, p-STAT1, p-STAT3 were significantly increased (all P<0.05). Compared with group M, SOCS-3 protein in group CM and group CH were significantly increased (all P<0.05), while the p-JAK2, p-STAT3 were significantly reduced (all P<0.05). Conclusion: Curcumin could promote the apoptosis of smooth muscle cells in rats with COPD, and improve the mean pulmonary artery pressure and RVMI through stimulating SOCS-3/JAK2/STAT signaling pathway.


Subject(s)
Apoptosis/drug effects , Arterial Pressure/drug effects , Curcumin/pharmacology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Animals , Apoptosis/physiology , Arterial Pressure/physiology , Hypertrophy, Right Ventricular/pathology , Hypertrophy, Right Ventricular/physiopathology , Janus Kinase 2/drug effects , Janus Kinase 2/physiology , Lung/chemistry , Lung/drug effects , Male , Proliferating Cell Nuclear Antigen/drug effects , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , STAT Transcription Factors , Suppressor of Cytokine Signaling 3 Protein/drug effects , Suppressor of Cytokine Signaling 3 Protein/physiology , Ventricular Pressure/drug effects , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...