Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
PLoS One ; 16(7): e0254712, 2021.
Article in English | MEDLINE | ID: mdl-34292971

ABSTRACT

Ischemic preconditioning (IPC) is the most powerful endogenous cardioprotective form of cellular adaptation. However, the inhibitory or augmenting mechanism underlying cardioprotection via IPC remains largely unknown. Suppressor of cytokine signaling-3 (SOCS3) is a cytokine-inducible potent negative feedback regulator of the signal transducer and activator of transcription-3 (STAT3) signaling pathway. Here, we aimed to determine whether cardiac SOCS3 deficiency and IPC would synergistically reduce infarct size after myocardial ischemia reperfusion injury. We evaluated STAT3 activation and SOCS3 induction after ischemic conditioning (IC) using western blot analysis and real-time PCR, and found that myocardial IC alone transiently activated myocardial STAT3 and correspondingly induced SOCS3 expression in wild-type mice. Compared with wild-type mice, cardiac-specific SOCS3 knockout (SOCS3-CKO) mice showed significantly greater and more sustained IC-induced STAT3 activation. Following ischemia reperfusion, IPC substantially reduced myocardial infarct size and significantly enhanced STAT3 phosphorylation in SOCS3-CKO mice compared to in wild-type mice. Real-time PCR array analysis revealed that SOCS3-CKO mice after IC exhibited significantly increased expressions of several anti-apoptotic genes and SAFE pathway-related genes. Moreover, real-time PCR analysis revealed that myocardial IC alone rapidly induced expression of the STAT3-activating cytokine erythropoietin in the kidney at 1 h post-IC. We also found that the circulating erythropoietin level was promptly increased at 1 h after myocardial IC. Myocardial SOCS3 deficiency and IPC exert synergistic effects in the prevention of myocardial injury after ischemia reperfusion. Our present results suggest that myocardial SOCS3 is a potent inhibitor of IPC-induced cardioprotection, and that myocardial SOCS3 inhibition augment IPC-mediated cardioprotection during ischemia reperfusion injury.


Subject(s)
Ischemic Preconditioning , Myocardial Reperfusion Injury , Myocytes, Cardiac/metabolism , Suppressor of Cytokine Signaling 3 Protein/deficiency , Animals , Erythropoietin/genetics , Erythropoietin/metabolism , Mice , Mice, Knockout , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/pathology
2.
J Innate Immun ; 12(3): 248-256, 2020.
Article in English | MEDLINE | ID: mdl-31574508

ABSTRACT

Mononuclear phagocytes, such as macrophages and microglia, are key regulators of organ homeostasis including vascularization processes. Here, we investigated the role of the suppressor of cytokine signaling 3 (SOCS3) in myeloid cells as a regulator of mononuclear phagocyte function and their interaction with endothelial cells in the context of sprouting angiogenesis. As compared to SOCS3-sufficient counterparts, SOCS3-deficient microglia and macrophages displayed an increased phagocytic activity toward primary apoptotic endothelial cells, which was associated with an enhanced expression of the opsonin growth arrest-specific 6 (Gas6), a major prophagocytic molecule. Furthermore, we found that myeloid SOCS3 deficiency significantly reduced angiogenesis in an ex vivo mouse aortic ring assay, which could be reversed by the inhibition of the Gas6 receptor Mer. Together, SOCS3 in myeloid cells regulates the Gas6/Mer-dependent phagocytosis of endothelial cells, and thereby angiogenesis-related processes. Our findings provide novel insights into the complex crosstalk between mononuclear phagocytes and endothelial cells, and may therefore provide a new platform for the development of new antiangiogenic therapies.


Subject(s)
Apoptosis/immunology , Endothelial Cells/immunology , Myeloid Cells/immunology , Neovascularization, Physiologic/immunology , Suppressor of Cytokine Signaling 3 Protein/deficiency , Animals , Apoptosis/genetics , Mice , Mice, Transgenic , Neovascularization, Physiologic/genetics , Phagocytosis , Suppressor of Cytokine Signaling 3 Protein/immunology
3.
J Cell Mol Med ; 23(12): 8101-8113, 2019 12.
Article in English | MEDLINE | ID: mdl-31559687

ABSTRACT

Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) are life-threatening condition in critically ill patients. Resveratrol (Res), a natural polyphenol, has therapeutic effect in animal model with ALI; however, whether Res attenuates ALI through modulation of macrophage phenotypes in the animal model remains unknown. We in this study treated LPS-induced murine ALI with 30 mg/kg Res and observed significantly reduced severity of ALI in the Res-treated mice 48 hours after Res treatment. Neutrophil infiltrates were significantly reduced, accompanied with lower infiltration of CD45+ Siglec F- phenotype macrophages, but higher population of CD45+ Siglec F+ and CD45+ CD206+ alternatively activated macrophages (M2 cells) in the Res-treated mice with ALI. In addition, the expression of IL-1beta and CXCL15 cytokines was suppressed in the treated mice. However, Res treatment in mice with myeloid cell-restricted SOCS3 deficiency did not significantly attenuate ALI severity and failed to increase population of both CD45+ Siglec F+ and CD45+ CD206+ M2 subtype macrophages in the murine ALI. Further studies in wild-type macrophages revealed that Res treatment effectively reduced the expression of IL-6 and CXCL15, and increased the expression of arginase-1, SIRT1 and SOCS3. However, macrophages' lack of SOCS3 expression were resistant to the Res-induced suppression of IL-6 and CXCL15 in vitro. Thus, we conclude that Res suppressed CD45+ Siglec F- and CD45+ CD206- M1 subtype macrophages through SOCS3 signalling in the LPS-induced murine ALI.


Subject(s)
Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Lectins, C-Type/metabolism , Leukocyte Common Antigens/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mannose-Binding Lectins/metabolism , Receptors, Cell Surface/metabolism , Resveratrol/therapeutic use , Suppressor of Cytokine Signaling 3 Protein/metabolism , Acetylation , Acute Lung Injury/chemically induced , Acute Lung Injury/genetics , Animals , Arginase/genetics , Arginase/metabolism , Chemokines, CXC/genetics , Chemokines, CXC/metabolism , Disease Models, Animal , Interleukin-6/genetics , Interleukin-6/metabolism , Lipopolysaccharides , Male , Mannose Receptor , Mice , Mice, Inbred C57BL , Mice, Knockout , Resveratrol/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Sirtuin 1/genetics , Sirtuin 1/metabolism , Suppressor of Cytokine Signaling 3 Protein/deficiency , Suppressor of Cytokine Signaling 3 Protein/genetics
4.
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
5.
Am J Pathol ; 188(4): 1007-1020, 2018 04.
Article in English | MEDLINE | ID: mdl-29452101

ABSTRACT

The suppressor of cytokine signaling protein 3 (SOCS3) critically controls immune cell activation, although its role in macrophage polarization and function remains controversial. Using experimental autoimmune uveoretinitis (EAU) as a model, we show that inflammation-mediated retinal degeneration is exaggerated and retinal angiogenesis is accelerated in mice with SOCS3 deficiency in myeloid cells (LysMCre/+SOCS3fl/fl). At the acute stage of EAU, the population of infiltrating neutrophils was increased and the population of macrophages decreased in LysMCre/+SOCS3fl/fl mice compared with that in wild-type (WT) mice. Real-time RT-PCR showed that the expression of tumor necrosis factor-α, IL-1ß, interferon-γ, granulocyte-macrophage colony-stimulating factor, and arginase-1 was significantly higher in the LysMCre/+SOCS3fl/fl EAU retina in contrast to the WT EAU retina. The percentage of arginase-1+ infiltrating cells was significantly higher in the LysMCre/+SOCS3fl/fl EAU retina than that in the WT EAU retina. In addition, bone marrow-derived macrophages and neutrophils from the LysMCre/+SOCS3fl/fl mice express significantly higher levels of chemokine (C-C motif) ligand 2 and arginase-1 compared with those from WT mice. Inhibition of arginase using an l-arginine analog amino-2-borono-6-hexanoic suppressed inflammation-induced retinal angiogenesis without affecting the severity of inflammation. Our results suggest that SOCS3 critically controls the phenotype and function of macrophages and neutrophils under inflammatory conditions and loss of SOCS3 promotes the angiogenic phenotype of the cells through up-regulation of arginase-1.


Subject(s)
Arginase/genetics , Autoimmune Diseases/genetics , Myeloid Cells/metabolism , Neovascularization, Pathologic/metabolism , Retinal Degeneration/pathology , Suppressor of Cytokine Signaling 3 Protein/deficiency , Up-Regulation/genetics , Uveal Diseases/genetics , Animals , Arginase/antagonists & inhibitors , Arginase/metabolism , Bone Marrow Cells/metabolism , Inflammation/pathology , Macrophages/metabolism , Mice, Inbred C57BL , Neutrophils/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism
6.
Exp Neurol ; 303: 120-133, 2018 05.
Article in English | MEDLINE | ID: mdl-29458059

ABSTRACT

Axons within the peripheral nervous system are capable of regeneration, but full functional recovery is rare. Recent work has shown that conditional deletion of two key signaling inhibitors of the PI3K and Jak/Stat pathways-phosphatase and tensin homolog (PTEN) and suppressor of cytokine signaling-3 (SOCS3), respectively-promotes regeneration of normally non-regenerative central nervous system axons. Moreover, in studies of optic nerve regeneration, co-deletion of both PTEN and SOCS3 has an even greater effect. Here, we test the hypotheses (1) that PTEN deletion enhances axon regeneration following sciatic nerve crush and (2) that PTEN/SOCS3 co-deletion further promotes regeneration. PTENfl/fl and PTEN/SOCS3fl/fl mice received direct injections of AAV-Cre into the fourth and fifth lumbar dorsal root ganglia (DRG) two weeks prior to sciatic nerve crush. Western blot analysis of whole cell lysates from DRG using phospho-specific antibodies revealed that PTEN deletion did not enhance or prolong PI3K signaling following sciatic nerve crush. However, PTEN/SOCS3 co-deletion activated PI3K for at least 7 days post-injury in contrast to controls, where activation peaked at 3 days. Quantification of SCG10-expressing regenerating sensory axons in the sciatic nerve after crush injury revealed longer distance regeneration at 3 days post-injury with both PTEN and PTEN/SOCS3 co-deletion. Additionally, analysis of noxious thermosensation and mechanosensation with PTEN/SOCS3 co-deletion revealed enhanced sensation at 14 and 21 days after crush, respectively, after which all treatment groups reached the same functional plateau. These findings indicate that co-deletion of PTEN and SOCS3 results in modest but measureable enhancement of early regeneration of DRG axons following crush injury.


Subject(s)
Ganglia, Spinal/metabolism , Nerve Regeneration/genetics , PTEN Phosphohydrolase/deficiency , Recovery of Function/genetics , Sciatic Neuropathy/pathology , Suppressor of Cytokine Signaling 3 Protein/deficiency , Animals , Disease Models, Animal , Ganglia, Spinal/physiopathology , Gene Expression Regulation/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hyperalgesia/physiopathology , Male , Mice , Mice, Transgenic , Motor Activity/genetics , PTEN Phosphohydrolase/genetics , Pain Measurement , Phosphatidylinositol 3-Kinases , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Time Factors
7.
J Am Heart Assoc ; 7(2)2018 01 17.
Article in English | MEDLINE | ID: mdl-29343476

ABSTRACT

BACKGROUND: Aortic dissection (AD) is a life-threatening medical emergency caused by the abrupt destruction of the intimomedial layer of the aortic walls. Given that previous studies have reported the involvement of proinflammatory cytokine interleukin-6 in AD pathogenesis, we investigated the role of signal transduction and activator of transcription 3 signaling, a downstream pathway of interleukin-6 in macrophages in pathogenesis of AD. METHODS AND RESULTS: We characterized the pathological and molecular events triggered by aortic stress, which can lead to AD. Aortic stress on the suprarenal aorta because of infrarenal aorta stiffening and angiotensin II infusion for 1 week caused focal medial rupture at the branching point of the celiac trunk and superior mesenteric artery. This focal medial rupture healed in 6 weeks in wild-type (WT) mice, but progressed to AD in mice with macrophage-specific deletion of Socs3 gene (mSocs3-KO). mSocs3-KO mice showed premature activation of cell proliferation, an inflammatory response, and skewed differentiation of macrophages toward the tissue-destructive phenotype. Concomitantly, they showed aberrant phenotypic modulation of smooth muscle cells and transforming growth factor beta signaling, which are likely to participate in tissue repair. Human AD samples revealed signal transduction and activator of transcription 3 activation in adventitial macrophages adjacent to the site of tissue destruction. CONCLUSIONS: These findings suggest that AD development is preceded by focal medial rupture, in which macrophage Socs3 maintains proper inflammatory response and differentiation of SMCs, thus promoting fibrotic healing to prevent tissue destruction and AD development. Understanding the sequence of the pathological and molecular events preceding AD development will help predict and prevent AD development and progression.


Subject(s)
Aorta/metabolism , Aortic Aneurysm/metabolism , Aortic Dissection/metabolism , Macrophages/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Vascular Remodeling , Aortic Dissection/chemically induced , Aortic Dissection/genetics , Aortic Dissection/pathology , Angiotensins , Animals , Aorta/pathology , Aortic Aneurysm/chemically induced , Aortic Aneurysm/genetics , Aortic Aneurysm/pathology , Calcium Chloride , Cell Differentiation , Cell Proliferation , Dilatation, Pathologic , Disease Models, Animal , Disease Progression , Fibrosis , Gene Regulatory Networks , Humans , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Phenotype , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/deficiency , Suppressor of Cytokine Signaling 3 Protein/genetics , Time Factors , Transcriptome
8.
Respir Res ; 18(1): 217, 2017 12 29.
Article in English | MEDLINE | ID: mdl-29284516

ABSTRACT

BACKGROUND: SOCS3 (suppressor of cytokine signaling 3) is a negative regulator of JAK/STAT3 signaling pathway and participates in the regulation of lung inflammation in a mouse model with acute lung injury (ALI). However, it is not well understood how SOCS3 regulates lung inflammation in the ALI mouse model. METHOD: In the present study, we investigated the effects of SOCS3 on modulation of Ly6C(+) monocyte phenotypes in a mouse model with lipopolysaccharide (LPS)-induced ALI. Conditional SOCS3(Lyz2cre) mice with myeloid cell-restricted depletion of SOCS3 gene were created by breeding transgenic Lyz2Cre mice with SOCS3(fl/fl) mice. Wilde-type (WT) and SOCS3(Lyz2cre) mice were intratracheal instilled with 5 mg/kg LPS for 2 days. Lung, bronchoalveolar lavage (BAL) and blood were collected for analysis by flow cytometry, ELISA, qRT-PCR and Western blot analysis. RESULTS: The studies in the ALI mouse model revealed that myeloid cell-restricted SOCS3 deficiency exacerbated the severity of ALI as compared to the WT mice. The increased severity of ALI in SOCS3-deficient mice was associated with higher populations of neutrophils, T lymphocytes and Ly6C(+) monocytes in the inflamed lung tissues. In addition, CCR2 and CXCL15 were elevated, and accompanied by greater expression and activation of STAT3 in the lung of SOCS3-deficient mice. SOCS3-deficient bone marrow-derived macrophages (BMDMs) expressed a higher amount of TNF-alpha, and adoptive transfer of the SOCS3-deficient Ly6C(+) BMDMs into WT mice enhanced the severity of ALI than adoptive transfer of WT control BMDMs. However, depletion of Ly6C(+) circulating monocytes by anti-Ly6C(+) neutralizing antibody moderately attenuated neutrophil infiltration and resulted in lower prevalence of Ly6C(+) cells in the lung of treated mice. CONCLUSION: Myeloid cell-restricted lack of SOCS3 induced more severe ALI through modulation of Ly6C(+) subtype macrophages. The results provide insight into a new role of SOCS3 in modulation of Ly6C(+) monocyte phenotypes and provide a novel therapeutic strategy for ALI by molecular intervention of macrophages subtypes.


Subject(s)
Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Antigens, Ly/metabolism , Lipopolysaccharides/toxicity , Macrophages/metabolism , Suppressor of Cytokine Signaling 3 Protein/deficiency , Acute Lung Injury/immunology , Animals , Antigens, Ly/immunology , Cells, Cultured , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Suppressor of Cytokine Signaling 3 Protein/immunology
9.
Neuroscience ; 365: 114-124, 2017 Dec 04.
Article in English | MEDLINE | ID: mdl-28987511

ABSTRACT

Previous studies have shown that leptin resistance is a key feature that leads to gestational metabolic adaptions. We hypothesized that leptin sensitivity in the ventromedial nucleus of the hypothalamus (VMH) plays a critical role regulating gestational metabolic changes. In the present study, we generated a mouse model carrying ablation of the suppressor of cytokine signaling 3 (SOCS3) in steroidogenic factor-1 (SF1) cells, which include the VMH, in order to investigate whether increased leptin sensitivity in this neuronal population prevents at least part of the metabolic changes typically observed during gestation and lactation. As predicted by the inhibitory effects of SOCS3 in leptin signaling, pregnant SF1 SOCS3 KO mice exhibited increased leptin sensitivity in the VMH, since an acute leptin injection induced a 95% increase in the STAT3 phosphorylation in this nucleus, compared to control animals (p = 0.02). Despite that, SF1 SOCS3 KO mice showed similar weight gain, food intake, hypothalamic neuropeptide expression and serum leptin levels during pregnancy compared to control littermates. Unexpectedly, SF1 SOCS3 KO mice exhibited glucose intolerance during pregnancy. SF1 SOCS3 KO mice also presented a lower body weight (-3%; p < 0.05) during mid and late lactation, although food intake, litter size and offspring growth were not affected. Our findings suggest that increased leptin sensitivity in the VMH causes modest metabolic effects and is not sufficient to prevent major metabolic adaptations of pregnancy and lactation.


Subject(s)
Lactation/metabolism , Neurons/metabolism , Pregnancy/metabolism , Steroidogenic Factor 1/metabolism , Suppressor of Cytokine Signaling 3 Protein/deficiency , Adiposity/drug effects , Adiposity/genetics , Animals , Body Weight/drug effects , Energy Metabolism/drug effects , Energy Metabolism/genetics , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glucose Tolerance Test , Insulin/metabolism , Lactation/drug effects , Leptin/pharmacology , Mice , Mice, Transgenic , Neurons/drug effects , RNA, Messenger/metabolism , Steroidogenic Factor 1/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Ventromedial Hypothalamic Nucleus/cytology
10.
PLoS One ; 12(9): e0184672, 2017.
Article in English | MEDLINE | ID: mdl-28926622

ABSTRACT

Improving axonal transport in the injured and diseased central nervous system has been proposed as a promising strategy to improve neuronal repair. However, the contribution of each cargo to the repair mechanism is unknown. DRG neurons globally increase axonal transport during regeneration. Because the transport of specific cargos after axonal insult has not been examined systematically in a model of enhanced regenerative capacity, it is unknown whether the transport of all cargos would be modulated equally in injured central nervous system neurons. Here, using a microfluidic culture system we compared neurons co-deleted for PTEN and SOCS3, an established model of high axonal regeneration capacity, to control neurons. We measured the axonal transport of three cargos (mitochondria, synaptic vesicles and late endosomes) in regenerating axons and found that the transport of mitochondria, but not the other cargos, was increased in PTEN/SOCS3 co-deleted axons relative to controls. The results reported here suggest a pivotal role for this organelle during axonal regeneration.


Subject(s)
Axons/physiology , Mitochondria/metabolism , Nerve Regeneration/physiology , Animals , Biological Transport , Cells, Cultured , Cerebral Cortex/cytology , Female , Immunohistochemistry , Mice, Transgenic , Microscopy, Confocal , Neurons/cytology , Neurons/metabolism , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Rats , Suppressor of Cytokine Signaling 3 Protein/deficiency , Suppressor of Cytokine Signaling 3 Protein/genetics , Time-Lapse Imaging , Tubulin/metabolism
11.
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
12.
J Neurosci ; 36(27): 7142-53, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27383590

ABSTRACT

UNLABELLED: The cellular processes that cause high caloric diet (HCD)-induced infertility are poorly understood but may involve upregulation of suppressor of cytokine signaling (SOCS-3) proteins that are associated with hypothalamic leptin resistance. Deletion of SOCS-3 from brain cells is known to protect mice from diet-induced obesity, but the effects on HCD-induced infertility are unknown. We used neuron-specific SOCS3 knock-out mice to elucidate this and the effects on regional hypothalamic leptin resistance. As expected, male and female neuron-specific SOCS3 knock-out mice were protected from HCD-induced obesity. While female wild-type mice became infertile after 4 months of HCD feeding, infertility onset in knock-out females was delayed by 4 weeks. Similarly, knock-out mice had delayed leptin resistance development in the medial preoptic area and anteroventral periventricular nucleus, regions important for generation of the surge of GnRH and LH that induces ovulation. We therefore tested whether the suppressive effects of HCD on the estradiol-induced GnRH/LH surge were overcome by neuron-specific SOCS3 knock-out. Although only 20% of control HCD-mice experienced a preovulatory-like LH surge, LH surges could be induced in almost all neuron-specific SOCS3 knock-out mice on this diet. In contrast to females, HCD-fed male mice did not exhibit any fertility decline compared with low caloric diet-fed males despite their resistance to the satiety effects of leptin. These data show that deletion of SOCS3 delays the onset of leptin resistance and infertility in HCD-fed female mice, but given continued HCD feeding this state does eventually occur, presumably in response to other mechanisms inhibiting leptin signal transduction. SIGNIFICANCE STATEMENT: Obesity is commonly associated with infertility in humans and other animals. Treatments for human infertility show a decreased success rate with increasing body mass index. A hallmark of obesity is an increase in circulating leptin levels; despite this, the brain responds as if there were low levels of leptin, leading to increased appetite and suppressed fertility. Here we show that leptin resistant infertility is caused in part by the leptin signaling molecule SOCS3. Deletion of SOCS3 from brain neurons delays the onset of diet-induced infertility.


Subject(s)
Hypothalamus/metabolism , Infertility/therapy , Leptin/metabolism , Luteinizing Hormone/blood , Neurons/physiology , Obesity/complications , Prosencephalon/pathology , Suppressor of Cytokine Signaling 3 Protein/deficiency , Age Factors , Animals , Body Weight , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Estrous Cycle/drug effects , Estrous Cycle/genetics , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Infertility/etiology , Male , Mice , Mice, Inbred DBA , Mice, Transgenic , Obesity/etiology , Suppressor of Cytokine Signaling 3 Protein/genetics
13.
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
14.
Am J Physiol Heart Circ Physiol ; 311(1): H146-56, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27106041

ABSTRACT

Carotid artery disease is a major contributor to stroke and cognitive deficits. Angiotensin II (Ang II) promotes vascular dysfunction and disease through mechanisms that include the IL-6/STAT3 pathway. Here, we investigated the importance of suppressor of cytokine signaling 3 (SOCS3) in models of Ang II-induced vascular dysfunction. We examined direct effects of Ang II on carotid arteries from SOCS3-deficient (SOCS3(+/-)) mice and wild-type (WT) littermates using organ culture and then tested endothelial function with acetylcholine (ACh). A low concentration of Ang II (1 nmol/l) did not affect ACh-induced vasodilation in WT but reduced that of SOCS3(+/-) mice by ∼50% (P < 0.05). In relation to mechanisms, effects of Ang II in SOCS3(+/-) mice were prevented by inhibitors of STAT3, IL-6, NF-κB, or superoxide. Systemic Ang II (1.4 mg/kg per day for 14 days) also reduced vasodilation to ACh in WT. Surprisingly, SOCS3 deficiency prevented most of the endothelial dysfunction. To examine potential underlying mechanisms, we performed bone marrow transplantation. WT mice reconstituted with SOCS3(+/-) bone marrow were protected from Ang II-induced endothelial dysfunction, whereas reconstitution of SOCS3(+/-) mice with WT bone marrow exacerbated Ang II-induced effects. The SOCS3 genotype of bone marrow-derived cells did not influence direct effects of Ang II on vascular function. These data provide new mechanistic insight into the influence of SOCS3 on the vasculature, including divergent effects depending on the source of Ang II. Bone marrow-derived cells deficient in SOCS3 protect against systemic Ang II-induced vascular dysfunction.


Subject(s)
Angiotensin II , Aorta/metabolism , Basilar Artery/metabolism , Bone Marrow Cells/metabolism , Carotid Arteries/metabolism , Hypertension/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Vasodilation , Animals , Aorta/drug effects , Aorta/physiopathology , Basilar Artery/drug effects , Basilar Artery/physiopathology , Bone Marrow Transplantation , Carotid Arteries/drug effects , Carotid Arteries/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Genotype , Hypertension/chemically induced , Hypertension/physiopathology , Hypertension/prevention & control , Interleukin-6/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/metabolism , Organ Culture Techniques , Phenotype , STAT3 Transcription Factor/metabolism , Signal Transduction , Superoxides/metabolism , Suppressor of Cytokine Signaling 3 Protein/deficiency , Suppressor of Cytokine Signaling 3 Protein/genetics , Vasodilation/drug effects , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...