Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Transl Med ; 21(1): 290, 2023 04 29.
Article in English | MEDLINE | ID: mdl-37120549

ABSTRACT

BACKGROUND: Oncostatin M (OSM) is a secreted cytokine of the interleukin (IL)-6 family that induces biological effects by activating functional receptor complexes of the common signal transducing component glycoprotein 130 (gp130) and OSM receptor ß (OSMR) or leukaemia inhibitory factor receptor (LIFR), which are mainly involved in chronic inflammatory and cardiovascular diseases. The effect and underlying mechanism of OSM/OSMR/LIFR on the development of cardiac hypertrophy remains unclear. METHODS AND RESULTS: OSMR-knockout (OSMR-KO) mice were subjected to aortic banding (AB) surgery to establish a model of pressure overload-induced cardiac hypertrophy. Echocardiographic, histological, biochemical and immunological analyses of the myocardium and the adoptive transfer of bone marrow-derived macrophages (BMDMs) were conducted for in vivo studies. BMDMs were isolated and stimulated with lipopolysaccharide (LPS) for the in vitro study. OSMR deficiency aggravated cardiac hypertrophy, fibrotic remodelling and cardiac dysfunction after AB surgery in mice. Mechanistically, the loss of OSMR activated OSM/LIFR/STAT3 signalling and promoted a proresolving macrophage phenotype that exacerbated inflammation and impaired cardiac repair during remodelling. In addition, adoptive transfer of OSMR-KO BMDMs to WT mice after AB surgery resulted in a consistent hypertrophic phenotype. Moreover, knockdown of LIFR in myocardial tissue with Ad-shLIFR ameliorated the effects of OSMR deletion on the phenotype and STAT3 activation. CONCLUSIONS: OSMR deficiency aggravated pressure overload-induced cardiac hypertrophy by modulating macrophages and OSM/LIFR/STAT3 signalling, which provided evidence that OSMR might be an attractive target for treating pathological cardiac hypertrophy and heart failure.


Subject(s)
Interleukin-6 , Receptors, OSM-LIF , Receptors, Oncostatin M , Signal Transduction , Animals , Mice , Cardiomegaly , Macrophages , Oncostatin M/genetics , Receptors, OSM-LIF/genetics , Receptors, Oncostatin M/genetics
2.
Biomark Med ; 16(6): 461-471, 2022 04.
Article in English | MEDLINE | ID: mdl-35321549

ABSTRACT

Aim: Ovarian cancer (OC) is a gynecological malignancy with a challenging judgment of prognosis due to complicated etiology and high recurrence rate. The oncostatin M receptor (OSMR) from members of the IL-6 receptor family is associated with tumor development. This study aims to explore the correlations between OSMR gene polymorphisms (rs2278329 [G/A, missense, Asp553Asn], rs2292016 [G/T, promoter, -100G/T]) and OC. Methods: This study enrolled 160 OC patients and 397 healthy controls. Genotypes of two single-nucleotide polymorphisms were distinguished using TaqMan SNP Genotyping Assay, and statistical analysis was performed using SPSS software. Results: A significantly decreased overall survival rate was found in serous OC patients carrying rs2278329 GA/AA genotypes. Meanwhile, TT genotype carriers of rs2292016 had an improved relapse rate, and the GT genotype showed a definitive correlation with a lower relapse rate. Conclusion:OSMR gene polymorphisms may be related to recurrence and overall survival of serous OC patients.


Subject(s)
Neoplasm Recurrence, Local , Ovarian Neoplasms , Carcinoma, Ovarian Epithelial , Case-Control Studies , China/epidemiology , Female , Genetic Predisposition to Disease , Genotype , Humans , Neoplasm Recurrence, Local/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Polymorphism, Single Nucleotide , Receptors, Oncostatin M/genetics
3.
Int J Mol Sci ; 23(3)2022 Feb 05.
Article in English | MEDLINE | ID: mdl-35163735

ABSTRACT

Oncostatin M (OSM), a member of the interleukin-6 family, functions as a major mediator of cardiomyocyte remodeling under pathological conditions. Its involvement in a variety of human cardiac diseases such as aortic stenosis, myocardial infarction, myocarditis, cardiac sarcoidosis, and various cardiomyopathies make the OSM receptor (OSMR) signaling cascades a promising therapeutic target. However, the development of pharmacological treatment strategies is highly challenging for many reasons. In mouse models of heart disease, OSM elicits opposing effects via activation of the type II receptor complex (OSMR/gp130). Short-term activation of OSMR/gp130 protects the heart after acute injury, whereas chronic activation promotes the development of heart failure. Furthermore, OSM has the ability to integrate signals from unrelated receptors that enhance fetal remodeling (dedifferentiation) of adult cardiomyocytes. Because OSM strongly stimulates the production and secretion of extracellular proteins, it is likely to exert systemic effects, which in turn, could influence cardiac remodeling. Compared with the mouse, the complexity of OSM signaling is even greater in humans because this cytokine also activates the type I leukemia inhibitory factor receptor complex (LIFR/gp130). In this article, we provide an overview of OSM-induced cardiomyocyte remodeling and discuss the consequences of OSMR/gp130 and LIFR/gp130 activation under acute and chronic conditions.


Subject(s)
Heart Failure , Interleukin-6 , Myocytes, Cardiac , Oncostatin M , Receptors, Oncostatin M , Animals , Cytokine Receptor gp130/metabolism , Humans , Interleukin-6/metabolism , Mice , Myocytes, Cardiac/metabolism , Oncostatin M/metabolism , Oncostatin M Receptor beta Subunit , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism
4.
Nat Commun ; 11(1): 4116, 2020 08 17.
Article in English | MEDLINE | ID: mdl-32807793

ABSTRACT

Glioblastoma contains a rare population of self-renewing brain tumor stem cells (BTSCs) which are endowed with properties to proliferate, spur the growth of new tumors, and at the same time, evade ionizing radiation (IR) and chemotherapy. However, the drivers of BTSC resistance to therapy remain unknown. The cytokine receptor for oncostatin M (OSMR) regulates BTSC proliferation and glioblastoma tumorigenesis. Here, we report our discovery of a mitochondrial OSMR that confers resistance to IR via regulation of oxidative phosphorylation, independent of its role in cell proliferation. Mechanistically, OSMR is targeted to the mitochondrial matrix via the presequence translocase-associated motor complex components, mtHSP70 and TIM44. OSMR interacts with NADH ubiquinone oxidoreductase 1/2 (NDUFS1/2) of complex I and promotes mitochondrial respiration. Deletion of OSMR impairs spare respiratory capacity, increases reactive oxygen species, and sensitizes BTSCs to IR-induced cell death. Importantly, suppression of OSMR improves glioblastoma response to IR and prolongs lifespan.


Subject(s)
Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Radiation, Ionizing , Receptors, Oncostatin M/metabolism , Animals , Cell Death/radiation effects , Cell Line, Tumor , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Male , Mice , Mice, SCID , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Neoplastic Stem Cells/radiation effects , Oncostatin M/metabolism , Oxidative Stress/radiation effects , Receptors, Oncostatin M/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/radiation effects
5.
Acta Neuropathol Commun ; 8(1): 42, 2020 04 05.
Article in English | MEDLINE | ID: mdl-32248843

ABSTRACT

Glioblastoma (GBM) is characterized by extensive tumor cell invasion, angiogenesis, and proliferation. We previously established subclones of GBM cells with distinct invasive phenotypes and identified annexin A2 (ANXA2) as an activator of angiogenesis and perivascular invasion. Here, we further explored the role of ANXA2 in regulating phenotypic transition in GBM. We identified oncostatin M receptor (OSMR) as a key ANXA2 target gene in GBM utilizing microarray analysis and hierarchical clustering analysis of the Ivy Glioblastoma Atlas Project and The Cancer Genome Atlas datasets. Overexpression of ANXA2 in GBM cells increased the expression of OSMR and phosphorylated signal transducer and activator of transcription 3 (STAT3) and enhanced cell invasion, angiogenesis, proliferation, and mesenchymal transition. Silencing of OSMR reversed the ANXA2-induced phenotype, and STAT3 knockdown reduced OSMR protein expression. Exposure of GBM cells to hypoxic conditions activated the ANXA2-STAT3-OSMR signaling axis. Mice bearing ANXA2-overexpressing GBM exhibited shorter survival times compared with control tumor-bearing mice, whereas OSMR knockdown increased the survival time and diminished ANXA2-mediated tumor invasion, angiogenesis, and growth. Further, we uncovered a significant relationship between ANXA2 and OSMR expression in clinical GBM specimens, and demonstrated their correlation with tumor histopathology and patient prognosis. Our results indicate that the ANXA2-STAT3-OSMR axis regulates malignant phenotypic changes and mesenchymal transition in GBM, suggesting that this axis is a promising therapeutic target to treat GBM aggressiveness.


Subject(s)
Annexin A2/genetics , Brain Neoplasms/genetics , Glioblastoma/genetics , Oncostatin M Receptor beta Subunit/genetics , STAT3 Transcription Factor/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Annexin A2/metabolism , Brain Neoplasms/blood supply , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation/genetics , Child , Dogs , Epithelial-Mesenchymal Transition/genetics , Female , Gene Knockdown Techniques , Gene Silencing , Glioblastoma/blood supply , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Oncostatin M Receptor beta Subunit/metabolism , Phenotype , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Survival Rate , Tumor Hypoxia/genetics
6.
J Pharmacol Exp Ther ; 372(1): 73-82, 2020 01.
Article in English | MEDLINE | ID: mdl-31771994

ABSTRACT

Itch stimuli are detected by specialized primary afferents that convey the signal to the spinal cord, but how itch transmission is regulated is still not completely known. Here, we investigated the roles of the neuropeptide Y (NPY)/Y2 receptor system on scratch behavior. The inhibitory Y2 receptor is expressed on mouse primary afferents, and intrathecal administration of the Y2 agonist peptide YY (PYY)3-36 reduced scratch episode frequency and duration induced by compound 48/80, an effect that could be reversed by intrathecal preadministration of the Y2 antagonist BIIE0246. Also, scratch episode duration induced by histamine could be reduced by PYY3-36 In contrast, scratch behavior induced by α-methyl-5HT, protease-activated receptor-2-activating peptide SLIGRL, chloroquine, topical dust mite extract, or mechanical itch induced by von Frey filaments was unaffected by stimulation of Y2 Primary afferent neurons expressing the Npy2r gene were found to coexpress itch-associated markers such as natriuretic peptide precursor b, oncostatin M receptor, and interleukin (IL) 31 receptor A. Accordingly, intrathecal PYY3-36 reduced the scratch behavior induced by IL-31. Our findings imply that the NPY/Y2 system reduces histaminergic and IL-31-associated itch through presynaptic inhibition of a subpopulation of itch-associated primary afferents. SIGNIFICANCE STATEMENT: The spinal neuropeptide Y system dampens scratching behavior induced by histaminergic compounds and interleukin 31, a cytokine involved in atopic dermatitis, through interactions with the Y2 receptor. The Y2 receptor is expressed by primary afferent neurons that are rich in itch-associated neurotransmitters and receptors such as somatostatin, natriuretic peptide precursor b, and interleukin 31 receptors.


Subject(s)
Antipruritics/pharmacology , Dermatitis, Atopic/metabolism , Neurons, Afferent/metabolism , Peptide Fragments/pharmacology , Peptide YY/pharmacology , Pruritus/metabolism , Receptors, Neuropeptide Y/metabolism , Animals , Antipruritics/administration & dosage , Antipruritics/therapeutic use , Arginine/analogs & derivatives , Arginine/toxicity , Benzazepines/toxicity , Cells, Cultured , Chloroquine/pharmacology , Dermatitis, Atopic/drug therapy , Ganglia, Spinal/cytology , Histamine/pharmacology , Histamine/toxicity , Interleukins/pharmacology , Interleukins/toxicity , Male , Mice , Mice, Inbred C57BL , Natriuretic Peptide, Brain/genetics , Natriuretic Peptide, Brain/metabolism , Neurons, Afferent/drug effects , Neurons, Afferent/physiology , Oligopeptides/pharmacology , Peptide Fragments/administration & dosage , Peptide Fragments/therapeutic use , Peptide YY/administration & dosage , Peptide YY/therapeutic use , Pruritus/drug therapy , Pruritus/etiology , Receptors, Neuropeptide Y/genetics , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism , Serotonin/pharmacology
7.
Biochem Biophys Res Commun ; 516(3): 951-956, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31272716

ABSTRACT

Oncostatin M (OSM) is a cytokine of the interleukin-6 family and plays a role in various disorders such as cancer and inflammatory diseases, which are often accompanied by skeletal muscle atrophy, or sarcopenia. However, the role of OSM in the regulation of skeletal muscle mass remains to be identified. In this study, we investigated the effect of OSM on C2C12 myotube formation in vitro. C2C12 myoblasts were induced to differentiate into myotubes for 3 days and then treated with OSM for 24 or 48 h. The diameter of differentiated C2C12 myotubes were reduced by 18.7% and 23.3% compared to control cells after treatment with OSM for 24 and 48 h, respectively. The expression levels of MyoD and myogenin were decreased, while those of atrogin-1, CCAAT/enhancer binding protein δ, and OSM receptor were increased in C2C12 myotubes treated with OSM for 24 h compared to control cells. Furthermore, the inhibitory effect of OSM on myotube formation was significantly attenuated by pretreatment with an inhibitor of signal transducer and activator of transcription (STAT) 3 or by knockdown of Stat3. Finally, the OSM-induced changes in the expression levels of MyoD, myogenin, and atrogin-1 were reversed by pretreatment with an inhibitor of STAT3 or by Stat3 knockdown in C2C12 myotubes. In conclusion, OSM induces C2C12 myotube atrophy by inhibiting myogenic differentiation and activating muscle degradation in a STAT3-dependent manner.


Subject(s)
Cell Differentiation/drug effects , Growth Inhibitors/pharmacology , Muscle Fibers, Skeletal/drug effects , Myoblasts/drug effects , Oncostatin M/pharmacology , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line, Transformed , Mice , Models, Biological , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Proteins/genetics , Muscle Proteins/metabolism , MyoD Protein/genetics , MyoD Protein/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Myogenin/genetics , Myogenin/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism , SKP Cullin F-Box Protein Ligases/genetics , SKP Cullin F-Box Protein Ligases/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Sarcopenia/chemically induced , Sarcopenia/genetics , Sarcopenia/metabolism , Sarcopenia/pathology , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
8.
Int J Mol Med ; 43(1): 209-220, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30431059

ABSTRACT

The present study aimed to investigate the effect of microRNA­183 (miR­183) on substantia nigra neurons by targeting oncostatin M receptor (OSMR) in a mouse model of Parkinson's disease (PD). The positive expression rates of OSMR and the apoptosis of substantia nigra neurons were detected by immunohistochemistry and terminal deoxynucleotidyl transferase­mediated dUTP­biotin nick end­labeling, respectively. Substantia nigra neurons in normal and PD mice were cultured in vitro. The association between miR­183 and OSMR was verified using a dual luciferase reporter gene assay. The expression of miR­183 and the phosphoinositide 3­kinase­Akt signaling pathway­associated genes were detected by reverse transcription­quantitative polymerase chain reaction and western blot analysis, respectively. Cell apoptosis was detected by flow cytometry. OSMR is the target gene of miR­183. The number of OSMR­positive cells and the apoptotic rate of substantia nigra neurons were increased in the PD group. Neurons transfected with miR­183 mimic exhibited elevated expression levels of miR­183, B­cell lymphoma 2 (Bcl­2)­associated X protein (Bax) and caspase­9 and increased apoptotic rate, and reduced expression levels of OSMR, Akt, phosphorylated (p­)Akt, glycogen synthase kinase­3 (GSK­3ß), p­GSK­3ß, Bcl­2, insulin­like growth factor 1 (IGF­1), mammalian target of rapamycin (mTOR) and p­mTOR. The miR­183 inhibitor decreased the expression levels of miR­183, Bax and caspase­9 and the apoptotic rate; however, increased the expression of OSMR, Akt, p­Akt, GSK­3ß, p­GSK­3ß, Bcl­2, IGF­1, mTOR and p­mTOR. The results of the present study provide evidence that the overexpression of miR­183 promotes the apoptosis of substantia nigra neurons by inhibiting the expression of OSMR.


Subject(s)
Apoptosis/genetics , MicroRNAs/genetics , Neurons/pathology , Parkinson Disease/genetics , Receptors, Oncostatin M/antagonists & inhibitors , Substantia Nigra/pathology , Animals , Base Sequence , Behavior, Animal , Caspase 9/metabolism , Disease Models, Animal , Insulin-Like Growth Factor I/metabolism , Male , Mice, Inbred C57BL , MicroRNAs/metabolism , Neurons/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism , TOR Serine-Threonine Kinases/metabolism , bcl-2-Associated X Protein/metabolism
9.
Theriogenology ; 125: 324-330, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30504073

ABSTRACT

Oncostatin M (OSM) and its receptor (OSMR) are members of the interleukin-6 family cytokines. Although OSM and OSMR expression was detected in human ovaries, their function and regulation during follicle development, ovulation and luteolysis have not been studied in any species. The aim of the present study was to investigate the levels of OSM and OSMR mRNA in bovine ovaries and the effect of OSM treatment on cultured granulosa cells. OSM mRNA was not detected in granulosa cells obtained from follicles around the time of follicular deviation and from pre-ovulatory follicles, whereas OSMR transcript levels were greater in granulosa cells of atretic subordinate follicles (P < 0.001). Abundance of OSMR mRNA increased in granulosa cells of preovulatory follicles, collected at 12 and 24 h after the ovulatory stimulus with gonadotropins (P < 0.001). In the luteal tissue, OSM mRNA abundance levels were higher at 24-48 h after PGF-induced luteolysis (P < 0.01) compared to 0 h, whereas OSMR mRNA was transiently increased at 2 h after PGF treatment (P < 0.05). In cultured granulosa cells, 10 ng/mL OSM in the presence of FSH increased BAX/BCL2 mRNA ratio (P < 0.05) compared to the control. Moreover, 100 ng/mL OSM in the presence of FSH increased OSMR (P < 0.05) and decreased XIAP mRNA (P < 0.05) levels, compared to the control group. These findings provide the first evidence that OSMR is regulated during follicle atresia, ovulation and luteolysis, and that OSM from other cells may mediate granulosa and luteal cell function, regulating the expression of genes involved in cell's viability.


Subject(s)
Gene Expression Regulation/physiology , Granulosa Cells/metabolism , Luteal Cells/metabolism , Oncostatin M/metabolism , RNA, Messenger/metabolism , Receptors, Oncostatin M/metabolism , Animals , Cattle , Cells, Cultured , Female , Luteolysis/physiology , Oncostatin M/genetics , Ovulation/physiology , RNA, Messenger/genetics , Receptors, Oncostatin M/genetics
10.
Mol Med ; 24(1): 28, 2018 06 05.
Article in English | MEDLINE | ID: mdl-30134804

ABSTRACT

BACKGROUND: Chronic autoimmune urticaria (CAU) is a common skin disease and remains unclear understanding of pathogenesis in the vast majority of cases. In order to explore a new therapy for CAU, the current study was performed to investigate the possible functioning of the Oncostatin M receptor (OSMR) gene in the autoimmunity of CAU via regulation of the JAK/STAT3 signaling pathway. METHODS: CAU skin tissues from 24 CAU patients and normal skin tissues from normal subjects were collected. Hematoxylin-eosin (HE) staining was conducted to count eosinophils, and immunohistochemistry was carried out to detect the positive rate of OSMR expression in two kinds of skin tissues. A total of 72 Kunming (KM) mice were selected, and 60 mice were used for establishing CAU models and later transfected with different plasmids. The expression of inflammatory factors was evaluated by enzyme-linked immunosorbent assays (ELISA). Expressions of janus kinase (JAK), signal transducer and activator of transcription 3 (STAT3), interferon-stimulated gene 15 (ISG15), CT10-regulated kinase (CRK), and interferon regulatory factor 9 (IRF9) were identified using Western blot assay and reverse transcription quantitative polymerase chain reaction (RT-qPCR). Epithelial cell proliferation was assessed by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT) assay, and cell cycle distribution and cell apoptosis were assessed using flow cytometry. RESULTS: The findings confirm that OSMR protein expression and histamine release rate are highly elevated in human CAU skin tissues, and the expression of the JAK/STAT3 signaling pathway-related genes (OSMR, JAK2, STAT3, ISG15, CRK and IRF9) was up-regulated. OSMR gene silencing in CAU mice significantly decreases the content of inflammatory factors (IL-1, IL-6, IFN-γ, and IgE), the number of eosinophils, and reduces the expression of the JAK/STAT3 signaling pathway related genes, and further enhances cell proliferation, promotes cell cycle entry and inhibits apoptosis of epithelial cells. CONCLUSION: All aforementioned results indicate that OSMR gene silencing inhibits the activation of the JAK/STAT3 signaling pathway, thereby suppressing the development of CAU.


Subject(s)
Autoimmune Diseases/genetics , Janus Kinases/metabolism , Receptors, Oncostatin M/genetics , STAT3 Transcription Factor/metabolism , Urticaria/genetics , Animals , Autoimmune Diseases/metabolism , Child , Child, Preschool , Chronic Disease , Female , Gene Silencing , Humans , Infant , Janus Kinases/genetics , Male , Mast Cells/metabolism , Mice , STAT3 Transcription Factor/genetics , Signal Transduction , Urticaria/metabolism
11.
Mol Cancer Ther ; 16(10): 2234-2245, 2017 10.
Article in English | MEDLINE | ID: mdl-28729401

ABSTRACT

Molecularly targeted drugs have yielded significant therapeutic advances in oncogene-driven non-small cell lung cancer (NSCLC), but a majority of patients eventually develop acquired resistance. Recently, the relation between proinflammatory cytokine IL6 and resistance to targeted drugs has been reported. We investigated the functional contribution of IL6 and the other members of IL6 family proinflammatory cytokine pathway to resistance to targeted drugs in NSCLC cells. In addition, we examined the production of these cytokines by cancer cells and cancer-associated fibroblasts (CAF). We also analyzed the prognostic significance of these molecule expressions in clinical NSCLC samples. In NSCLC cells with acquired resistance to targeted drugs, we observed activation of the IL6-cytokine pathway and STAT3 along with epithelial-to-mesenchymal transition (EMT) features. In particular, IL6 family cytokine oncostatin-M (OSM) induced a switch to the EMT phenotype and protected cells from targeted drug-induced apoptosis in OSM receptors (OSMRs)/JAK1/STAT3-dependent manner. The cross-talk between NSCLC cells and CAFs also preferentially activated the OSM/STAT3 pathway via a paracrine mechanism and decreased sensitivity to targeted drugs. The selective JAK1 inhibitor filgotinib effectively suppressed STAT3 activation and OSMR expression, and cotargeting inhibition of the oncogenic pathway and JAK1 reversed resistance to targeted drugs. In the analysis of clinical samples, OSMR gene expression appeared to be associated with worse prognosis in patients with surgically resected lung adenocarcinoma. Our data suggest that the OSMRs/JAK1/STAT3 axis contributes to resistance to targeted drugs in oncogene-driven NSCLC cells, implying that this pathway could be a therapeutic target. Mol Cancer Ther; 16(10); 2234-45. ©2017 AACR.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Janus Kinase 1/genetics , Molecular Targeted Therapy , Oncostatin M/genetics , STAT3 Transcription Factor/genetics , Aged , Apoptosis/drug effects , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cytokines/antagonists & inhibitors , Cytokines/genetics , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Janus Kinase 1/antagonists & inhibitors , Male , Middle Aged , Neoplasm Staging , Oncostatin M/antagonists & inhibitors , Receptors, Oncostatin M/genetics , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects
12.
Cell Death Differ ; 23(6): 1073-85, 2016 06.
Article in English | MEDLINE | ID: mdl-26768665

ABSTRACT

Atopic dermatitis (AD) is the most common inflammatory skin disease with no well-delineated cause or effective cure. Here we show that the p53 family member p63, specifically the ΔNp63, isoform has a key role in driving keratinocyte activation in AD. We find that overexpression of ΔNp63 in transgenic mouse epidermis results in a severe skin phenotype that shares many of the key clinical, histological and molecular features associated with human AD. This includes pruritus, epidermal hyperplasia, aberrant keratinocyte differentiation, enhanced expression of selected cytokines and chemokines and the infiltration of large numbers of inflammatory cells including type 2 T-helper cells - features that are highly representative of AD dermatopathology. We further demonstrate several of these mediators to be direct transcriptional targets of ΔNp63 in keratinocytes. Of particular significance are two p63 target genes, IL-31 and IL-33, both of which are key players in the signaling pathways implicated in AD. Importantly, we find these observations to be in good agreement with elevated levels of ΔNp63 in skin lesions of human patients with AD. Our studies reveal an important role for ΔNp63 in the pathogenesis of AD and offer new insights into its etiology and possible therapeutic targets.


Subject(s)
Dermatitis, Atopic/pathology , Interleukin-33/metabolism , Phosphoproteins/metabolism , Trans-Activators/metabolism , Animals , Cytokines/metabolism , Dermatitis, Atopic/metabolism , Enzyme-Linked Immunosorbent Assay , Epidermis/metabolism , Humans , Immunoglobulin E/blood , Interleukins/genetics , Interleukins/metabolism , Mice , Mice, Transgenic , Microscopy, Fluorescence , Phenotype , Phosphoproteins/genetics , Protein Binding , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism , Signal Transduction , Skin/metabolism , Skin/pathology , Th2 Cells/cytology , Th2 Cells/immunology , Th2 Cells/metabolism , Trans-Activators/genetics
13.
Apoptosis ; 20(9): 1150-63, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26093524

ABSTRACT

Oncostatin M (OSM) exhibits many unique biological activities by activating the Oß receptor. However, its role in myocardial ischemia/reperfusion injury (I/R injury) in mice remains unknown. We investigated whether Notch3/Akt signaling is involved in the regulation of OSM-induced protection against cardiac I/R injury. The effects of OSM were assessed in mice that underwent myocardial I/R injury by OSM treatment or by genetic deficiency of the OSM receptor Oß. We investigated its effects on cardiomyocyte apoptosis and mitochondrial biogenesis and whether Notch3/Akt signaling was involved in the regulation of OSM-induced protection against cardiac I/R injury. The mice underwent 30 min of ischemia followed by 3 h of reperfusion and were randomized to be treated with Notch3 siRNA (siNotch3) or lentivirus carrying Notch3 cDNA (Notch3) 72 h before coronary artery ligation. Myocardial infarct size, cardiac function, cardiomyocyte apoptosis and mitochondria morphology in mice that underwent cardiac I/R injury were compared between groups. OSM alleviated cardiac I/R injury by inhibiting cardiomyocyte apoptosis through promotion of Notch3 production, thus activating the PI3K/Akt pathway. OSM enhanced mitochondrial biogenesis and mitochondrial function in mice subjected to cardiac I/R injury. In contrast, OSM receptor Oß knock out exacerbated cardiac I/R injury, decreased Notch3 production, enhanced cardiomyocyte apoptosis, and impaired mitochondrial biogenesis in cardiac I/R injured mice. The mechanism of OSM on cardiac I/R injury is partly mediated by the Notch3/Akt pathway. These results suggest a novel role of Notch3/Akt signaling that contributes to OSM-induced protection against cardiac I/R injury.


Subject(s)
Growth Inhibitors/pharmacology , Myocardial Reperfusion Injury/prevention & control , Oncostatin M/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/drug effects , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac , RNA, Small Interfering/metabolism , Rats , Receptor, Notch3 , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism
14.
J Cell Mol Med ; 19(6): 1296-307, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25752217

ABSTRACT

Oncostatin M (OSM) exhibits many unique biological activities by activating Oß receptor. However, its role in myocardial I/R injury in diabetic mice remains unknown. The involvement of OSM was assessed in diabetic mice which underwent myocardial I/R injury by OSM treatment or genetic deficiency of OSM receptor Oß. Its mechanism on cardiomyocyte apoptosis, mitochondrial biogenesis and insulin sensitivity were further studied. OSM alleviated cardiac I/R injury by inhibiting cardiomyocyte apoptosis through inhibition of inositol pyrophosphate 7 (IP7) production, thus activating PI3K/Akt/BAD pathway, decreasing Bax expression while up-regulating Bcl-2 expression and decreasing the ratio of Bax to Bcl-2 in db/db mice. OSM enhanced mitochondrial biogenesis and mitochondrial function in db/db mice subjected to cardiac I/R injury. On the contrary, OSM receptor Oß knockout exacerbated cardiac I/R injury, increased IP7 production, enhanced cardiomyocyte apoptosis, impaired mitochondrial biogenesis, glucose homoeostasis and insulin sensitivity in cardiac I/R injured diabetic mice. Inhibition of IP7 production by TNP (IP6K inhibitor) exerted similar effects of OSM. The mechanism of OSM on cardiac I/R injury in diabetic mice is partly associated with IP7/Akt and adenine mononucleotide protein kinase/PGC-1α pathway. OSM protects against cardiac I/R Injury by regulating apoptosis, insulin sensitivity and mitochondrial biogenesis in diabetic mice through inhibition of IP7 production.


Subject(s)
Apoptosis/drug effects , Diabetes Mellitus, Experimental/physiopathology , Insulin Resistance , Myocardial Reperfusion Injury/prevention & control , Oncostatin M/pharmacology , Animals , Apoptosis/genetics , Blotting, Western , Diabetes Mellitus, Experimental/genetics , Glucose/metabolism , Homeostasis/genetics , Inositol Phosphates/antagonists & inhibitors , Inositol Phosphates/metabolism , Mice, 129 Strain , Mice, Knockout , Mice, Mutant Strains , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/physiopathology , Myocardium/metabolism , Myocardium/pathology , Organelle Biogenesis , Protective Agents/pharmacology , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism
15.
Neoplasia ; 17(2): 225-37, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25748242

ABSTRACT

Glioblastoma (GBM), the most malignant of the brain tumors is classified on the basis of molecular signature genes using TCGA data into four subtypes- classical, mesenchymal, proneural and neural. The mesenchymal phenotype is associated with greater aggressiveness and low survival in contrast to GBMs enriched with proneural genes. The proinflammatory cytokines secreted in the microenvironment of gliomas play a key role in tumor progression. The study focused on the role of Oncostatin-M (OSM), an IL-6 family cytokine in inducing mesenchymal properties in GBM. Analysis of TCGA and REMBRANDT data revealed that expression of OSMR but not IL-6R or LIFR is upregulated in GBM and has negative correlation with survival. Amongst the GBM subtypes, OSMR level was in the order of mesenchymal > classical > neural > proneural. TCGA data and RT-PCR analysis in primary cultures of low and high grade gliomas showed a positive correlation between OSMR and mesenchymal signature genes-YKL40/CHI3L1, fibronectin and vimentin and a negative correlation with proneural signature genes-DLL3, Olig2 and BCAN. OSM enhanced transcript and protein level of fibronectin and YKL-40 and reduced the expression of Olig2 and DLL3 in GBM cells. OSM-regulated mesenchymal phenotype was associated with enhanced MMP-9 activity, increased cell migration and invasion. Importantly, OSM induced mesenchymal markers and reduced proneural genes even in primary cultures of grade-III glioma cells. We conclude that OSM-mediated signaling contributes to aggressive nature associated with mesenchymal features via STAT3 signaling in glioma cells. The data suggest that OSMR can be explored as potential target for therapeutic intervention.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Mesenchymal Stem Cells/metabolism , Oncostatin M/physiology , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Blotting, Western , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation/physiology , Glioma/pathology , Humans , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Phenotype , Real-Time Polymerase Chain Reaction/methods , Receptors, Interleukin-6/genetics , Receptors, Oncostatin M/genetics
16.
Arthritis Rheumatol ; 66(9): 2391-402, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24839265

ABSTRACT

OBJECTIVE: To examine the impact of the gp130 cytokine family on murine articular cartilage and to explore a potential regulatory role of suppressor of cytokine signaling 3 (SOCS-3) in murine chondrocytes. METHODS: In wild-type (WT) mouse chondrocytes, baseline receptor expression levels and gp130 cytokine-induced JAK/STAT signaling were determined by flow cytometry, and expression of SOCS-3 was assessed by quantitative polymerase chain reaction. The role of endogenous SOCS-3 was examined in cartilage explants and chondrocytes from mice with conditional deletion of Socs3 driven by the Col2a1 promoter in vitro (Socs3(Δ/Δcol2) ) and from mice during CD4+ T cell-dependent inflammatory monarthritis. Bone erosions in the murine joints were analyzed by micro-computed tomography. RESULTS: On chondrocytes from WT mice, gp130 and the oncostatin M (OSM) receptor were strongly expressed, whereas the transmembrane interleukin-6 (IL-6) receptor was expressed at much lower levels. Compared to other gp130 cytokines, OSM was the most potent activator of the JAK/STAT pathway and of SOCS-3 induction. Treatment of Socs3(Δ/Δcol2) mouse cartilage explants and chondrocytes with gp130 cytokines prolonged JAK/STAT signaling, enhanced cartilage degradation, increased the expression of Adamts4, Adamts5, and RANKL, and elevated the production of IL-6, granulocyte colony-stimulating factor, CXCL1, and CCL2. Socs3(Δ/Δcol2) mice developed exacerbated inflammation and joint damage in response to gp130 cytokine injections, and these histopathologic features were also observed in mice with inflammatory monarthritis. CONCLUSION: The results of this study highlight a key role for SOCS-3 in regulating chondrocyte responses during inflammatory arthritis. Within the gp130 cytokine family, OSM is a potent stimulus of chondrocyte responses, while IL-6 probably signals via trans-signaling. The gp130 cytokine-driven production of RANKL in chondrocytes may link chondrocyte activation and bone remodeling during inflammatory arthritis. Thus, these findings suggest that the inhibition of OSM might reduce the development and severity of structural joint damage during inflammatory arthritis.


Subject(s)
Arthritis, Experimental/metabolism , Chondrocytes/metabolism , Cytokine Receptor gp130/metabolism , Signal Transduction/physiology , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Cartilage, Articular/metabolism , Cytokine Receptor gp130/genetics , Knee Joint/metabolism , Mice , Mice, Knockout , Oncostatin M/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics
17.
Cell Mol Life Sci ; 71(10): 1907-16, 2014 May.
Article in English | MEDLINE | ID: mdl-24322910

ABSTRACT

Cardiomyocytes continuously generate the contractile force to circulate blood through the body. Imbalances in contractile performance or energy supply cause adaptive responses of the heart resulting in adverse rearrangement of regular structures, which in turn might lead to heart failure. At the cellular level, cardiomyocyte remodeling includes (1) restructuring of the contractile apparatus; (2) rearrangement of the cytoskeleton; and (3) changes in energy metabolism. Dedifferentiation represents a key feature of cardiomyocyte remodeling. It is characterized by reciprocal changes in the expression pattern of "mature" and "immature" cardiomyocyte-specific genes. Dedifferentiation may enable cardiomyocytes to cope with hypoxic stress by disassembly of the energy demanding contractile machinery and by reduction of the cellular energy demand. Dedifferentiation during myocardial repair might provide cardiomyocytes with additional plasticity, enabling survival under hypoxic conditions and increasing the propensity to enter the cell cycle. Although dedifferentiation of cardiomyocytes has been described during tissue regeneration in zebrafish and newts, little is known about corresponding mechanisms and regulatory circuits in mammals. The recent finding that the cytokine oncostatin M (OSM) is pivotal for cardiomyocyte dedifferentiation and exerts strong protective effects during myocardial infarction highlights the role of cytokines as potent stimulators of cardiac remodeling. Here, we summarize the current knowledge about transient dedifferentiation of cardiomyocytes in the context of myocardial remodeling, and propose a model for the role of OSM in this process.


Subject(s)
Heart/physiology , Myocytes, Cardiac/cytology , Animals , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cell Dedifferentiation , Humans , Myocytes, Cardiac/metabolism , Oncostatin M/metabolism , Receptors, Oncostatin M/antagonists & inhibitors , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism , Regeneration , Ventricular Remodeling
18.
J Clin Endocrinol Metab ; 99(2): E217-25, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24297795

ABSTRACT

CONTEXT: Adipose tissue is a highly active endocrine organ that secretes many factors that affect other tissues and whole-body metabolism. Adipocytes are responsive to several glycoprotein 130 (gp130) cytokines, some of which have been targeted as potential antiobesity therapeutics. OBJECTIVE: Oncostatin M (OSM) is a gp130 family member known to inhibit adipocyte differentiation in vitro, but its effects on other adipocyte properties are not characterized. The expression of OSM in white adipose tissue (WAT) has not been evaluated in the context of obesity. Thus, our objective was to examine the expression of adipose tissue OSM in obese animals and humans. DESIGN: OSM expression was examined in adipose tissues from mice with diet-induced and genetic obesity and in obese humans as well as in fractionated adipose tissue from mice. Murine adipocytes were used to examine OSM receptor expression and the effects of OSM on adipocytes, including the secretion of factors such as plasminogen activator inhibitor 1 and IL-6, which are implicated in metabolic diseases. RESULTS: OSM expression is increased in rodent and human obesity/type 2 diabetes mellitus. In humans, OSM levels correlate with body weight and insulin and are inversely correlated with glucose disposal rate as measured by hyperinsulinemic-euglycemic clamp. OSM is not produced from the adipocytes in WAT but derives from cells in the stromovascular fraction, including F4/80(+) macrophages. The specific receptor of OSM, OSM receptor-ß, is expressed in adipocytes and adipose tissue and increased in both rodent models of obesity examined. OSM acts on adipocytes to induce the expression and secretion of plasminogen activator inhibitor 1 and IL-6. CONCLUSIONS: These data indicate that WAT macrophages are a source of OSM and that OSM levels are significantly induced in murine and human obesity/type 2 diabetes mellitus. These studies suggest that OSM produced from immune cells in WAT acts in a paracrine manner on adipocytes to promote a proinflammatory phenotype in adipose tissue.


Subject(s)
Adipose Tissue/metabolism , Diabetes Mellitus, Type 2/metabolism , Obesity/metabolism , Oncostatin M/metabolism , Receptors, Oncostatin M/metabolism , 3T3-L1 Cells , Animals , Diabetes Mellitus, Type 2/genetics , Gene Expression Regulation , Humans , Male , Mice , Obesity/genetics , Oncostatin M/genetics , Receptors, Oncostatin M/genetics
19.
PLoS One ; 7(8): e43155, 2012.
Article in English | MEDLINE | ID: mdl-22937020

ABSTRACT

Evaluation of a pathophysiological role of the interleukin-6-type cytokine oncostatin M (OSM) for human diseases has been complicated by the fact that mouse models of diseases targeting either OSM or the OSM receptor (OSMR) complex cannot fully reflect the human situation. This is due to earlier findings that human OSM utilizes two receptor complexes, glycoprotein 130 (gp130)/leukemia inhibitory factor receptor (LIFR) (type I) and gp130/OSMR (type II), both with wide expression profiles. Murine OSM on the other hand only binds to the gp130/OSMR (type II) receptor complex with high affinity. Here, we characterize the receptor usage for rat OSM. Using different experimental approaches (knock-down of the OSMR expression by RNA interference, blocking of the LIFR by LIF-05, an antagonistic LIF variant and stably transfected Ba/F3 cells) we can clearly show that rat OSM surprisingly utilizes both, the type I and type II receptor complex, therefore mimicking the human situation. Furthermore, it displays cross-species activities and stimulates cells of human as well as murine origin. Its signaling capacities closely mimic those of human OSM in cell types of different origin in the way that strong activation of the Jak/STAT, the MAP kinase as well as the PI3K/Akt pathways can be observed. Therefore, rat disease models would allow evaluation of the relevance of OSM for human biology.


Subject(s)
Receptors, Cytokine/metabolism , Receptors, Oncostatin M/metabolism , Animals , Blotting, Western , Cell Line , Cell Proliferation , Cells, Cultured , Humans , Mice , Protein Binding , RNA, Small Interfering , Rats , Receptors, Cytokine/genetics , Receptors, Oncostatin M/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology
20.
Mol Hum Reprod ; 18(2): 88-95, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21965347

ABSTRACT

Fetal ovarian development and primordial follicle formation are imperative for adult fertility in the female. Data suggest the interleukin (IL)6-type cytokines, leukaemia inhibitory factor (LIF), IL6, oncostatin M (OSM) and ciliary neurotrophic factor (CNTF), are able to regulate the survival, proliferation and differentiation of fetal murine germ cells (GCs) in vivo and in vitro. We postulated that these factors may play a similar role during early human GC development and primordial follicle formation. To test this hypothesis, we have investigated the expression and regulation of IL6-type cytokines, using quantitative reverse transcription polymerase chain reaction and immunohistochemistry. Expression of transcripts encoding OSM increased significantly across the gestational range examined (8-20 weeks), while expression of IL6 increased specifically between the first (8-11 weeks) and early second (12-16 weeks) trimesters, co-incident with the initiation of meiosis. LIF and CNTF expression remained unchanged. Expression of the genes encoding the LIF and IL6 receptors, and their common signalling subunit gp130, was also found to be developmentally regulated, with expression increasing significantly with increasing gestation. LIF receptor and gp130 proteins localized exclusively to GCs, including oocytes in primordial follicles, indicating this cell type to be the sole target of IL6-type cytokine signalling in the human fetal ovary. These data establish that IL6-type cytokines and their receptors are expressed in the human fetal ovary and may directly influence GC development at multiple stages of maturation.


Subject(s)
Gene Expression Regulation, Developmental , Oocytes/metabolism , Ovarian Follicle/metabolism , RNA, Messenger/biosynthesis , Signal Transduction/genetics , Adult , Ciliary Neurotrophic Factor/genetics , Ciliary Neurotrophic Factor/metabolism , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Female , Fetus , Gestational Age , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Leukemia Inhibitory Factor/genetics , Leukemia Inhibitory Factor/metabolism , Oncostatin M/genetics , Oncostatin M/metabolism , Oocytes/growth & development , Ovarian Follicle/growth & development , Pregnancy , Pregnancy Trimesters , Real-Time Polymerase Chain Reaction , Receptor, Ciliary Neurotrophic Factor/genetics , Receptor, Ciliary Neurotrophic Factor/metabolism , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...