Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Int J Radiat Oncol Biol Phys ; 118(1): 203-217, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37610394

RESUMO

PURPOSE: Radiation-induced heart fibrosis (RIHF) is a severe consequence of radiation-induced heart damage (RIHD) leading to impaired cardiac function. The involvement of oncostatin M (OSM) and its receptor (OSMR) in RIHD remains unclear. This study aimed to investigate the specific mechanism of OSM/OSMR in RIHF/RIHD. METHODS AND MATERIALS: RNA sequencing was performed on heart tissues from a RIHD mouse model. OSM levels were assessed in serum samples obtained from patients receiving thoracic radiation therapy (RT), as well as in RIHF mouse heart tissues and serum using enzyme-linked immunosorbent assay. Fiber activation was evaluated through costimulation of primary cardiac fibroblasts and NIH3T3 cells with RT and OSM, using Western blotting, immunofluorescence, and quantitative Polymerase Chain Reaction (qPCR). Adeno-associated virus serotype 9-mediated overexpression or silencing of OSM specifically in the heart was performed in vivo to assess cardiac fibrosis levels by transthoracic echocardiography and pathologic examination. The regulatory mechanism of OSM on the transcription level of SMAD4 was further explored in vitro using mass spectrometric analysis, chromatin immunoprecipitation-qPCR, and DNA pull-down. RESULTS: OSM levels were elevated in the serum of patients after thoracic RT as well as in RIHF mouse cardiac endothelial cells and mouse serum. The OSM rate (post-RT/pre-RT) and the heart exposure dose in RT patients showed a positive correlation. Silencing OSMR in RIHF mice reduced fibrosis, while OSMR overexpression increased fibrotic responses. Furthermore, increased OSM promoted histone acetylation (H3K27ac) in the SMAD4 promoter region, influencing SMAD4 transcription and subsequently enhancing fibrotic response. CONCLUSIONS: The findings demonstrated that OSM/OSMR signaling promotes SMAD4 transcription in cardiac fibroblasts through H3K27 hyperacetylation, thereby promoting radiation-induced cardiac fibrosis and manifestations of RIHD.


Assuntos
Células Endoteliais , Fibroblastos , Animais , Humanos , Camundongos , Fibroblastos/metabolismo , Fibrose , Células NIH 3T3 , Oncostatina M/genética , Oncostatina M/metabolismo , Oncostatina M/farmacologia , Receptores de Oncostatina M/metabolismo , Proteína Smad4
2.
Nature ; 617(7962): 827-834, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37165186

RESUMO

Skeletal muscle atrophy is a hallmark of the cachexia syndrome that is associated with poor survival and reduced quality of life in patients with cancer1. Muscle atrophy involves excessive protein catabolism and loss of muscle mass and strength2. An effective therapy against muscle wasting is currently lacking because mechanisms driving the atrophy process remain incompletely understood. Our gene expression analysis in muscle tissues indicated upregulation of ectodysplasin A2 receptor (EDA2R) in tumour-bearing mice and patients with cachectic cancer. Here we show that activation of EDA2R signalling promotes skeletal muscle atrophy. Stimulation of primary myotubes with the EDA2R ligand EDA-A2 triggered pronounced cellular atrophy by induction of the expression of muscle atrophy-related genes Atrogin1 and MuRF1. EDA-A2-driven myotube atrophy involved activation of the non-canonical NFĸB pathway and was dependent on NFκB-inducing kinase (NIK) activity. Whereas EDA-A2 overexpression promoted muscle wasting in mice, deletion of either EDA2R or muscle NIK protected tumour-bearing mice from loss of muscle mass and function. Tumour-induced oncostatin M (OSM) upregulated muscle EDA2R expression, and muscle-specific oncostatin M receptor (OSMR)-knockout mice were resistant to tumour-induced muscle wasting. Our results demonstrate that EDA2R-NIK signalling mediates cancer-associated muscle atrophy in an OSM-OSMR-dependent manner. Thus, therapeutic targeting of these pathways may be beneficial in prevention of muscle loss.


Assuntos
Caquexia , Atrofia Muscular , Neoplasias , Transdução de Sinais , Receptor Xedar , Animais , Camundongos , Caquexia/complicações , Caquexia/etiologia , Caquexia/metabolismo , Caquexia/patologia , Fibras Musculares Esqueléticas/metabolismo , Atrofia Muscular/etiologia , Atrofia Muscular/metabolismo , Atrofia Muscular/patologia , Atrofia Muscular/prevenção & controle , Neoplasias/complicações , Neoplasias/metabolismo , Neoplasias/patologia , Receptor Xedar/metabolismo , Humanos , Ligantes , Receptores de Oncostatina M/metabolismo , Oncostatina M/metabolismo , Quinase Induzida por NF-kappaB
3.
Mol Biol Rep ; 50(5): 4735-4741, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36929287

RESUMO

BACKGROUND: The human oncostatin M receptor subunit , commonly known as the oncostatin M receptor (OSMR), is a cell surface protein and belongs to the family of type I cytokine receptors. It is highly expressed in several cancers and is a potential therapeutic target. Structurally, OSMR consists of three major domains: the extracellular, transmembrane, and cytoplasmic domains. The extracellular domain further comprises four Type III fibronectin subdomains. The functional relevance of these type III fibronectin domains is not known yet, and it is of great interest to us to understand their role in OSMR-mediated interactions with other oncogenic proteins. METHODS & RESULTS: The four type III fibronectin domains of hOSMR were amplified by PCR using the pUNO1-hOSMR construct as a template. The molecular size of the amplified products was confirmed by agarose gel electrophoresis. The amplicons were then cloned into a pGEX4T3 vector containing GST as an N-terminal tag. Positive clones with domain inserts were identified by restriction digestion and overexpressed in E. coli Rosetta (DE3) cells. The optimum conditions for overexpression were found to be 1 mM IPTG and an incubation temperature of 37 °C. The overexpression of the fibronectin domains was confirmed by SDS-PAGE, and they are affinity purified by using glutathione agarose beads in three repetitive steps. The purity of the isolated domains analyzed by SDS-PAGE and western blotting showed that they were exactly at their corresponding molecular weights as a single distinct band. CONCLUSION: In this study, we have successfully cloned, expressed, and purified four Type III fibronectin subdomains of hOSMR.


Assuntos
Escherichia coli , Fibronectinas , Humanos , Fibronectinas/genética , Fibronectinas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Western Blotting , Receptores de Oncostatina M/metabolismo , Clonagem Molecular
4.
J Oral Pathol Med ; 52(2): 136-144, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36207791

RESUMO

BACKGROUND: Oncostatin M receptor is an interleukin 6 receptor with great influence on inflammation and cancer progression. However, the function of oncostatin M receptor in oral squamous cell carcinoma remains unknown. METHODS: Oncostatin M receptor expression was explored with TIMER and TCGA databases. The mRNA and protein expressions of oncostatin M receptor were detected in oral tissues. The association between oncostatin M receptor expression and clinicopathological characteristics was analyzed, and the prognostic value of oncostatin M receptor was determined. Immune statues of oncostatin M receptor were analyzed by TIMER and TISIDB. The underlying mechanisms of oncostatin M receptor in oral squamous cell carcinoma was also explored preliminarily. RESULTS: Oncostatin M receptor was dysregulated in many cancers. Both mRNA and protein levels of oncostatin M receptor in oral squamous cell carcinoma tissues were significantly higher than that in normal oral tissues. Oncostatin M receptor expression was connected to differentiation, lymph node metastasis, tumor node metastasis (TNM) stage, perineural invasion and vascular invasion. Oncostatin M receptor expression was an independent prognostic factor associated with overall survivals. Oncostatin M receptor expression was significantly related to CD8+ T cell and interleukin 6 receptor. High oncostatin M receptor expression was associated with focal adhesion, extracellular matrix (ECM) receptor interaction, and JAK/STAT signaling pathway. CONCLUSION: Oncostatin M receptor was overexpressed in oral squamous cell carcinoma and related to overall survival. Oncostatin M receptor expression has potential to become an effective prognostic biomarker for oral squamous cell carcinoma patients.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Bucais , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço , Carcinoma de Células Escamosas/patologia , Receptores de Oncostatina M/metabolismo , Transdução de Sinais , Prognóstico , RNA Mensageiro
5.
Int J Mol Sci ; 23(19)2022 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-36232392

RESUMO

Oncostatin M (OSM) is a member of the interleukin-6 (IL-6) family of cytokines and can bind two different receptors, Leukemia inhibitory factor receptor (LIFR) and Oncostatin M receptor (OSMR), through a complex containing the common glycoprotein 130 (gp130) subunit [...].


Assuntos
Citocinas , Interleucina-6 , Receptor gp130 de Citocina/metabolismo , Interleucina-6/metabolismo , Oncostatina M/metabolismo , Receptores de Citocinas/metabolismo , Receptores de OSM-LIF , Receptores de Oncostatina M/metabolismo
6.
J Pathol Clin Res ; 8(3): 279-293, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35289095

RESUMO

Pure squamous cell carcinoma (SCC) is the most common pure variant form of bladder cancer, found in 2-5% of cases. It often presents late and is unresponsive to cisplatin-based chemotherapy. The molecular features of these tumours have not been elucidated in detail. We carried out whole-exome sequencing (WES), copy number, and transcriptome analysis of bladder SCC. Muscle-invasive bladder cancer (MIBC) samples with no evidence of squamous differentiation (non-SD) were used for comparison. To assess commonality of features with urothelial carcinoma with SD, we examined data from SD samples in The Cancer Genome Atlas (TCGA) study of MIBC. TP53 was the most commonly mutated gene in SCC (64%) followed by FAT1 (45%). Copy number analysis revealed complex changes in SCC, many differing from those in samples with SD. Gain of 5p and 7p was the most common feature, and focal regions on 5p included OSMR and RICTOR. In addition to 9p deletions, we found some samples with focal gain of 9p24 containing CD274 (PD-L1). Loss of 4q35 containing FAT1 was found in many samples such that all but one sample analysed by WES had FAT1 mutation or deletion. Expression features included upregulation of oncostatin M receptor (OSMR), metalloproteinases, metallothioneins, keratinisation genes, extracellular matrix components, inflammatory response genes, stem cell markers, and immune response modulators. Exploration of differentially expressed transcription factors identified BNC1 and TFAP2A, a gene repressed by PPARG, as the most upregulated factors. Known urothelial differentiation factors were downregulated along with 72 Kruppel-associated (KRAB) domain-containing zinc finger family protein (KZFP) genes. Novel therapies are urgently needed for these tumours. In addition to upregulated expression of EGFR, which has been suggested as a therapeutic target in basal/squamous bladder cancer, we identified expression signatures that indicate upregulated OSMR and YAP/TAZ signalling. Preclinical evaluation of the effects of inhibition of these pathways alone or in combination is merited.


Assuntos
Carcinoma de Células Escamosas , Carcinoma de Células de Transição , Neoplasias da Bexiga Urinária , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Humanos , Subunidade beta de Receptor de Oncostatina M , Receptores de Oncostatina M/metabolismo , Bexiga Urinária/metabolismo , Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/genética
7.
Int J Mol Sci ; 23(3)2022 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-35163735

RESUMO

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.


Assuntos
Insuficiência Cardíaca , Interleucina-6 , Miócitos Cardíacos , Oncostatina M , Receptores de Oncostatina M , Animais , Receptor gp130 de Citocina/metabolismo , Humanos , Interleucina-6/metabolismo , Camundongos , Miócitos Cardíacos/metabolismo , Oncostatina M/metabolismo , Subunidade beta de Receptor de Oncostatina M , Receptores de Oncostatina M/genética , Receptores de Oncostatina M/metabolismo
8.
Nat Commun ; 12(1): 7336, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34921158

RESUMO

Pancreatic ductal adenocarcinoma (PDA) is a lethal malignancy with a complex microenvironment. Dichotomous tumour-promoting and -restrictive roles have been ascribed to the tumour microenvironment, however the effects of individual stromal subsets remain incompletely characterised. Here, we describe how heterocellular Oncostatin M (OSM) - Oncostatin M Receptor (OSMR) signalling reprograms fibroblasts, regulates tumour growth and metastasis. Macrophage-secreted OSM stimulates inflammatory gene expression in cancer-associated fibroblasts (CAFs), which in turn induce a pro-tumourigenic environment and engage tumour cell survival and migratory signalling pathways. Tumour cells implanted in Osm-deficient (Osm-/-) mice display an epithelial-dominated morphology, reduced tumour growth and do not metastasise. Moreover, the tumour microenvironment of Osm-/- animals exhibit increased abundance of α smooth muscle actin positive myofibroblasts and a shift in myeloid and T cell phenotypes, consistent with a more immunogenic environment. Taken together, these data demonstrate how OSM-OSMR signalling coordinates heterocellular interactions to drive a pro-tumourigenic environment in PDA.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Oncostatina M/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Receptores de Oncostatina M/metabolismo , Transdução de Sinais , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Comunicação Celular , Linhagem Celular Tumoral , Proliferação de Células , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Terapia de Imunossupressão , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/patologia , Masculino , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Células Estreladas do Pâncreas/metabolismo , Células Estreladas do Pâncreas/patologia , Microambiente Tumoral
9.
J Exp Med ; 218(8)2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34132743

RESUMO

The wound healing process that occurs after spinal cord injury is critical for maintaining tissue homeostasis and limiting tissue damage, but eventually results in a scar-like environment that is not conducive to regeneration and repair. A better understanding of this dichotomy is critical to developing effective therapeutics that target the appropriate pathobiology, but a major challenge has been the large cellular heterogeneity that results in immensely complex cellular interactions. In this study, we used single-cell RNA sequencing to assess virtually all cell types that comprise the mouse spinal cord injury site. In addition to discovering novel subpopulations, we used expression values of receptor-ligand pairs to identify signaling pathways that are predicted to regulate specific cellular interactions during angiogenesis, gliosis, and fibrosis. Our dataset is a valuable resource that provides novel mechanistic insight into the pathobiology of not only spinal cord injury but also other traumatic disorders of the CNS.


Assuntos
Comunicação Celular , Análise de Célula Única , Traumatismos da Medula Espinal/patologia , Angiopoietinas/metabolismo , Animais , Astrócitos/metabolismo , Quimiotaxia , Feminino , Fibroblastos/metabolismo , Fibrose , Gliose/complicações , Gliose/patologia , Inflamação/patologia , Interleucina-6/metabolismo , Ligantes , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Células Mieloides/patologia , Neuroglia/patologia , Oncostatina M/metabolismo , Receptores de Oncostatina M/metabolismo , Transdução de Sinais , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/imunologia , Fatores de Tempo , Transcriptoma/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
10.
Int J Mol Sci ; 23(1)2021 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-35008777

RESUMO

Oncostatin M (OSM) and leukemia inhibitory factor (LIF) signaling protects the heart after myocardial infarction (MI). In mice, oncostatin M receptor (OSMR) and leukemia inhibitory factor receptor (LIFR) are selectively activated by the respective cognate ligands while OSM activates both the OSMR and LIFR in humans, which prevents efficient translation of mouse data into potential clinical applications. We used an engineered human-like OSM (hlOSM) protein, capable to signal via both OSMR and LIFR, to evaluate beneficial effects on cardiomyocytes and hearts after MI in comparison to selective stimulation of either LIFR or OSMR. Cell viability assays, transcriptome and immunoblot analysis revealed increased survival of hypoxic cardiomyocytes by mLIF, mOSM and hlOSM stimulation, associated with increased activation of STAT3. Kinetic expression profiling of infarcted hearts further specified a transient increase of OSM and LIF during the early inflammatory phase of cardiac remodeling. A post-infarction delivery of hlOSM but not mOSM or mLIF within this time period combined with cardiac magnetic resonance imaging-based strain analysis uncovered a global cardioprotective effect on infarcted hearts. Our data conclusively suggest that a simultaneous and rapid activation of OSMR and LIFR after MI offers a therapeutic opportunity to preserve functional and structural integrity of the infarcted heart.


Assuntos
Cardiotônicos/metabolismo , Infarto do Miocárdio/prevenção & controle , Oncostatina M/metabolismo , Receptores de OSM-LIF/metabolismo , Animais , Hipóxia Celular/genética , Sobrevivência Celular , Células Cultivadas , Humanos , Cinética , Fator Inibidor de Leucemia/metabolismo , Camundongos , Contração Miocárdica , Infarto do Miocárdio/genética , Miócitos Cardíacos/metabolismo , Engenharia de Proteínas , Proteínas Proto-Oncogênicas c-myc/metabolismo , Receptores de Oncostatina M/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais , Especificidade da Espécie , Transcriptoma/genética
11.
Nat Commun ; 11(1): 4116, 2020 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-32807793

RESUMO

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.


Assuntos
Glioblastoma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Radiação Ionizante , Receptores de Oncostatina M/metabolismo , Animais , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos da radiação , Células Cultivadas , Imunofluorescência , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Humanos , Masculino , Camundongos , Camundongos SCID , NADH Desidrogenase/genética , NADH Desidrogenase/metabolismo , Células-Tronco Neoplásicas/efeitos da radiação , Oncostatina M/metabolismo , Estresse Oxidativo/efeitos da radiação , Receptores de Oncostatina M/genética , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos da radiação
12.
Acta Neuropathol Commun ; 8(1): 42, 2020 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-32248843

RESUMO

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.


Assuntos
Anexina A2/genética , Neoplasias Encefálicas/genética , Glioblastoma/genética , Subunidade beta de Receptor de Oncostatina M/genética , Fator de Transcrição STAT3/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Anexina A2/metabolismo , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Proliferação de Células/genética , Criança , Cães , Transição Epitelial-Mesenquimal/genética , Feminino , Técnicas de Silenciamento de Genes , Inativação Gênica , Glioblastoma/irrigação sanguínea , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Transplante de Neoplasias , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Subunidade beta de Receptor de Oncostatina M/metabolismo , Fenótipo , Receptores de Oncostatina M/genética , Receptores de Oncostatina M/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Taxa de Sobrevida , Hipóxia Tumoral/genética
13.
J Pharmacol Exp Ther ; 372(1): 73-82, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31771994

RESUMO

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.


Assuntos
Antipruriginosos/farmacologia , Dermatite Atópica/metabolismo , Neurônios Aferentes/metabolismo , Fragmentos de Peptídeos/farmacologia , Peptídeo YY/farmacologia , Prurido/metabolismo , Receptores de Neuropeptídeo Y/metabolismo , Animais , Antipruriginosos/administração & dosagem , Antipruriginosos/uso terapêutico , Arginina/análogos & derivados , Arginina/toxicidade , Benzazepinas/toxicidade , Células Cultivadas , Cloroquina/farmacologia , Dermatite Atópica/tratamento farmacológico , Gânglios Espinais/citologia , Histamina/farmacologia , Histamina/toxicidade , Interleucinas/farmacologia , Interleucinas/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Peptídeo Natriurético Encefálico/genética , Peptídeo Natriurético Encefálico/metabolismo , Neurônios Aferentes/efeitos dos fármacos , Neurônios Aferentes/fisiologia , Oligopeptídeos/farmacologia , Fragmentos de Peptídeos/administração & dosagem , Fragmentos de Peptídeos/uso terapêutico , Peptídeo YY/administração & dosagem , Peptídeo YY/uso terapêutico , Prurido/tratamento farmacológico , Prurido/etiologia , Receptores de Neuropeptídeo Y/genética , Receptores de Oncostatina M/genética , Receptores de Oncostatina M/metabolismo , Serotonina/farmacologia
14.
Biochem Biophys Res Commun ; 516(3): 951-956, 2019 08 27.
Artigo em Inglês | MEDLINE | ID: mdl-31272716

RESUMO

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.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Inibidores do Crescimento/farmacologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Mioblastos/efeitos dos fármacos , Oncostatina M/farmacologia , Animais , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Linhagem Celular Transformada , Camundongos , Modelos Biológicos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Proteína MyoD/genética , Proteína MyoD/metabolismo , Mioblastos/citologia , Mioblastos/metabolismo , Miogenina/genética , Miogenina/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Oncostatina M/genética , Receptores de Oncostatina M/metabolismo , Proteínas Ligases SKP Culina F-Box/genética , Proteínas Ligases SKP Culina F-Box/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Sarcopenia/induzido quimicamente , Sarcopenia/genética , Sarcopenia/metabolismo , Sarcopenia/patologia , Proteínas com Motivo Tripartido/genética , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo
15.
Front Immunol ; 10: 1165, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31191538

RESUMO

For a long time, the central nervous system (CNS) was believed to be an immune privileged organ. In the last decades, it became apparent that the immune system interacts with the CNS not only in pathological, but also in homeostatic situations. It is now clear that immune cells infiltrate the healthy CNS as part of immune surveillance and that immune cells communicate through cytokines with CNS resident cells. In pathological conditions, an enhanced infiltration of immune cells takes place to fight the pathogen. A well-known family of cytokines is the interleukin (IL)-6 cytokine family. All members are important in cell communication and cell signaling in the immune system. One of these members is oncostatin M (OSM), for which the receptor is expressed on several cells of the CNS. However, the biological function of OSM in the CNS is not studied in detail. Here, we briefly describe the general aspects related to OSM biology, including signaling and receptor binding. Thereafter, the current understanding of OSM during CNS homeostasis and pathology is summarized.


Assuntos
Sistema Nervoso Central/fisiologia , Oncostatina M/genética , Oncostatina M/metabolismo , Animais , Suscetibilidade a Doenças , Homeostase , Humanos , Receptores de Oncostatina M/metabolismo , Transdução de Sinais , Especificidade da Espécie
16.
Theriogenology ; 125: 324-330, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30504073

RESUMO

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.


Assuntos
Regulação da Expressão Gênica/fisiologia , Células da Granulosa/metabolismo , Células Lúteas/metabolismo , Oncostatina M/metabolismo , RNA Mensageiro/metabolismo , Receptores de Oncostatina M/metabolismo , Animais , Bovinos , Células Cultivadas , Feminino , Luteólise/fisiologia , Oncostatina M/genética , Ovulação/fisiologia , RNA Mensageiro/genética , Receptores de Oncostatina M/genética
17.
Int J Mol Med ; 43(1): 209-220, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30431059

RESUMO

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.


Assuntos
Apoptose/genética , MicroRNAs/genética , Neurônios/patologia , Doença de Parkinson/genética , Receptores de Oncostatina M/antagonistas & inibidores , Substância Negra/patologia , Animais , Sequência de Bases , Comportamento Animal , Caspase 9/metabolismo , Modelos Animais de Doenças , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/metabolismo , Neurônios/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Oncostatina M/genética , Receptores de Oncostatina M/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Proteína X Associada a bcl-2/metabolismo
18.
Br J Cancer ; 115(2): 212-22, 2016 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-27351213

RESUMO

BACKGROUND: Copy-number gain of the oncostatin-M receptor (OSMR) occurs frequently in cervical squamous cell carcinoma (SCC) and is associated with adverse clinical outcome. We previously showed that OSMR overexpression renders cervical SCC cells more sensitive to the major ligand oncostatin-M (OSM), which increases migration and invasion in vitro. We hypothesised that a major contribution to this phenotype would come from epithelial-mesenchymal transition (EMT). METHODS: We performed a comprehensive integrated study, involving in vitro cell line studies, in vivo animal models and numerous clinical samples from a variety of anatomical sites. RESULTS: In independent sets of cervical, head/neck and lung SCC tissues, OSMR expression levels correlated with multiple EMT-associated phenotypic markers and transcription factors. OSM treatment of OSMR overexpressing cervical SCC cells produced consistent EMT changes and increased tumour sphere formation in suspension culture. In a mouse model, OSMR overexpressing SCC cells treated with OSM showed significant increases in lung colonisation. The biological effects of exogenous OSM were mirrored by highly significant adverse overall survival in cervical SCCs with OSMR overexpression (N=251). CONCLUSIONS: OSM:OSMR interactions are able to induce EMT, increased cancer stem cell-like properties and enhanced lung colonisation in SCC cells. These changes are likely to contribute to the highly significant adverse outcome associated with OSMR overexpression in cervical SCCs.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Transição Epitelial-Mesenquimal , Receptores de Oncostatina M/metabolismo , Análise de Sobrevida , Neoplasias do Colo do Útero/metabolismo , Animais , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Feminino , Xenoenxertos , Humanos , Janus Quinase 2/metabolismo , Camundongos , Metástase Neoplásica , Fator de Transcrição STAT3/metabolismo , Neoplasias do Colo do Útero/patologia
19.
Cell Death Differ ; 23(6): 1073-85, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26768665

RESUMO

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.


Assuntos
Dermatite Atópica/patologia , Interleucina-33/metabolismo , Fosfoproteínas/metabolismo , Transativadores/metabolismo , Animais , Citocinas/metabolismo , Dermatite Atópica/metabolismo , Ensaio de Imunoadsorção Enzimática , Epiderme/metabolismo , Humanos , Imunoglobulina E/sangue , Interleucinas/genética , Interleucinas/metabolismo , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Fenótipo , Fosfoproteínas/genética , Ligação Proteica , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Receptores de Oncostatina M/genética , Receptores de Oncostatina M/metabolismo , Transdução de Sinais , Pele/metabolismo , Pele/patologia , Células Th2/citologia , Células Th2/imunologia , Células Th2/metabolismo , Transativadores/genética
20.
Apoptosis ; 20(9): 1150-63, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26093524

RESUMO

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.


Assuntos
Inibidores do Crescimento/farmacologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Oncostatina M/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mitocôndrias/efeitos dos fármacos , Infarto do Miocárdio/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos , RNA Interferente Pequeno/metabolismo , Ratos , Receptor Notch3 , Receptores de Oncostatina M/genética , Receptores de Oncostatina M/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA