Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Comput Biol Med ; 155: 106709, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36854228

RESUMEN

Small molecule inhibitors (SMIs) targeting oncostatin M (OSM) signaling pathway represent new therapeutics to combat cancer, inflammatory bowel disease (IBD) and CNS disease. Recently, the first-in-class SMI named SMI-10B that target OSM and block its interaction with receptor (OSMR) were reported. However, the binding pocket and interaction mode of the compound on OSM remain poorly understood, which hampering the rational design of SMIs that target OSM. Here, using SMI-10B as a probe, the multiple pockets on OSM for small molecules binding were extensively explored by unbiased molecular dynamics (MD) simulations. Then, the near-native structure of the complex was identified by molecular mechanics generalized Born surface area (MM/GBSA) binding energy funnel. Moreover, the binding stabilities of the protein-ligand complexes in near- and non-native conformations were verified by additional independent MD runs and absolute free energy perturbation (FEP) calculation. In summary, the unique feature of SMI-10B spontaneously binds to OSM characterized here not only provide detailed information for understanding the molecular mechanism of SMI-10B binding to OSM, but also will facilitate the rational design of novel and more potent SMIs to block OSM signaling.


Asunto(s)
Simulación de Dinámica Molecular , Subunidad beta del Receptor de Oncostatina M , Oncostatina M/metabolismo , Oncostatina M/farmacología , Subunidad beta del Receptor de Oncostatina M/química , Subunidad beta del Receptor de Oncostatina M/metabolismo , Unión Proteica , Transducción de Señal
2.
Stem Cell Res ; 63: 102842, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35714449

RESUMEN

Mutations in the tumor suppressor M receptor (OSMR) gene are associated with primary localized cutaneous amyloidosis (PLCA). Recently, we confirmed that OSMR loss-of-function mutations enhance epidermal keratinocyte differentiation via inactivation of the STAT5/KLF7 signaling. However, no disease model was available for PLCA. Accordingly, we generated an OSMR c.1538G > A mutant human embryonic stem cell line (SMUDHe010-A-82) using CRISPR/Cas9-mediated homologous recombination. The cell line preserves normal karyotype, pluripotency and the ability to differentiate into all three germ layers. Moreover, the cell line can be used to prepare human skin organoid, which may provide a disease model for PLCA.


Asunto(s)
Células Madre Embrionarias Humanas , Sistemas CRISPR-Cas/genética , Línea Celular , Recombinación Homóloga , Homocigoto , Células Madre Embrionarias Humanas/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Mutación/genética , Subunidad beta del Receptor de Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/metabolismo
3.
Stem Cell Res Ther ; 13(1): 278, 2022 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-35765036

RESUMEN

BACKGROUND: Oncostatin M receptor (OSMR), as one of the receptors for oncostatin M (OSM), has previously been shown to mediate the stimulatory role of OSM in osteoclastogenesis and bone resorption. However, it remains to be clarified whether and how OSMR affects the differentiation of osteoblasts. METHODS: The expression level of OSMR during osteoblast and adipocyte differentiation was examined. The role of OSMR in the differentiation was investigated using in vitro gain-of-function and loss-of-function experiments. The mechanisms by which OSMR regulates bone cell differentiation were explored. Finally, in vivo function of OSMR in cell fate determination and bone homeostasis was studied after transplantation of OSMR-silenced bone marrow stromal cells (BMSCs) to the marrow of ovariectomized mice. RESULTS: OSMR was regulated during osteogenic and adipogenic differentiation of marrow stromal progenitor cells and increased in the metaphysis of ovariectomized mice. OSMR suppressed osteogenic differentiation and stimulated adipogenic differentiation of progenitor cells. Mechanistic investigations showed that OSMR inhibited extracellular signal-regulated kinase (ERK) and autophagy signaling. The downregulation of autophagy, which was mediated by ERK inhibition, suppressed osteogenic differentiation of progenitor cells. Additionally, inactivation of ERK/autophagy signaling attenuated the stimulation of osteogenic differentiation induced by Osmr siRNA. Furthermore, transplantation of BMSCs in which OSMR was silenced to the marrow of mice promoted osteoblast differentiation, attenuated fat accumulation and osteoclast differentiation, and thereby relieved the osteopenic phenotype in the ovariectomized mice. CONCLUSIONS: Our study has for the first time established the direct role of OSMR in regulating osteogenic differentiation of marrow stromal progenitor cells through ERK-mediated autophagy signaling. OSMR thus contributes to bone homeostasis through dual regulation of osteoblasts and osteoclasts. It also suggests that OSMR may be a potential target for the treatment of metabolic disorders such as osteoporosis.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular , Sistema de Señalización de MAP Quinasas , Subunidad beta del Receptor de Oncostatina M , Osteoblastos , Osteogénesis , Animales , Autofagia/fisiología , Diferenciación Celular/fisiología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Ratones , Subunidad beta del Receptor de Oncostatina M/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo
4.
Biochem Biophys Res Commun ; 614: 114-119, 2022 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-35576682

RESUMEN

Oncostatin M receptor beta (OSMRß) mediates signaling of Oncostatin M (OSM) and interleukine-31 (IL-31), two key cytokines involved in many important biological processes including inflammation and cancer progression. More importantly, OSMRß might be a potential biomarker and therapeutic target for some diseases, such as inflammatory bowel disease, pruritus and ovarian cancer. In this study, soluble recombinant canine OSMRß (cOSMRß) was experimentally expressed as a native antigen to develop an effective cOSMRß-specific monoclonal antibody (mAb), 2O2, using hybridoma technology. It was demonstrated that 2O2 is able to detect OSMRß expressed on cell surface using immunofluorescence assay (IFA) and flow cytometry (FACS). This mAb exhibits very high binding affinity to cOSMRß with the KD and half-maximal effective concentration (EC50) values of 2.49 nM and 96.96 ng/ml, respectively. Meanwhile, it didn't show any cross-relativities with feline OSMRß (fOSMRß) and human OSMRß (hOSMRß). Moreover, we determined the binding epitope of 2O2, which localizes in the domain VI (DVI, amino acids 623-734) of cOSMRß. In conclusion, this novel mAb, 2O2, can be used in immunoassays, including IFA, FACS and enzyme-linked immunosorbent assay (ELISA) to facilitate studies in dogs.


Asunto(s)
Subunidad beta del Receptor de Oncostatina M , Transducción de Señal , Animales , Anticuerpos Monoclonales , Gatos , Perros , Inflamación , Ratones , Oncostatina M/metabolismo , Subunidad beta del Receptor de Oncostatina M/metabolismo , Prurito
5.
Int J Mol Sci ; 22(21)2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34769079

RESUMEN

The IL-6 family cytokine Oncostatin M (OSM) is involved in cell development, growth, hematopoiesis, inflammation, and cancer. Intriguingly, OSM has proliferative and antiproliferative effects depending on the target cell. The molecular mechanisms underlying these opposing effects are not fully understood. Previously, we found OSM upregulation in different myeloproliferative syndromes. However, OSM receptor (OSMR) expression was detected on stromal cells but not the malignant cells themselves. In the present study, we, therefore, investigated the effect of murine OSM (mOSM) on proliferation in stromal and fibroblast cell lines. We found that mOSM impairs the proliferation of bone marrow (BM) stromal cells, whereas fibroblasts responded to mOSM with increased proliferation. When we set out to reveal the mechanisms underlying these opposing effects, we detected increased expression of the OSM receptors OSMR and LIFR in stromal cells. Interestingly, Osmr knockdown and Lifr overexpression attenuated the OSM-mediated effect on proliferation in both cell lines indicating that mOSM affected the proliferation signaling mainly through the OSMR. Furthermore, mOSM induced activation of the JAK-STAT, PI3K-AKT, and MAPK-ERK pathways in OP9 and NIH/3T3 cells with differences in total protein levels between the two cell lines. Our findings offer new insights into the regulation of proliferation by mOSM.


Asunto(s)
Proliferación Celular , Fibroblastos/citología , Células Madre Mesenquimatosas/citología , Subunidad beta del Receptor de Oncostatina M/metabolismo , Oncostatina M/metabolismo , Animales , Línea Celular , Fibroblastos/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Células 3T3 NIH , Transducción de Señal
6.
Cancer Res ; 81(20): 5336-5352, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34380633

RESUMEN

Although patients with advanced ovarian cancer may respond initially to treatment, disease relapse is common, and nearly 50% of patients do not survive beyond five years, indicating an urgent need for improved therapies. To identify new therapeutic targets, we performed single-cell and nuclear RNA-seq data set analyses on 17 human ovarian cancer specimens, revealing the oncostatin M receptor (OSMR) as highly expressed in ovarian cancer cells. Conversely, oncostatin M (OSM), the ligand of OSMR, was highly expressed by tumor-associated macrophages and promoted proliferation and metastasis in cancer cells. Ovarian cancer cell lines and additional patient samples also exhibited elevated levels of OSMR when compared with other cell types in the tumor microenvironment or to normal ovarian tissue samples. OSMR was found to be important for ovarian cancer cell proliferation and migration. Binding of OSM to OSMR caused OSMR-IL6ST dimerization, which is required to produce oncogenic signaling cues for prolonged STAT3 activation. Human monoclonal antibody clones B14 and B21 directed to the extracellular domain of OSMR abrogated OSM-induced OSMR-IL6ST heterodimerization, promoted the internalization and degradation of OSMR, and effectively blocked OSMR-mediated signaling in vitro. Importantly, these antibody clones inhibited the growth of ovarian cancer cells in vitro and in vivo by suppressing oncogenic signaling through OSMR and STAT3 activation. Collectively, this study provides a proof of principle that anti-OSMR antibody can mediate disruption of OSM-induced OSMR-IL6ST dimerization and oncogenic signaling, thus documenting the preclinical therapeutic efficacy of human OSMR antagonist antibodies for immunotherapy in ovarian cancer. SIGNIFICANCE: This study uncovers a role for OSMR in promoting ovarian cancer cell proliferation and metastasis by activating STAT3 signaling and demonstrates the preclinical efficacy of antibody-based OSMR targeting for ovarian cancer treatment.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Subunidad beta del Receptor de Oncostatina M/antagonistas & inhibidores , Neoplasias Ováricas/prevención & control , Factor de Transcripción STAT3/antagonistas & inhibidores , Microambiente Tumoral , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Fibroblastos Asociados al Cáncer/inmunología , Proliferación Celular , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/metabolismo , Femenino , Humanos , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Oncostatina M/genética , Oncostatina M/metabolismo , Subunidad beta del Receptor de Oncostatina M/inmunología , Subunidad beta del Receptor de Oncostatina M/metabolismo , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Pronóstico , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cancer Cell ; 39(6): 779-792.e11, 2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-34087162

RESUMEN

The mesenchymal subtype of glioblastoma is thought to be determined by both cancer cell-intrinsic alterations and extrinsic cellular interactions, but remains poorly understood. Here, we dissect glioblastoma-to-microenvironment interactions by single-cell RNA sequencing analysis of human tumors and model systems, combined with functional experiments. We demonstrate that macrophages induce a transition of glioblastoma cells into mesenchymal-like (MES-like) states. This effect is mediated, both in vitro and in vivo, by macrophage-derived oncostatin M (OSM) that interacts with its receptors (OSMR or LIFR) in complex with GP130 on glioblastoma cells and activates STAT3. We show that MES-like glioblastoma states are also associated with increased expression of a mesenchymal program in macrophages and with increased cytotoxicity of T cells, highlighting extensive alterations of the immune microenvironment with potential therapeutic implications.


Asunto(s)
Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/patología , Glioblastoma/inmunología , Glioblastoma/patología , Linfocitos T/inmunología , Macrófagos Asociados a Tumores/inmunología , Animales , Neoplasias Encefálicas/genética , Células Cultivadas , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/metabolismo , Citotoxicidad Inmunológica , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Humanos , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/genética , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Oncostatina M/metabolismo , Subunidad beta del Receptor de Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Microambiente Tumoral , Macrófagos Asociados a Tumores/patología
8.
Breast Cancer Res ; 23(1): 56, 2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-34011405

RESUMEN

BACKGROUND: Invasive ductal carcinoma (IDC) is a serious problem for patients as it metastasizes, decreasing 5-year patient survival from > 95 to ~ 27%. The breast tumor microenvironment (TME) is often saturated with proinflammatory cytokines, such as oncostatin M (OSM), which promote epithelial-to-mesenchymal transitions (EMT) in IDC and increased metastasis. The extracellular matrix (ECM) also plays an important role in promoting invasive and metastatic potential of IDC. Specifically, the reorganization and alignment of collagen fibers in stromal ECM leads to directed tumor cell motility, which promotes metastasis. Lysyl oxidase like-2 (LOXL2) catalyzes ECM remodeling by crosslinking of collagen I in the ECM. We propose a novel mechanism whereby OSM induces LOXL2 expression, mediating stromal ECM remodeling of the breast TME. METHODS: Bioinformatics was utilized to determine survival and gene correlation in patients. IDC cell lines were treated with OSM (also IL-6, LIF, and IL-1ß) and analyzed for LOXL2 expression by qRT-PCR and immunolabelling techniques. Collagen I contraction assays, 3D invasion assays, and confocal microscopy were performed with and without LOXL2 inhibition to determine the impact of OSM-induced LOXL2 on the ECM. RESULTS: Our studies demonstrate that IDC patients with high LOXL2 and OSM co-expression had worse rates of metastasis-free survival than those with high levels of either, individually, and LOXL2 expression is positively correlated to OSM/OSM receptor (OSMR) expression in IDC patients. Furthermore, human IDC cells treated with OSM resulted in a significant increase in LOXL2 mRNA, which led to upregulated protein expression of secreted, glycosylated, and enzymatically active LOXL2. The expression of LOXL2 in IDC cells did not affect OSM-promoted EMT, and LOXL2 was localized to the cytoplasm and/or secreted. OSM-induced LOXL2 promoted an increase in ECM collagen I fiber crosslinking, which led to significant fiber alignment between cells and increased IDC cell invasion. CONCLUSIONS: Aligned collagen fibers in the ECM provide pathways for tumor cells to migrate more easily through the stroma to nearby vasculature and tissue. These results provide a new paradigm through which proinflammatory cytokine OSM promotes tumor progression. Understanding the nuances in IDC metastasis will lead to better potential therapeutics to combat against the possibility.


Asunto(s)
Aminoácido Oxidorreductasas/metabolismo , Neoplasias de la Mama/metabolismo , Matriz Extracelular/metabolismo , Oncostatina M/metabolismo , Aminoácido Oxidorreductasas/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patología , Línea Celular Tumoral , Colágeno Tipo I/metabolismo , Transición Epitelial-Mesenquimal/genética , Femenino , Glicosilación , Humanos , Inflamación , Metástasis de la Neoplasia , Oncostatina M/genética , Oncostatina M/farmacología , Subunidad beta del Receptor de Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/metabolismo , Pronóstico , Transducción de Señal , Microambiente Tumoral , Regulación hacia Arriba/genética
9.
Braz J Microbiol ; 52(3): 1057-1066, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33851342

RESUMEN

Helicobacter pylori, a human pathogen that colonizes the stomach of 50% of the world's population, is associated with gastritis, gastric adenocarcinoma, and mucosa-associated lymphoid tissue (MALT) lymphoma. Diseases are characterized by severe inflammatory responses in the stomach that are induced by various chemokines and cytokines. Recently, oncostatin M (OSM), an IL-6 family cytokine, was detected in early gastric cancer biopsies. In this study, we showed that Helicobacter pylori induced secretion of OSM and overexpression of its type II receptor OSMRß (OSM/OSMRß) in a human gastric adenocarcinoma cell line (AGS) over 24 h of infection. Furthermore, we showed that the induction of OSM and OSMRß was carried out by heat-sensitive Helicobacter pylori outer membrane vesicle (OMV) protein. Collectively, our results established, for the first time, a direct relation between Helicobacter pylori OMVs and the OSM/OSMRß signaling axis.


Asunto(s)
Adenocarcinoma , Membrana Externa Bacteriana , Infecciones por Helicobacter , Oncostatina M , Neoplasias Gástricas , Adenocarcinoma/metabolismo , Mucosa Gástrica , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/genética , Humanos , Oncostatina M/metabolismo , Subunidad beta del Receptor de Oncostatina M/metabolismo , Transducción de Señal , Neoplasias Gástricas/metabolismo
11.
Sci Rep ; 10(1): 17150, 2020 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-33051515

RESUMEN

Oncostatin M (OSM), a member of the IL-6 family of cytokines, has important roles in renal diseases. The relationship between OSM and kidney stone disease, however, remains unclear. To investigate the roles of OSM in the development of kidney stone disease, we generated a mouse model of renal crystal formation using OSM receptor ß (OSMRß)-deficient mice (OSMRß-/- mice). There were fewer renal crystal deposits in OSMRß-/- mice than in wild-type (WT) mice. Crystal-binding molecules (osteopontin, annexin A1, and annexin A2), inflammatory cytokines (TNF-α and IL-1ß), and fibrosis markers (TGF-ß, collagen 1a2, and α-smooth muscle actin) were also decreased in the kidneys of OSMRß-/- mice compared with those in WT mice. Immunofluorescence staining showed that OSMRß was expressed in renal tubular epithelial cells (RTECs) and renal fibroblasts in the model of renal crystal formation. In the cultured RTECs and renal fibroblasts, OSM directly induced the expression of crystal-binding molecules and fibrosis markers. Expressions of inflammatory cytokines were increased by stimulation with OSM in cultured renal fibroblasts. OSM may promote the formation of renal crystal deposits by directly acting on RTECs and renal fibroblasts to produce crystal-binding molecules and inflammatory cytokines.


Asunto(s)
Riñón/metabolismo , Riñón/patología , Subunidad beta del Receptor de Oncostatina M/metabolismo , Oncostatina M/metabolismo , Animales , Biomarcadores/metabolismo , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis/metabolismo , Fibrosis/patología , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/metabolismo
12.
Acta Neuropathol Commun ; 8(1): 42, 2020 04 05.
Artículo en Inglés | MEDLINE | ID: mdl-32248843

RESUMEN

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.


Asunto(s)
Anexina A2/genética , Neoplasias Encefálicas/genética , Glioblastoma/genética , Subunidad beta del Receptor de Oncostatina M/genética , Factor de Transcripción STAT3/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Anexina A2/metabolismo , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proliferación Celular/genética , Niño , Perros , Transición Epitelial-Mesenquimal/genética , Femenino , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Glioblastoma/irrigación sanguínea , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica/genética , Trasplante de Neoplasias , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Subunidad beta del Receptor de Oncostatina M/metabolismo , Fenotipo , Receptores de Oncostatina M/genética , Receptores de Oncostatina M/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Tasa de Supervivencia , Hipoxia Tumoral/genética
13.
Cells ; 9(1)2019 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-31861914

RESUMEN

BACKGROUND: Hepatic myofibroblasts (MFs) can originate from hepatic stellate cells, portal fibroblasts, or bone marrow-derived mesenchymal stem cells and can migrate towards the site of injury by aligning with nascent and established fibrotic septa in response to several mediators. Oncostatin M (OSM) is known to orchestrate hypoxia-modulated hepatic processes involving the hypoxia-inducible factor 1 (HIF-1). METHODS: In vivo and in vitro experiments were performed to analyze the expression of OSM and OSM-receptor (OSMR) in three murine models of non-alcoholic-fatty liver disease (NAFLD) and -steatohepatitis (NASH) and in human NASH patients as well as the action of OSM on phenotypic responses of human MFs. RESULTS: Hepatic OSM and OSMR levels were overexpressed in three murine NASH models and in NASH patients. OSM stimulates migration in human MFs by involving early intracellular ROS generation and activation of Ras/Erk, JNK1/2, PI3K/Akt as well as STAT1/STAT3 pathways and HIF-1α. OSM-dependent migration relies on a biphasic mechanism requiring early intracellular generation of reactive oxygen species (ROS) and late HIF1-dependent expression and release of VEGF. CONCLUSION: OSM is overexpressed in experimental and human progressive NAFLD and can act as a profibrogenic factor by directly stimulating migration of hepatic MFs.


Asunto(s)
Miofibroblastos/citología , Enfermedad del Hígado Graso no Alcohólico/genética , Subunidad beta del Receptor de Oncostatina M/genética , Oncostatina M/genética , Regulación hacia Arriba , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Miofibroblastos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Oncostatina M/metabolismo , Subunidad beta del Receptor de Oncostatina M/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
PLoS One ; 14(8): e0221477, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31461490

RESUMEN

OBJECTIVE: Previous studies indicate a role for Oncostatin M (OSM) in atherosclerosis and other chronic inflammatory diseases for which inhibitory antibodies are in development. However, to date no intervention studies with OSM have been performed, and its relation to coronary heart disease (CHD) has not been studied. APPROACH AND RESULTS: Gene expression analysis on human normal arteries (n = 10) and late stage/advanced carotid atherosclerotic arteries (n = 127) and in situ hybridization on early human plaques (n = 9) showed that OSM, and its receptors, OSM receptor (OSMR) and Leukemia Inhibitory Factor Receptor (LIFR) are expressed in normal arteries and atherosclerotic plaques. Chronic OSM administration in APOE*3Leiden.CETP mice (n = 15/group) increased plasma E-selectin levels and monocyte adhesion to the activated endothelium independently of cholesterol but reduced the amount of inflammatory Ly-6CHigh monocytes and atherosclerotic lesion size and severity. Using aptamer-based proteomics profiling assays high circulating OSM levels were shown to correlate with post incident CHD survival probability in the AGES-Reykjavik study (n = 5457). CONCLUSIONS: Chronic OSM administration in APOE*3Leiden.CETP mice reduced atherosclerosis development. In line, higher serum OSM levels were correlated with improved post incident CHD survival probability in patients, suggesting a protective cardiovascular effect.


Asunto(s)
Apolipoproteínas E/metabolismo , Aterosclerosis/patología , Proteínas de Transferencia de Ésteres de Colesterol/metabolismo , Oncostatina M/metabolismo , Animales , Aterosclerosis/sangre , Aterosclerosis/genética , Biomarcadores/metabolismo , Enfermedad Coronaria/sangre , Enfermedad Coronaria/genética , Enfermedad Coronaria/mortalidad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Humanos , Inflamación/patología , Interleucina-6/metabolismo , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/genética , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Ratones Transgénicos , Monocitos/patología , Oncostatina M/sangre , Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/metabolismo , Fenotipo , Placa Aterosclerótica/genética , Placa Aterosclerótica/patología , Probabilidad , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Supervivencia , Molécula 1 de Adhesión Celular Vascular/metabolismo
15.
Gastric Cancer ; 22(5): 955-966, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30778797

RESUMEN

BACKGROUND: Oncostatin M receptor (OSMR) is a member of the interleukin 6 (IL-6) receptor family that transduces signaling events of Oncostatin M (OSM). OSM-OSMR signaling plays a key role in inflammation and cancer progression. However, the role of OSM-OSMR in gastric cancer (GC) is still unknown. METHODS: OSMR expression in GC was determined by real-time PCR (RT-PCR), immunohistochemistry (IHC) and Western blot. The effects of OSM-OSMR on GC cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) in vitro and metastasis in vivo were examined. The pathways underlying OSM-OSMR signaling were explored by Western blot. Regulatory mechanism between SP1 and OSMR was explored in vitro. RESULTS: OSMR was highly expressed in GC tissues and its expression level was closely associated with age, T stage, Lauren classification, lymph node metastasis, TNM stage and worse prognosis of patients with GC. Knockdown of OSMR expression in GC cells significantly inhibited cell proliferation, migration, invasion, and EMT in vitro, as well as tumorigenesis and peritoneal metastasis in vivo induced by OSM. These effects mediated by OSM-OSMR were dependent on the activation of STAT3/FAK/Src signaling. SP1 could bind to the promoter region of human OSMR gene from - 255 to - 246 bp, and transcriptionally regulated OSMR overexpression in GC cells. CONCLUSIONS: OSM-OSMR contributes to GC progression through activating STAT3/FAK/Src signaling, and OSMR is transcriptionally activated by SP1.


Asunto(s)
Adenocarcinoma/secundario , Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Subunidad beta del Receptor de Oncostatina M/metabolismo , Oncostatina M/farmacología , Factor de Transcripción Sp1/metabolismo , Neoplasias Gástricas/patología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis , Biomarcadores de Tumor/genética , Movimiento Celular , Proliferación Celular , Femenino , Estudios de Seguimiento , Humanos , Metástasis Linfática , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Subunidad beta del Receptor de Oncostatina M/genética , Pronóstico , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factor de Transcripción Sp1/genética , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Biol Chem ; 293(52): 20181-20199, 2018 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-30373773

RESUMEN

The pleiotropic interleukin-6 (IL-6)-type cytokine oncostatin M (OSM) signals in multiple cell types, affecting processes such as cell differentiation, hematopoiesis, and inflammation. In humans, OSM exerts its effects through activation of either of two different heterodimeric receptor complexes, formed by glycoprotein 130 (gp130) and either OSM receptor (OSMR) or leukemia inhibitory factor receptor (LIFR). In contrast, the mouse OSM orthologue acts mainly through dimers containing OSMR and gp130 and shows limited activity through mouse LIFR. Despite their structural similarity, neither human nor mouse OSM signal through the other species' OSMR. The molecular basis for such species-specific signaling, however, remains poorly understood. To identify key molecular features of OSM that determine receptor activation in humans and mice, we generated chimeric mouse-human cytokines. Replacing regions within binding site III of murine OSM with the human equivalents showed that the cytokine's AB loop was critical for receptor selection. Substitutions of individual amino acids within this region demonstrated that residues Asn-37, Thr-40, and Asp-42 of the murine cytokine were responsible for limited LIFR activation and absence of human OSMR/LIFR signaling. In human OSM, Lys-44 appeared to be the main residue preventing mouse OSMR activation. Our data reveal that individual amino acids within the AB loop of OSM determine species-specific activities. These mutations might reflect a key step in the evolutionary process of this cytokine, in which receptor promiscuity gives way to ligand-receptor specialization.


Asunto(s)
Oncostatina M/metabolismo , Transducción de Señal , Animales , Línea Celular , Humanos , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/genética , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Ratones , Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/metabolismo , Multimerización de Proteína/genética , Estructura Secundaria de Proteína , Especificidad de la Especie
17.
Cardiovasc Res ; 114(12): 1667-1679, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29850784

RESUMEN

Aims: Myocardial infarction (MI) causes a massive increase of macrophages in the heart, which serve various non-redundant functions for cardiac repair. The identities of signals controlling recruitment of functionally distinct cardiac macrophages to sites of injury are only partially known. Previous work identified Regenerating islet-derived protein 3 beta (Reg3ß) as a novel factor directing macrophages to sites of myocardial injury. Herein, we aim to characterize functionally distinct macrophage subsets and understand the impact of different members of the Reg protein family including Reg3ß, Reg3γ, and Reg4 on their accumulation in the infarcted heart. Methods and results: We have determined dynamic changes of three phenotypically distinct tissue macrophage subpopulations in the mouse heart after MI by flow cytometry. RNA sequencing and bioinformatics analysis identified inflammatory gene expression patterns in MHC-IIhi/Ly6Clo and MHC-IIlo/Ly6Clo cardiac tissue macrophages while Ly6Chi cardiac tissue macrophages are characterized by gene activities associated with healing and revascularization of damaged tissue. Loss- and gain-of-function experiments revealed specific roles of Reg proteins for recruitment of cardiac tissue macrophage subpopulations to the site of myocardial injury. We found that expression of Reg3ß, Reg3γ, and Reg4 is strongly increased after MI in mouse and human hearts with Reg3ß providing the lead, followed by Reg3γ and Reg4. Inactivation of the Reg3ß gene prevented the increase of all types of cardiac tissue macrophages shortly after MI whereas local delivery of Reg3ß, Reg3γ, and Reg4 selectively stimulated recruitment of MHC-IIhi/Ly6Clo and MHC-IIlo/Ly6Clo but repressed accumulation of Ly6Chi cardiac tissue macrophages. Conclusion: We conclude that distinct cardiac macrophage subpopulations are characterized by substantially different gene expression patterns reflecting their pathophysiological role after MI. We argue that sequential, local production of Reg proteins orchestrates accumulation of macrophage subsets, which seem to act in a parallel or partially overlapping rather than in a successive manner.


Asunto(s)
Quimiotaxis , Macrófagos/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Asociadas a Pancreatitis/metabolismo , Animales , Antígenos Ly/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Proteínas de Neoplasias/genética , Subunidad beta del Receptor de Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/metabolismo , Proteínas Asociadas a Pancreatitis/deficiencia , Proteínas Asociadas a Pancreatitis/genética , Fenotipo , Ratas Sprague-Dawley , Transducción de Señal
18.
Stem Cell Reports ; 10(6): 1920-1934, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-29779898

RESUMEN

Understanding the molecular pathways controlling hematopoietic stem cell specification and expansion is a necessary milestone to perform regenerative medicine. Here, we used the zebrafish model to study the role of the ckit signaling pathway in this process. We show the importance of kitb/kitlgb signaling in the specification and expansion of hematopoietic stem cells (HSCs), in the hemogenic endothelium and caudal hematopoietic tissue (CHT), respectively. Moreover, we identified the zebrafish ortholog of Oncostatin M (osm) in the zebrafish genome. We show that the osm/osmr pathway acts upstream of kitb during specification of the hemogenic endothelium, while both pathways act synergistically to expand HSCs in the CHT. Moreover, we found that osm, in addition to its role in promoting HSC proliferation, inhibits HSC commitment to the lymphoid fate. Altogether, our data identified two cytokines, kitlgb and osm, secreted by the vascular niche, that control HSCs during early embryonic development.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Oncostatina M/farmacología , Factor de Células Madre/farmacología , Pez Cebra , Animales , Biomarcadores , Expresión Génica , Células Madre Hematopoyéticas/efectos de los fármacos , Inmunohistoquímica , Linfocitos/citología , Linfocitos/efectos de los fármacos , Linfocitos/metabolismo , Modelos Biológicos , Subunidad beta del Receptor de Oncostatina M/metabolismo , Transducción de Señal/efectos de los fármacos
20.
J Biol Chem ; 293(18): 7017-7029, 2018 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-29511087

RESUMEN

Oncostatin M (OSM) and leukemia inhibitory factor (LIF) are closely related members of the interleukin-6 (IL-6) cytokine family. Both cytokines share a common origin and structure, and both interact through a specific region, termed binding site III, to activate a dimeric receptor complex formed by glycoprotein 130 (gp130) and LIF receptor (LIFR) in humans. However, only OSM activates the OSM receptor (OSMR)-gp130 complex. The molecular features that enable OSM to specifically activate the OSMR are currently unknown. To define specific sequence motifs within OSM that are critical for initiating signaling via OSMR, here we generated chimeric OSM-LIF cytokines and performed alanine-scanning experiments. Replacement of the OSM AB loop within OSM's binding site III with that of LIF abrogated OSMR activation, measured as STAT3 phosphorylation at Tyr-705, but did not compromise LIFR activation. Correspondingly, substitution of the AB loop and D-helix in LIF with their OSM counterparts was sufficient for OSMR activation. The alanine-scanning experiments revealed that residues Tyr-34, Gln-38, Gly-39, and Leu-45 (in the AB loop) and Pro-153 (in the D-helix) had specific roles in activating OSMR but not LIFR signaling, whereas Leu-40 and Cys-49 (in the AB loop), and Phe-160 and Lys-163 (in the D-helix) were required for activation of both receptors. Because most of the key amino acid residues identified here are conserved between LIF and OSM, we concluded that comparatively minor differences in a few amino acid residues within binding site III account for the differential biological effects of OSM and LIF.


Asunto(s)
Subunidad beta del Receptor de Oncostatina M/metabolismo , Oncostatina M/metabolismo , Aminoácidos/química , Aminoácidos/metabolismo , Sitios de Unión , Receptor gp130 de Citocinas/metabolismo , Citocinas/metabolismo , Humanos , Factor Inhibidor de Leucemia/metabolismo , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Mutagénesis Sitio-Dirigida , Oncostatina M/química , Oncostatina M/genética , Subunidad beta del Receptor de Oncostatina M/química , Subunidad beta del Receptor de Oncostatina M/genética , Fosforilación , Unión Proteica , Receptores OSM-LIF/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA