Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
1.
Comput Biol Med ; 155: 106709, 2023 03.
Article in English | MEDLINE | ID: mdl-36854228

ABSTRACT

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.


Subject(s)
Molecular Dynamics Simulation , Oncostatin M Receptor beta Subunit , Oncostatin M/metabolism , Oncostatin M/pharmacology , Oncostatin M Receptor beta Subunit/chemistry , Oncostatin M Receptor beta Subunit/metabolism , Protein Binding , Signal Transduction
2.
Stem Cell Res ; 63: 102842, 2022 08.
Article in English | MEDLINE | ID: mdl-35714449

ABSTRACT

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.


Subject(s)
Human Embryonic Stem Cells , CRISPR-Cas Systems/genetics , Cell Line , Homologous Recombination , Homozygote , Human Embryonic Stem Cells/metabolism , Humans , Kruppel-Like Transcription Factors/genetics , Mutation/genetics , Oncostatin M Receptor beta Subunit/genetics , Oncostatin M Receptor beta Subunit/metabolism
3.
Stem Cell Res Ther ; 13(1): 278, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35765036

ABSTRACT

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.


Subject(s)
Extracellular Signal-Regulated MAP Kinases , MAP Kinase Signaling System , Oncostatin M Receptor beta Subunit , Osteoblasts , Osteogenesis , Animals , Autophagy/physiology , Cell Differentiation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Oncostatin M Receptor beta Subunit/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism
4.
Biochem Biophys Res Commun ; 614: 114-119, 2022 07 23.
Article in English | MEDLINE | ID: mdl-35576682

ABSTRACT

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.


Subject(s)
Oncostatin M Receptor beta Subunit , Signal Transduction , Animals , Antibodies, Monoclonal , Cats , Dogs , Inflammation , Mice , Oncostatin M/metabolism , Oncostatin M Receptor beta Subunit/metabolism , Pruritus
5.
Int J Mol Sci ; 22(21)2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34769079

ABSTRACT

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.


Subject(s)
Cell Proliferation , Fibroblasts/cytology , Mesenchymal Stem Cells/cytology , Oncostatin M Receptor beta Subunit/metabolism , Oncostatin M/metabolism , Animals , Cell Line , Fibroblasts/metabolism , Mesenchymal Stem Cells/metabolism , Mice , NIH 3T3 Cells , Signal Transduction
6.
Cancer Res ; 81(20): 5336-5352, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34380633

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Oncostatin M Receptor beta Subunit/antagonists & inhibitors , Ovarian Neoplasms/prevention & control , STAT3 Transcription Factor/antagonists & inhibitors , Tumor Microenvironment , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cancer-Associated Fibroblasts/immunology , Cell Proliferation , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Female , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Oncostatin M/genetics , Oncostatin M/metabolism , Oncostatin M Receptor beta Subunit/immunology , Oncostatin M Receptor beta Subunit/metabolism , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Prognosis , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Cancer Cell ; 39(6): 779-792.e11, 2021 06 14.
Article in English | MEDLINE | ID: mdl-34087162

ABSTRACT

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.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/pathology , Glioblastoma/immunology , Glioblastoma/pathology , T-Lymphocytes/immunology , Tumor-Associated Macrophages/immunology , Animals , Brain Neoplasms/genetics , Cells, Cultured , Cytokine Receptor gp130/genetics , Cytokine Receptor gp130/metabolism , Cytotoxicity, Immunologic , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Humans , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Oncostatin M/metabolism , Oncostatin M Receptor beta Subunit/genetics , Oncostatin M Receptor beta Subunit/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Tumor Microenvironment , Tumor-Associated Macrophages/pathology
8.
Breast Cancer Res ; 23(1): 56, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34011405

ABSTRACT

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.


Subject(s)
Amino Acid Oxidoreductases/metabolism , Breast Neoplasms/metabolism , Extracellular Matrix/metabolism , Oncostatin M/metabolism , Amino Acid Oxidoreductases/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Collagen Type I/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Glycosylation , Humans , Inflammation , Neoplasm Metastasis , Oncostatin M/genetics , Oncostatin M/pharmacology , Oncostatin M Receptor beta Subunit/genetics , Oncostatin M Receptor beta Subunit/metabolism , Prognosis , Signal Transduction , Tumor Microenvironment , Up-Regulation/genetics
9.
Braz J Microbiol ; 52(3): 1057-1066, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33851342

ABSTRACT

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.


Subject(s)
Adenocarcinoma , Bacterial Outer Membrane , Helicobacter Infections , Oncostatin M , Stomach Neoplasms , Adenocarcinoma/metabolism , Gastric Mucosa , Helicobacter Infections/metabolism , Helicobacter pylori/genetics , Humans , Oncostatin M/metabolism , Oncostatin M Receptor beta Subunit/metabolism , Signal Transduction , Stomach Neoplasms/metabolism
10.
Int J Mol Sci ; 22(8)2021 Apr 07.
Article in English | MEDLINE | ID: mdl-33917126

ABSTRACT

Hirschsprung (HSCR) Associated Enterocolitis (HAEC) is a common life-threatening complication in HSCR. HAEC is suggested to be due to a loss of gut homeostasis caused by impairment of immune system, barrier defense, and microbiome, likely related to genetic causes. No gene has been claimed to contribute to HAEC occurrence, yet. Genetic investigation of HAEC by Whole-Exome Sequencing (WES) on 24 HSCR patients affected (HAEC) or not affected (HSCR-only) by enterocolitis and replication of results on a larger panel of patients allowed the identification of the HAEC susceptibility variant p.H187Q in the Oncostatin-M receptor (OSMR) gene (14.6% in HAEC and 5.1% in HSCR-only, p = 0.0024). Proteomic analysis on the lymphoblastoid cell lines from one HAEC patient homozygote for this variant and one HAEC patient not carrying the variant revealed two well distinct clusters of proteins significantly up or downregulated upon OSM stimulation. A marked enrichment in immune response pathways (q < 0.0001) was shown in the HAEC H187 cell line, while proteins upregulated in the HAEC Q187 lymphoblasts sustained pathways likely involved in pathogen infection and inflammation. In conclusion, OSMR p.H187Q is an HAEC susceptibility variant and perturbates the downstream signaling cascade necessary for the gut immune response and homeostasis maintenance.


Subject(s)
Disease Susceptibility , Enterocolitis/etiology , Enterocolitis/metabolism , Hirschsprung Disease/complications , Hirschsprung Disease/genetics , Oncostatin M Receptor beta Subunit/genetics , Signal Transduction , Alleles , Enterocolitis/pathology , Gene Expression , Gene Frequency , Genetic Variation , Genotype , Hirschsprung Disease/diagnosis , Humans , Models, Molecular , Oncostatin M Receptor beta Subunit/chemistry , Oncostatin M Receptor beta Subunit/metabolism , Protein Conformation , Proteomics/methods , Structure-Activity Relationship , Exome Sequencing , Whole Genome Sequencing
11.
Immunity ; 54(3): 484-498.e8, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33581044

ABSTRACT

Pathologic roles of innate immunity in neurologic disorders are well described, but their beneficial aspects are less understood. Dectin-1, a C-type lectin receptor (CLR), is largely known to induce inflammation. Here, we report that Dectin-1 limited experimental autoimmune encephalomyelitis (EAE), while its downstream signaling molecule, Card9, promoted the disease. Myeloid cells mediated the pro-resolution function of Dectin-1 in EAE with enhanced gene expression of the neuroprotective molecule, Oncostatin M (Osm), through a Card9-independent pathway, mediated by the transcription factor NFAT. Furthermore, we find that the Osm receptor (OsmR) functioned specifically in astrocytes to reduce EAE severity. Notably, Dectin-1 did not respond to heat-killed Mycobacteria, an adjuvant to induce EAE. Instead, endogenous Dectin-1 ligands, including galectin-9, in the central nervous system (CNS) were involved to limit EAE. Our study reveals a mechanism of beneficial myeloid cell-astrocyte crosstalk regulated by a Dectin-1 pathway and identifies potential therapeutic targets for autoimmune neuroinflammation.


Subject(s)
Astrocytes/immunology , Brain/pathology , CARD Signaling Adaptor Proteins/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Lectins, C-Type/metabolism , Multiple Sclerosis/immunology , Myeloid Cells/immunology , Neurogenic Inflammation/immunology , Receptors, Mitogen/metabolism , Animals , Cell Communication , Cells, Cultured , Disease Models, Animal , Galectins/metabolism , Gene Expression Regulation , Lectins, C-Type/genetics , Mice, Inbred C57BL , Mice, Knockout , Myelin-Oligodendrocyte Glycoprotein/immunology , Oncostatin M/genetics , Oncostatin M/metabolism , Oncostatin M Receptor beta Subunit/metabolism , Peptide Fragments/immunology , Receptors, Mitogen/genetics , Signal Transduction
12.
Exp Dermatol ; 30(6): 804-810, 2021 06.
Article in English | MEDLINE | ID: mdl-33428793

ABSTRACT

Prurigo nodularis (PN) is a chronic skin dermatosis with hyperkeratotic and intensely pruritic nodules. Managing PN-associated itch is difficult because its aetiology is still unknown. This study aimed to investigate the correlation between itch intensity in PN and the expression of a pruritogenic cytokine interleukin (IL)-31, its receptor complex components IL-31 receptor α (IL-31RA) and oncostatin M receptor ß (OSMRß), and oncostatin M (OSM), which is a ligand of OSMR ß, through immunofluorescence staining examination. Itch intensity in PN was closely correlated with the number of dermal IL-31(+) cells (Spearman's r = 0.551, p < 0.05), dermal IL-31RA(+) cells (r = 0.475, p < 0.05) and dermal OSM(+) cells (r = 0.505, p < 0.05). In addition, the number of dermal OSMRß (+) cells was increased in PN (t test, p < 0.05), despite not being correlated with itch intensity (Spearman's r = 0.375, p > 0.05). Major cellular sources of dermal IL-31 were T cells (27.0% of total IL-31-expressing cells) and macrophages (35.0%), while those of OSM were mainly T cells (49.8%) and mast cells (26.8%). IL-31RA-expressing dermal cells were mostly mast cells (49.3%) and macrophages (36.6%), and OSMRß was mainly expressed by macrophages (51.8%) in the dermis. These findings indicate that IL-31 (mainly from macrophages and T cells) and OSM (principally from T cells and mast cells) stimulate dermal cells expressing IL-31RA and OSMRß (e.g. macrophages), which may further promote itch and inflammation in PN. This complex dermal milieu of cell/cytokine/receptor network can be a therapeutic target for PN-associated itch.


Subject(s)
Interleukins/metabolism , Oncostatin M Receptor beta Subunit/metabolism , Oncostatin M/metabolism , Prurigo/metabolism , Pruritus/metabolism , Receptors, Interleukin/metabolism , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult
14.
Phytomedicine ; 80: 153372, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33113505

ABSTRACT

BACKGROUND: Feiyangchangweiyan capsule (FYC) is a traditional Chinese medicine formulation used in the clinical treatment of acute and chronic gastroenteritis and bacterial dysentery. However, the effect of FYC on ulcerative colitis (UC) and the mechanism thereof remains unknown. PURPOSE: To investigate the protective effect of FYC on UC mice induced by dextran sulfate sodium and illustrate the potential mechanism of this effect. METHODS: Here, we established a model of UC mice by dextran sulfate sodium and administered with FYC. The disease activity index (DAI), colon length, myeloperoxidase (MPO) content in serum, pathological structure and ultrastructural changes, and inflammatory cell infiltration of colon tissue were evaluated. Transcriptome and 16S rDNA sequencing were employed to illuminate the mechanism of FYC in the protection of UC mice. RESULTS: FYC significantly alleviates the pathological damage and the infiltration of inflammatory cells in colon tissue of dextran sulfate sodium induced UC mice, rescues shortened colon length, reduces DAI score, MPO content in serum, and pro-inflammatory factors including IL-1ß, IL-6, CCL11, MCP-1 and MIP-2, and increases anti-inflammatory factors such as IL-10. Transcriptomics revealed that Oncostatin M (OSM) and its receptor (OSMR) are the critical pathway for UC treatment by FYC. OSM and OSMR increased in UC mice compared to control mice, and decreased with FYC, which was verified via measurement of OSM and OSMR mRNA and protein levels. Furthermore, we observed that FYC modulates intestinal microbiome composition (e.g., the proportion of Barnesiella/Proteobacteria) by affecting the inflammatory factors. CONCLUSION: FYC exerts an effect on UC by inhibiting the OSM/OSMR pathway and regulating inflammatory factors to improve the intestinal flora.


Subject(s)
Colitis, Ulcerative/drug therapy , Drugs, Chinese Herbal/pharmacology , Gastrointestinal Microbiome/drug effects , Oncostatin M Receptor beta Subunit/metabolism , Oncostatin M/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Capsules , Chemokines/blood , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/microbiology , Colon/drug effects , Colon/metabolism , Colon/pathology , Colon/ultrastructure , Cytokines/blood , Dextran Sulfate/toxicity , Disease Models, Animal , Gastrointestinal Microbiome/genetics , Male , Mice, Inbred C57BL , Oncostatin M/genetics , Oncostatin M Receptor beta Subunit/genetics , Protective Agents/pharmacology
15.
Sci Rep ; 10(1): 17150, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33051515

ABSTRACT

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.


Subject(s)
Kidney/metabolism , Kidney/pathology , Oncostatin M Receptor beta Subunit/metabolism , Oncostatin M/metabolism , Animals , Biomarkers/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/metabolism , Fibrosis/pathology , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism
16.
Biochemistry ; 59(23): 2171-2181, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32459958

ABSTRACT

Interleukin-31 (IL-31) is a major protein involved in severe inflammatory skin disorders. Its signaling pathway is mediated through two type I cytokine receptors, IL-31RA (also known as the gp130-like receptor) and the oncostatin M receptor (OSMR). Understanding molecular details in these interactions would be helpful for developing antagonist anti-IL-31 monoclonal antibodies (mAbs) as potential therapies. Previous studies suggest that human IL-31 binds to IL-31RA and then recruits OSMR to form a ternary complex. In this model, OSMR cannot interact with IL-31 in the absence of IL-31RA. In this work, we show that feline IL-31 (fIL-31) binds independently with feline OSMR using surface plasmon resonance, an enzyme-linked immunosorbent assay, and yeast surface display. Moreover, competition experiments suggest that OSMR shares a partially overlapping epitope with IL-31RA. We then used deep mutational scanning to map the binding sites of both receptors on fIL-31. In agreement with previous studies of the human homologue, the binding site for IL31-RA contains fIL-31 positions E20 and K82, while the binding site for OSMR comprises the "PADNFERK" motif (P103-K110) and position G38. However, our results also revealed a new overlapping site, composed of positions R69, R72, P73, D76, D81, and E97, between both receptors that we called the "shared site". The conformational epitope of an anti-feline IL-31 mAb that inhibits both OSMR and IL-31RA also mapped to this shared site. Combined, our results show that fIL-31 binds IL-31RA and OSMR independently through a partially shared epitope. These results suggest reexamination of the putative canonical mechanisms for IL-31 signaling in higher animals.


Subject(s)
Epitopes/metabolism , Interleukins/metabolism , Oncostatin M Receptor beta Subunit/metabolism , Receptors, Interleukin/metabolism , Animals , Cats , Epitopes/chemistry , Humans , Interleukins/chemistry , Models, Molecular , Oncostatin M Receptor beta Subunit/chemistry , Receptors, Interleukin/chemistry
17.
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
18.
Front Immunol ; 11: 612013, 2020.
Article in English | MEDLINE | ID: mdl-33854494

ABSTRACT

Adipocytes are the largest cell type in terms of volume, but not number, in adipose tissue. Adipocytes are prominent contributors to systemic metabolic health. Obesity, defined by excess adipose tissue (AT), is recognized as a low-grade chronic inflammatory state. Cytokines are inflammatory mediators that are produced in adipose tissue (AT) and function in both AT homeostatic as well as pathological conditions. AT inflammation is associated with systemic metabolic dysfunction and obesity-associated infiltration and proliferation of immune cells occurs in a variety of fat depots in mice and humans. AT immune cells secrete a variety of chemokines and cytokines that act in a paracrine manner on adjacent adipocytes. TNFα, IL-6, and MCP-1, are well studied mediators of AT inflammation. Oncostatin M (OSM) is another proinflammatory cytokine that is elevated in AT in human obesity, and its specific receptor (OSMRß) is also induced in conditions of obesity and insulin resistance. OSM production and paracrine signaling in AT regulates adipogenesis and the functions of AT. This review summarizes the roles of the oncostatin M receptor (OSMRß) as a modulator of adipocyte development and function its contributions to immunological adaptations in AT in metabolic disease states.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Inflammation Mediators/metabolism , Obesity/metabolism , Oncostatin M Receptor beta Subunit/metabolism , Oncostatin M/metabolism , Panniculitis/metabolism , Adipocytes/immunology , Adipose Tissue/immunology , Adipose Tissue/physiopathology , Animals , Energy Metabolism , Homeostasis , Humans , Insulin Resistance , Obesity/immunology , Obesity/physiopathology , Panniculitis/immunology , Panniculitis/physiopathology , Signal Transduction
19.
Cells ; 9(1)2019 12 20.
Article in English | MEDLINE | ID: mdl-31861914

ABSTRACT

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.


Subject(s)
Myofibroblasts/cytology , Non-alcoholic Fatty Liver Disease/genetics , Oncostatin M Receptor beta Subunit/genetics , Oncostatin M/genetics , Up-Regulation , Animals , Cell Line , Cell Movement/drug effects , Disease Models, Animal , Humans , Hypoxia-Inducible Factor 1/metabolism , Mice , Myofibroblasts/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Oncostatin M/metabolism , Oncostatin M Receptor beta Subunit/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
20.
Neuroscience ; 422: 12-20, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31705893

ABSTRACT

Oncostatin M (OSM) is a cytokine of the interleukin (IL)-6 family members. It induces blood-brain barrier (BBB) dysfunction by activating Janus-activated kinase (JAK) and signal transducer and activator of transcription (STAT) 3 pathways in brain endothelial cells. Brain pericytes located around microvessels are one of the BBB constituents. Pericytes work as a boundary surface between the blood circulation and brain parenchyma, and their functions are altered under pathophysiological conditions, leading to BBB dysregulation. However, it remains unknown whether pericytes are associated with OSM-induced BBB dysfunction. We demonstrated that pericyte exposure to OSM (100 ng/mL) elevated phosphorylation of STAT3, a main OSM signaling pathway, and that pericytes expressed OSM receptors (OSMRs) including OSMRß and glycoprotein 130. These results suggest that pericytes are able to respond to OSM. To determine the effects of OSM-reactive pericytes on BBB functions, rat brain endothelial cell (RBEC) monolayers were cultured with OSM-treated pericytes. The presence of pericytes exposed to 100 ng/mL of OSM for 48 h aggravated both the elevated permeability to sodium fluorescein and the lowered transendothelial electrical resistance which were induced by OSM in RBECs. This OSM-reactive pericyte-induced aggravation of lowered RBEC barrier function was reversed by ruxolitinib, a JAK inhibitor. These findings suggest that activated JAK/STAT3 signaling in pericytes contributes to OSM-produced BBB breakdown. Thus, OSM-reactive pericytes may have to be considered a characteristic machinery in the formation and progression of BBB breakdown under pathological conditions associated with increased OSM levels.


Subject(s)
Blood-Brain Barrier/physiopathology , Janus Kinases/metabolism , Oncostatin M/pharmacology , Oncostatin M/physiology , STAT3 Transcription Factor/metabolism , Animals , Cytokine Receptor gp130/metabolism , Nitric Oxide Synthase Type III/metabolism , Nitriles , Oncostatin M/antagonists & inhibitors , Oncostatin M Receptor beta Subunit/metabolism , Pericytes/drug effects , Pericytes/metabolism , Phosphorylation/drug effects , Primary Cell Culture , Pyrazoles/pharmacology , Pyrimidines , Rats , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...