Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
J Dermatol Sci ; 110(2): 53-60, 2023 May.
Article in English | MEDLINE | ID: mdl-37100691

ABSTRACT

BACKGROUND: Primary localized cutaneous amyloidosis (PLCA) is a chronic skin disease characterized by aberrant keratinocyte differentiation, epidermal hyperproliferation, and amyloid deposits. Previously, we demonstrated OSMR loss-function mutants enhanced basal keratinocyte differentiation through the OSMR/STAT5/KLF7 signaling in PLCA patients. OBJECTIVE: To investigate the underlying mechanisms involved in basal keratinocyte proliferation in PLCA patients that remain unclear. METHODS: Patients with pathologically confirmed PLCA visiting the dermatologic outpatient clinic were involved in the study. Laser capture microdissection and mass spectrometry analysis, gene-edited mice, 3D human epidermis culture, flow cytometry, western blot, qRT-PCR and RNA sequencing were used to explore the underlying molecular mechanisms. RESULTS: In this study, we found that AHNAK peptide fragments were enriched in the lesions of PLCA patients, as detected by laser capture microdissection and mass spectrometry analysis. The upregulated expression of AHNAK was further confirmed using immunohistochemical staining. qRT-PCR and flow cytometry revealed that pre-treatment with OSM can inhibit AHNAK expression in HaCaT cells, NHEKs, and 3D human skin models, but OSMR knockout or OSMR mutations abolished this down-regulation trend. Similar results were obtained in wild-type and OSMR knockout mice. More importantly, EdU incorporation and FACS assays demonstrated the knockdown of AHNAK could induce G1 phase cell cycle arrest and inhibit keratinocyte proliferation. Furthermore, RNA sequencing revealed that AHNAK knockdown regulated keratinocyte differentiation. CONCLUSION: Taken together, these data indicated that the elevated expression of AHNAK by OSMR mutations led to hyperproliferation and overdifferentiation of keratinocytes, and the discovered mechanism might provide insights into potential therapeutic targets for PLCA.


Subject(s)
Amyloidosis, Familial , Skin Diseases, Genetic , Humans , Animals , Mice , Skin Diseases, Genetic/pathology , Skin/pathology , Amyloidosis, Familial/genetics , Keratinocytes/metabolism , Kruppel-Like Transcription Factors/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Oncostatin M/pharmacology , Oncostatin M Receptor beta Subunit/genetics
2.
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
3.
Acta Neuropathol ; 144(2): 259-281, 2022 08.
Article in English | MEDLINE | ID: mdl-35666306

ABSTRACT

Oncostatin M (OSM) is an IL-6 family member which exerts neuroprotective and remyelination-promoting effects after damage to the central nervous system (CNS). However, the role of OSM in neuro-inflammation is poorly understood. Here, we investigated OSM's role in pathological events important for the neuro-inflammatory disorder multiple sclerosis (MS). We show that OSM receptor (OSMRß) expression is increased on circulating lymphocytes of MS patients, indicating their elevated responsiveness to OSM signalling. In addition, OSM production by activated myeloid cells and astrocytes is increased in MS brain lesions. In experimental autoimmune encephalomyelitis (EAE), a preclinical model of MS, OSMRß-deficient mice exhibit milder clinical symptoms, accompanied by diminished T helper 17 (Th17) cell infiltration into the CNS and reduced BBB leakage. In vitro, OSM reduces BBB integrity by downregulating the junctional molecules claudin-5 and VE-cadherin, while promoting secretion of the Th17-attracting chemokine CCL20 by inflamed BBB-endothelial cells and reactive astrocytes. Using flow cytometric fluorescence resonance energy transfer (FRET) quantification, we found that OSM-induced endothelial CCL20 promotes activation of lymphocyte function-associated antigen 1 (LFA-1) on Th17 cells. Moreover, CCL20 enhances Th17 cell adhesion to OSM-treated inflamed endothelial cells, which is at least in part ICAM-1 mediated. Together, these data identify an OSM-CCL20 axis, in which OSM contributes significantly to BBB impairment during neuro-inflammation by inducing permeability while recruiting Th17 cells via enhanced endothelial CCL20 secretion and integrin activation. Therefore, care should be taken when considering OSM as a therapeutic agent for treatment of neuro-inflammatory diseases such as MS.


Subject(s)
Blood-Brain Barrier , Encephalomyelitis, Autoimmune, Experimental , Oncostatin M , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Oncostatin M/metabolism , Oncostatin M/pharmacology , Oncostatin M Receptor beta Subunit/biosynthesis , Oncostatin M Receptor beta Subunit/genetics , Th17 Cells/metabolism , Th17 Cells/pathology
4.
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
5.
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
6.
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
7.
J Pathol Clin Res ; 8(3): 279-293, 2022 05.
Article in English | MEDLINE | ID: mdl-35289095

ABSTRACT

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.


Subject(s)
Carcinoma, Squamous Cell , Carcinoma, Transitional Cell , Urinary Bladder Neoplasms , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Humans , Oncostatin M Receptor beta Subunit , Receptors, Oncostatin M/metabolism , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urinary Bladder Neoplasms/genetics
8.
Int J Mol Sci ; 23(3)2022 Feb 05.
Article in English | MEDLINE | ID: mdl-35163735

ABSTRACT

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


Subject(s)
Heart Failure , Interleukin-6 , Myocytes, Cardiac , Oncostatin M , Receptors, Oncostatin M , Animals , Cytokine Receptor gp130/metabolism , Humans , Interleukin-6/metabolism , Mice , Myocytes, Cardiac/metabolism , Oncostatin M/metabolism , Oncostatin M Receptor beta Subunit , Receptors, Oncostatin M/genetics , Receptors, Oncostatin M/metabolism
10.
Cells ; 10(12)2021 11 27.
Article in English | MEDLINE | ID: mdl-34943841

ABSTRACT

Ependymal cells reside in the adult spinal cord and display stem cell properties in vitro. They proliferate after spinal cord injury and produce neurons in lower vertebrates but predominantly astrocytes in mammals. The mechanisms underlying this glial-biased differentiation remain ill-defined. We addressed this issue by generating a molecular resource through RNA profiling of ependymal cells before and after injury. We found that these cells activate STAT3 and ERK/MAPK signaling post injury and downregulate cilia-associated genes and FOXJ1, a central transcription factor in ciliogenesis. Conversely, they upregulate 510 genes, seven of them more than 20-fold, namely Crym, Ecm1, Ifi202b, Nupr1, Rbp1, Thbs2 and Osmr-the receptor for oncostatin, a microglia-specific cytokine which too is strongly upregulated after injury. We studied the regulation and role of Osmr using neurospheres derived from the adult spinal cord. We found that oncostatin induced strong Osmr and p-STAT3 expression in these cells which is associated with reduction of proliferation and promotion of astrocytic versus oligodendrocytic differentiation. Microglial cells are apposed to ependymal cells in vivo and co-culture experiments showed that these cells upregulate Osmr in neurosphere cultures. Collectively, these results support the notion that microglial cells and Osmr/Oncostatin pathway may regulate the astrocytic fate of ependymal cells in spinal cord injury.


Subject(s)
Cell Lineage , Ependyma/metabolism , Gene Expression Profiling , Gene Expression Regulation , Oncostatin M/metabolism , RNA/genetics , Spinal Cord Injuries/genetics , Stem Cells/pathology , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cilia/genetics , Down-Regulation/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , Oncostatin M Receptor beta Subunit , RNA/metabolism , Spheroids, Cellular/metabolism , Spinal Cord/pathology , Up-Regulation/genetics
11.
Sci Transl Med ; 13(619): eabe3037, 2021 11 10.
Article in English | MEDLINE | ID: mdl-34757808

ABSTRACT

Chronic itch is a major symptom of many inflammatory skin diseases. This type of pruritus is thought to be facilitated by cytokines that activate cutaneous nerve fibers; however, the molecular components and mechanisms involved are poorly understood. We found that the cytokine oncostatin M (OSM) is highly up-regulated in psoriasis, atopic dermatitis, and cutaneous T cell lymphoma, diseases associated with chronic itch. OSM receptor (OSMR) is expressed by itch-selective natriuretic polypeptide B (Nppb) neurons, and single-cell sequencing showed that OSM is mainly produced by dermal T cells and monocytes. Unlike canonical pruritogens, OSM does not activate sensory neurons. Instead, it sensitizes neurons by potentiating neural responses to pruritogens and by enhancing neural excitability. Knockout of OSMR in sensory neurons attenuated OSM-sensitized itch and inflammatory itch in mice, and pharmacological antagonism of the OSMR complex effectively alleviated pruritus in experimental inflammatory dermatitis in a rodent model. Together, our results uncover OSM as an itch neuromodulator and reveal OSM signal transduction as a potential target for antipruritic therapy.


Subject(s)
Pruritus , Sensory Receptor Cells , Animals , Gene Knockout Techniques , Mice , Mice, Knockout , Oncostatin M/pharmacology , Oncostatin M Receptor beta Subunit/genetics , Sensory Receptor Cells/drug effects , Signal Transduction , Skin
12.
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
13.
J Dermatol Sci ; 104(2): 95-103, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34674925

ABSTRACT

BACKGROUND: Oncostatin M (OSM), an interleukin-6 (IL-6) family proinflammatory cytokine, plays a critical role in inflammatory skin diseases, but its mechanism of action is not well understood. OBJECTIVE: To demonstrate the mechanism of OSM induced pyropotosis in normal human epidermal keratinocytes (NHEKs) and immortalized human keratinocytes (HaCaT cells). METHODS: NHEKs and HaCaT cells were treated with OSM. Knockout of OSM receptor (OSMR) with CRISPR/Cas9 system, knockdown of GSDME with small interfering RNA and primary keratinocytes from Osmr-/- and Gsdme-/- mice were used to study the effect of OSMR and GSDME. After treatment of OSM, NHEKs and HaCaT cells were irradiated with UVB. The mRNA was analyzed by quantitative real-time polymerase chain reaction (qRT-PCR) and RNA sequencing, protein level was detected by Western Blotting, Elisa and immunofluorescence. Cell death was examined by lactate dehydrogenase (LDH) releasing. RESULTS: Here we found that OSM induced pyropotosis in NHEKs and HaCaT cells, but knockout of OSMR abolished pyropotosis. RNA sequencing revealed an upregulation of several key genes involved in NLRP3 inflammasome activation following OSM treatment, among which NLRP3, GSDME, and IL-1ß were confirmed by qRT-PCR and Western Blotting. Knockdown of GSDME alleviated OSM-induced pyropotosis. Pretreatment of OSM boosted UVB-induced pyroptosis and inflammation in NHEKs and HaCaT cells, and this priming function was lost in keratinocytes of Osmr-/- and Gsdme-/- mice. Similar results were obtained in a 3-dimensional culture of human epidermis. CONCLUSION: OSM functions as a priming cytokine to enhance UVB-induced inflammation in keratinocytes, providing insight into the pathogenesis of inflammatory skin diseases.


Subject(s)
Keratinocytes/radiation effects , Oncostatin M/pharmacology , Pore Forming Cytotoxic Proteins/metabolism , Pyroptosis/drug effects , Ultraviolet Rays , Animals , Epidermis/metabolism , Gene Knockout Techniques , HaCaT Cells , Humans , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Oncostatin M Receptor beta Subunit/genetics , Pore Forming Cytotoxic Proteins/genetics , Pyroptosis/genetics , Pyroptosis/radiation effects , RNA, Small Interfering , Up-Regulation
14.
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
15.
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
16.
J Pediatr Gastroenterol Nutr ; 73(3): 352-357, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34117193

ABSTRACT

OBJECTIVES: This study aimed to determine whether mRNA expression of oncostatin-M (OSM) and its receptor (OSMR) in initial, pre-treatment intestinal biopsies is predictive of response to tumor necrosis factor antagonists (anti-TNF) in a pediatric inflammatory bowel disease (IBD) cohort. Secondary outcomes correlated OSM and OSMR expression with demographic variables; IBD type, extent, phenotype, and severity; laboratory values; and endoscopic findings. METHODS: A retrospective chart review was conducted on 98 pediatric patients. Patients' clinical courses were stratified as follows: failed anti-TNF (n = 14), quiescent on anti-TNF (n = 36), anti-TNF naïve (n = 19), and age-matched non-IBD controls (n = 29). The mRNA from each patient's pre-treatment ileal or colonic biopsy was isolated, and expression of OSM and OSMR was analyzed. RESULTS: There was no difference in OSM or OSMR expression among the three IBD groups; however, expression was significantly higher in patients with IBD than non-IBD controls (P < 0.001). OSM and OSMR were more highly expressed in patients with ulcerative colitis (UC) with a Mayo score of 3 (P = 0.0092 and P = 0.0313, respectively). High OSM expression correlated with severe disease activity indices at diagnosis (P = 0.002), anemia at diagnosis (P = 0.0236), and need for immunomodulators (P = 0.0193) and steroids (P = 0.0273) during patients' clinical courses. CONCLUSIONS: OSM and OSMR expression were not predictive of response to anti-TNF in our pediatric cohort. OSM expression did correlate with IBD compared with healthy controls as well as with several clinical indicators of severe IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Oncostatin M Receptor beta Subunit/genetics , Oncostatin M/genetics , Child , Humans , Inflammatory Bowel Diseases/drug therapy , Retrospective Studies , Tumor Necrosis Factor Inhibitors
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...