Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Immunity ; 54(3): 484-498.e8, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33581044

RESUMEN

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.


Asunto(s)
Astrocitos/inmunología , Encéfalo/patología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Encefalomielitis Autoinmune Experimental/inmunología , Lectinas Tipo C/metabolismo , Esclerosis Múltiple/inmunología , Células Mieloides/inmunología , Inflamación Neurogénica/inmunología , Receptores Mitogénicos/metabolismo , Animales , Comunicación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Galectinas/metabolismo , Regulación de la Expresión Génica , Lectinas Tipo C/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito/inmunología , Oncostatina M/genética , Oncostatina M/metabolismo , Subunidad beta del Receptor de Oncostatina M/metabolismo , Fragmentos de Péptidos/inmunología , Receptores Mitogénicos/genética , Transducción de Señal
2.
J Biol Chem ; 300(5): 107251, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38569939

RESUMEN

Ciliary neurotrophic factor (CNTF) activates cells via the non-signaling α-receptor CNTF receptor (CNTFR) and the two signaling ß-receptors glycoprotein 130 (gp130) and leukemia inhibitory factor receptor (LIFR). The CNTF derivate, Axokine, was protective against obesity and insulin resistance, but clinical development was halted by the emergence of CNTF antibodies. The chimeric cytokine IC7 used the framework of interleukin (IL-)6 with the LIFR-binding site from CNTF to activate cells via IL-6R:gp130:LIFR complexes. Similar to CNTF/Axokine, IC7 protected mice from obesity and insulin resistance. Here, we developed CNTF-independent chimeras that specifically target the IL-6R:gp130:LIFR complex. In GIL-6 and GIO-6, we transferred the LIFR binding site from LIF or OSM to IL-6, respectively. While GIO-6 signals via gp130:IL-6R:LIFR and gp130:IL-6R:OSMR complexes, GIL-6 selectively activates the IL-6R:gp130:LIFR receptor complex. By re-evaluation of IC7 and CNTF, we discovered the Oncostatin M receptor (OSMR) as an alternative non-canonical high-affinity receptor leading to IL-6R:OSMR:gp130 and CNTFR:OSMR:gp130 receptor complexes, respectively. The discovery of OSMR as an alternative high-affinity receptor for IC7 and CNTF designates GIL-6 as the first truly selective IL-6R:gp130:LIFR cytokine, whereas GIO-6 is a CNTF-free alternative for IC7.


Asunto(s)
Factor Neurotrófico Ciliar , Receptor gp130 de Citocinas , Interleucina-6 , Transducción de Señal , Animales , Humanos , Ratones , Factor Neurotrófico Ciliar/metabolismo , Factor Neurotrófico Ciliar/genética , Receptor gp130 de Citocinas/metabolismo , Receptor gp130 de Citocinas/genética , Interleucina-6/metabolismo , Interleucina-6/genética , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/genética , Modelos Moleculares , Ingeniería de Proteínas/métodos , Estructura Terciaria de Proteína , Receptores de Interleucina-6/metabolismo , Receptores de Interleucina-6/genética , Receptores OSM-LIF/metabolismo , Receptores OSM-LIF/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes de Fusión/genética , Ratones Endogámicos C57BL
3.
J Transl Med ; 21(1): 290, 2023 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-37120549

RESUMEN

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


Asunto(s)
Interleucina-6 , Receptores OSM-LIF , Receptores de Oncostatina M , Transducción de Señal , Animales , Ratones , Cardiomegalia , Macrófagos , Oncostatina M/genética , Receptores OSM-LIF/genética , Receptores de Oncostatina M/genética
4.
FASEB J ; 36(6): e22368, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35596683

RESUMEN

Chronic itch is a complex sensation of the skin frequently associated with skin diseases, such as atopic dermatitis (AD) and psoriasis. Although Serpin E1 is implicated in chronic itch, its receptor and signaling pathways involved in itch are not known. In this study, the clinical relevance of a putative Serpin E1 receptor PLAUR to chronic itch, and the neuro-cutaneous Serpin E1-PLAUR signaling are explored. We found that PLAUR is overexpressed in skin specimens of human lesional AD and lesional psoriasis, and sensory neurons innervating MC903-induced AD-like murine skin. Murine PLAUR+ sensory neurons responded to Serpin E1, resulting in enrichment of numerous itch- and inflammation-related genes and their protein release. PLAUR resides in TLR2+ neurons and Serpin E1 stimulus led to transcriptional upregulation of TLR2 and its co-signaling proteins. Agonists of TLR2 propagated itch-related gene transcription including BNP, OSM, and PAR2. OSM induced acute itch in mice and promoted G-CSF and IL-8 release from human keratinocytes. Serpin E1 inhibitor reduced MC903-induced itch, epidermal hyperplasia, immunocyte infiltration, and resulted in lower transcription/expression levels of Serpin E1 and OSM. Taken together, the PLAUR-TLR2-OSM signaling promotes skin-nerve communication, cutaneous inflammation, and itch, all feeding into an aggravation of AD and exaggerated itch circuits.


Asunto(s)
Prurito , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Animales , Dermatitis Atópica/genética , Inflamación , Ratones , Inhibidor 1 de Activador Plasminogénico/genética , Prurito/genética , Psoriasis/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Piel/metabolismo , Receptor Toll-Like 2/genética
5.
J Oral Pathol Med ; 52(2): 136-144, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36207791

RESUMEN

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


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello , Carcinoma de Células Escamosas/patología , Receptores de Oncostatina M/metabolismo , Transducción de Señal , Pronóstico , ARN Mensajero
6.
Cytokine ; 148: 155550, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34217594

RESUMEN

Cytokines control immune related events and are critically involved in a plethora of patho-physiological processes including autoimmunity and cancer development. In rare cases, single nucleotide polymorphisms (SNPs) or single nucleotide variations (SNVs) in cytokine receptors eventually cause detrimental ligand-independent, constitutive activation of signal transduction. Most SNPs have, however, no or only marginal influences on gene expression, protein stability, localization and function and thereby only slightly affecting pathogenesis probability. The SNP database (dbSNP) is an archive for a broad collection of polymorphisms in which SNPs are categorized and marked with a locus accession number "reference SNP" (rs). Here, we engineered an algorithm to directly align dbSNP information to DNA and protein sequence information to clearly illustrate a genetic SNP landscape exemplified for all tall cytokine receptors of the IL-6/IL-12 family, including IL-23R, IL-12Rß1, IL-12Rß2, gp130, LIFR, OSMR and WSX-1. This information was complemented by a comprehensive literature summary and structural insights of relevant disease-causing SNPs in cytokine/cytokine receptor interfaces. In summary, we present a general strategy with potential to apply to other cytokine receptor networks.


Asunto(s)
Interleucina-12/genética , Interleucina-6/genética , Polimorfismo de Nucleótido Simple/genética , Receptores de Citocinas/genética , Animales , Humanos , Publicaciones
7.
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
8.
Int J Mol Sci ; 22(8)2021 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-33917126

RESUMEN

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.


Asunto(s)
Susceptibilidad a Enfermedades , Enterocolitis/etiología , Enterocolitis/metabolismo , Enfermedad de Hirschsprung/complicaciones , Enfermedad de Hirschsprung/genética , Subunidad beta del Receptor de Oncostatina M/genética , Transducción de Señal , Alelos , Enterocolitis/patología , Expresión Génica , Frecuencia de los Genes , Variación Genética , Genotipo , Enfermedad de Hirschsprung/diagnóstico , Humanos , Modelos Moleculares , Subunidad beta del Receptor de Oncostatina M/química , Subunidad beta del Receptor de Oncostatina M/metabolismo , Conformación Proteica , Proteómica/métodos , Relación Estructura-Actividad , Secuenciación del Exoma , Secuenciación Completa del Genoma
9.
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
10.
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
11.
J Cell Physiol ; 234(8): 14221-14233, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30684280

RESUMEN

BACKGROUND: Acute kidney injury (AKI) shows several kinds of disorders, which acutely harm the kidney. However, the current medical methods have limited therapeutic effects. The present study aimed to find out the molecular mechanism of AKI pathogenesis, which may provide new insights for future therapy. METHODS: Bioinformatic analysis was conducted using the R language (AT&T BellLaboratories, University of Auckland, New Zealand) to acquire the differentially expressed long noncoding RNAs (lncRNAs) and messenger RNAs (mRNAs) in AKI. The expression levels of RNAs and related proteins in tissues and cells were detected by quantitative real-time PCR (qRT-PCR) and western blot. Dual-luciferase reporter gene assays were performed to verify the target relationship between microRNA (miRNA) and lncRNA as well as miRNA and mRNA. Flow cytometry and tunnel assay were used to detect the cell apoptotic rate in AKI. RESULTS: LINC00520, miR-27b-3p, and OSMR form an axis to regulate AKI. Knockdown of LINC00520 reduced acute renal injury both in vitro and in vivo. LINC00520 activated the PI3K/AKT pathway to aggravate renal ischemia/reperfusion injury, while upregulation of miR-27b-3p or downregulation of OSMR could accelerate the recovery of AKI. CONCLUSION: Overexpression of LINC00520 contributes to the aggravation of AKI by targeting miR-27b-3p/ OSMR.


Asunto(s)
Lesión Renal Aguda/genética , MicroARNs/genética , Subunidad beta del Receptor de Oncostatina M/genética , ARN Largo no Codificante/genética , Lesión Renal Aguda/patología , Animales , Línea Celular , Proliferación Celular/genética , Regulación de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Humanos , Proteína Oncogénica v-akt/genética , Fosfatidilinositol 3-Quinasas/genética , Ratas , Transducción de Señal/genética
12.
Mol Med ; 24(1): 28, 2018 06 05.
Artículo en Inglés | MEDLINE | ID: mdl-30134804

RESUMEN

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


Asunto(s)
Enfermedades Autoinmunes/genética , Quinasas Janus/metabolismo , Receptores de Oncostatina M/genética , Factor de Transcripción STAT3/metabolismo , Urticaria/genética , Animales , Enfermedades Autoinmunes/metabolismo , Niño , Preescolar , Enfermedad Crónica , Femenino , Silenciador del Gen , Humanos , Lactante , Quinasas Janus/genética , Masculino , Mastocitos/metabolismo , Ratones , Factor de Transcripción STAT3/genética , Transducción de Señal , Urticaria/metabolismo
13.
Cancer Invest ; 36(2): 141-151, 2018 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-29420094

RESUMEN

This study systematically reviewed previous literatures and analyzed the genotype-phenotype relationship between the multiple endocrine neoplasia type 2A (MEN 2A)-cutaneous lichen amyloidosis (CLA) and RET/OSMR/IL31RA mutations. RET/OSMR/IL31RA screening was performed on 8 RET-carriers from 3 independent Chinese MEN 2A families. Besides, 51 MEN 2A-CLA patients in 116 RET carriers from literatures were clustered and analyzed. Our results indicated that almost all MEN 2A-CLA patients exhibited CLA which was located in the scapular region and carried RET mutation at codon 634. Meanwhile, we firstly described MEN 2A-CLA here in Chinese Han patient with RET p.C634F mutation.


Asunto(s)
Amiloidosis/complicaciones , Pueblo Asiatico/genética , Marcadores Genéticos , Neoplasia Endocrina Múltiple Tipo 2a/complicaciones , Mutación , Proteínas Proto-Oncogénicas c-ret/genética , Enfermedades Cutáneas Metabólicas/complicaciones , Adulto , Amiloidosis/genética , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasia Endocrina Múltiple Tipo 2a/genética , Subunidad beta del Receptor de Oncostatina M/genética , Linaje , Fenotipo , Proto-Oncogenes Mas , Receptores de Interleucina/genética , Enfermedades Cutáneas Metabólicas/genética
14.
Int J Mol Sci ; 19(6)2018 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-29882921

RESUMEN

Aberrant methylation of multiple promoter CpG islands could be related to the biology of ovarian tumors and its determination could help to improve treatment strategies. DNA methylation profiling was performed using the Methylation Ligation-dependent Macroarray (MLM), an array-based analysis. Promoter regions of 41 genes were analyzed in 102 ovarian tumors and 17 normal ovarian samples. An average of 29% of hypermethylated promoter genes was observed in normal ovarian tissues. This percentage increased slightly in serous, endometrioid, and mucinous carcinomas (32%, 34%, and 45%, respectively), but decreased in germ cell tumors (20%). Ovarian tumors had methylation profiles that were more heterogeneous than other epithelial cancers. Unsupervised hierarchical clustering identified four groups that are very close to the histological subtypes of ovarian tumors. Aberrant methylation of three genes (BRCA1, MGMT, and MLH1), playing important roles in the different DNA repair mechanisms, were dependent on the tumor subtype and represent powerful biomarkers for precision therapy. Furthermore, a promising relationship between hypermethylation of MGMT, OSMR, ESR1, and FOXL2 and overall survival was observed. Our study of DNA methylation profiling indicates that the different histotypes of ovarian cancer should be treated as separate diseases both clinically and in research for the development of targeted therapies.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Metilación de ADN/genética , Neoplasias Ováricas/genética , Análisis por Conglomerados , Femenino , Humanos , Estimación de Kaplan-Meier , Regiones Promotoras Genéticas
15.
J Biol Chem ; 290(41): 24747-59, 2015 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-26306032

RESUMEN

Interleukin-31 (IL-31) is a T helper type 2 cell-derived cytokine tightly associated with inflammatory skin disorders. IL-31-induced signaling is mediated by a receptor complex composed of oncostatin M receptor ß and the cytokine-specific receptor subunit IL-31Rα, of which there are several isoforms. The latter can be classified as long or short isoforms with respect to their intracellular domain. At present, the signaling capabilities of the different isoforms remain inchoately understood, and potential mechanisms involved in negative regulation of IL-31Rα signaling have so far not been studied in detail. Here, we show that both the long and short isoforms of IL-31Rα are capable of inducing STAT signaling. However, the presence of a functional JAK-binding box within IL-31Rα is an essential prerequisite for functional IL-31-mediated STAT3 signaling. Moreover, both the long and short isoforms require oncostatin M receptor ß for their activity. We also show that IL-31 induces expression of four suppressor of cytokine signaling family members and provide evidence that SOCS3 acts as a potent feedback inhibitor of IL-31-induced signaling. Taken together, this study identifies crucial requirements for IL-31 signaling and shows its counter-regulation by SOCS3.


Asunto(s)
Retroalimentación Fisiológica , Interleucinas/metabolismo , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Secuencias de Aminoácidos , Membrana Celular/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Espacio Intracelular/metabolismo , Subunidad beta del Receptor de Oncostatina M/metabolismo , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Receptores de Interleucina/química , Receptores de Interleucina/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Tirosina/metabolismo
16.
Cell Mol Gastroenterol Hepatol ; 17(3): 453-479, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37993018

RESUMEN

BACKGROUND & AIMS: HNF4α, a master regulator of liver development and the mature hepatocyte phenotype, is down-regulated in chronic and inflammatory liver disease. We used contemporary transcriptomics and epigenomics to study the cause and effects of this down-regulation and characterized a multicellular etiology. METHODS: Progressive changes in the rat carbon tetrachloride model were studied by deep RNA sequencing and genome-wide chromatin immunoprecipitation sequencing analysis of transcription factor (TF) binding and chromatin modification. Studies compared decompensated cirrhosis with liver failure after 26 weeks of treatment with earlier compensated cirrhosis and with additional rat models of chronic fibrosis. Finally, to resolve cell-specific responses and intercellular signaling, we compared transcriptomes of liver, nonparenchymal, and inflammatory cells. RESULTS: HNF4α was significantly lower in 26-week cirrhosis, part of a general reduction of TFs that regulate metabolism. Nevertheless, increased binding of HNF4α contributed to strong activation of major phenotypic genes, whereas reduced binding to other genes had a moderate phenotypic effect. Decreased Hnf4a expression was the combined effect of STAT3 and nuclear factor kappa B (NFκB) activation, which similarly reduced expression of other metabolic TFs. STAT/NFκB also induced de novo expression of Osmr by hepatocytes to complement induced expression of Osm by nonparenchymal cells. CONCLUSIONS: Liver decompensation by inflammatory STAT3 and NFκB signaling was not a direct consequence of progressive cirrhosis. Despite significant reduction of Hnf4a expression, residual levels of this abundant TF still stimulated strong new gene expression. Reduction of HNF4α was part of a broad hepatocyte transcriptional response to inflammation.


Asunto(s)
Factor Nuclear 4 del Hepatocito , Fallo Hepático , Animales , Ratas , Factor Nuclear 4 del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Hepatocitos/metabolismo , Cirrosis Hepática/patología , Fallo Hepático/metabolismo
17.
Ann Transl Med ; 11(2): 69, 2023 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-36819551

RESUMEN

Background: Despite receiving standard treatment, the prognosis of glioblastoma (GBM) patients is still poor. Considering the heterogeneity of each patient, it is imperative to identify reliable risk model that can effectively predict the prognosis of each GBM patient to guide the personalized treatment. Methods: Transcriptomic gene expression profiles and corresponding clinical data of GBM patients were downloaded from The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) databases. Inflammatory response-related genes were extracted from Gene Set Enrichment Analysis (GSEA) website. Univariate Cox regression analysis was used for prognosis-related inflammatory genes (P<0.05). A polygenic prognostic risk model was constructed using least absolute shrinkage and selection operator (LASSO) Cox regression analysis. Validation was performed through CGGA cohort. Overall survival (OS) was compared by Kaplan-Meier analysis. A nomogram was plotted to accurately predict the prognosis for each patient. GSEA was used for the pathway enrichment analysis. The single sample GSEA (ssGSEA) algorithm was implemented to conduct the immune infiltration analysis. The potential role of oncostatin M receptor (OSMR) in GBM was investigated through the in vitro experiment. Results: A prognostic risk model consisting of 4 genes (PTPRN, OSMR, MYD88, and EFEMP2) was developed. GBM patients in the high-risk group had worse OS. The time-dependent ROC curves showed an area under the curve (AUC) of 0.782, 0.765, and 0.784 for 1-, 2-, and 3-year survival in TCGA cohort, while the AUC in the CGGA cohort was 0.589, 0.684, and 0.785 at 1, 2, and 3 years, respectively. The risk score, primary-recurrent-secondary (PRS) type, and isocitrate dehydrogenase (IDH) mutation could predict the prognosis of GBM patients well. The nomogram accurately predicted the 1-, 2-, and 3-year OS for each patient. Immune cell infiltration was associated with the risk score and the model could predict immunotherapy responsiveness. The expression of the prognostic gene was correlated with the sensitivity to antitumor drugs. Interference of OSMR inhibited proliferation and migration and promoted apoptosis of GBM cells. Conclusions: The prognostic model based on 4 inflammatory response-related genes had reliable predictive power to effectively predict clinical outcome in GBM patients and provided the guide for the personalized treatment.

18.
J Dermatol Sci ; 110(2): 53-60, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37100691

RESUMEN

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.


Asunto(s)
Amiloidosis Familiar , Enfermedades Cutáneas Genéticas , Humanos , Animales , Ratones , Enfermedades Cutáneas Genéticas/patología , Piel/patología , Amiloidosis Familiar/genética , Queratinocitos/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Oncostatina M/farmacología , Subunidad beta del Receptor de Oncostatina M/genética
19.
Biomedicines ; 11(12)2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38137445

RESUMEN

OBJECTIVES: The cytokine oncostatin M (OSM) elicits pathogenic effects involving disruption of the epithelial barrier function as a part of immunological response networks. It is unclear how these integrated cytokine signals influence inflammation and other physiological processes in the pathology of chronic rhinosinusitis (CRS). We investigated the expression and distribution of OSM and OSM receptor (OSMR) in CRS patients' sinonasal specimens, and we compared the results with a panel of inflammatory cytokine levels and clinical features. PATIENTS AND METHODS: We classified CRS patients as eosinophilic (ECRS, n = 36) or non-eosinophilic (non-ECRS, n = 35) based on the Japanese Epidemiological Survey of Refractory Eosinophilic Chronic Rhinosinusitis phenotypic criteria and compared their cases with those of 20 control subjects. We also examined OSM's stimulatory effects on cytokine receptor expression levels using the human bronchial epithelium cell line BEAS-2B. RESULTS: RT-PCR showed that the OSM mRNA levels were significantly increased in the CRS patients' ethmoid sinus mucosa. The OSM mRNA levels were positively correlated with those of TNF-α, IL-1ß, IL-13, and OSMR-ß. In BEAS-2B cells, OSM treatment induced significant increases in the OSMRß, IL-1R1, and IL-13Ra mRNA levels. CONCLUSIONS: OSM is involved in the pathogenesis of CRS in both type 1 and type 2 inflammation, suggesting the OSM signaling pathway as a potential therapeutic target for modulating epithelial stromal interactions.

20.
Pharmaceuticals (Basel) ; 15(6)2022 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-35745569

RESUMEN

Synovial sarcoma (SS) is a pediatric muscle cancer that primarily affects adolescents and young adults and has few treatment options. Complicating the treatment of synovial sarcoma is the low mutational burden of SS. Inflammatory pathways have been identified as being upregulated in some SS, leading to the discovery of upregulated oncostatin M receptor (OSMR). It was found that OSMR is upregulated in SS by RNAseq analysis and quantitative PCR, highlighting its potential in the treatment of SS. Also, OSMR is upregulated in mouse models for synovial sarcoma as demonstrated by western blot and immunohistochemistry, and the protein is present in both primary and metastatic sites of disease. Using a radioimmune therapy drug model, targeted therapy was synthesized for use in OSMR expressing SS and it was demonstrated that this drug is stable, while capable of efficient OSMR binding and isotope capture. Finally, this antibody conjugate exhibited ideal pharmacokinetics and targeted sites of disease in our mouse model and was taken up in both primary and metastatic diseased tissue. This suggests OSMR as an ideal target for therapy and this radioimmune therapy provides a novel treatment option for a disease with few therapy choices.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda