Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
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
2.
Nature ; 569(7754): 131-135, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30996350

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis largely owing to inefficient diagnosis and tenacious drug resistance. Activation of pancreatic stellate cells (PSCs) and consequent development of dense stroma are prominent features accounting for this aggressive biology1,2. The reciprocal interplay between PSCs and pancreatic cancer cells (PCCs) not only enhances tumour progression and metastasis but also sustains their own activation, facilitating a vicious cycle to exacerbate tumorigenesis and drug resistance3-7. Furthermore, PSC activation occurs very early during PDAC tumorigenesis8-10, and activated PSCs comprise a substantial fraction of the tumour mass, providing a rich source of readily detectable factors. Therefore, we hypothesized that the communication between PSCs and PCCs could be an exploitable target to develop effective strategies for PDAC therapy and diagnosis. Here, starting with a systematic proteomic investigation of secreted disease mediators and underlying molecular mechanisms, we reveal that leukaemia inhibitory factor (LIF) is a key paracrine factor from activated PSCs acting on cancer cells. Both pharmacologic LIF blockade and genetic Lifr deletion markedly slow tumour progression and augment the efficacy of chemotherapy to prolong survival of PDAC mouse models, mainly by modulating cancer cell differentiation and epithelial-mesenchymal transition status. Moreover, in both mouse models and human PDAC, aberrant production of LIF in the pancreas is restricted to pathological conditions and correlates with PDAC pathogenesis, and changes in the levels of circulating LIF correlate well with tumour response to therapy. Collectively, these findings reveal a function of LIF in PDAC tumorigenesis, and suggest its translational potential as an attractive therapeutic target and circulating marker. Our studies underscore how a better understanding of cell-cell communication within the tumour microenvironment can suggest novel strategies for cancer therapy.


Asunto(s)
Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Factor Inhibidor de Leucemia/metabolismo , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Comunicación Paracrina , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Carcinogénesis/genética , Carcinoma Ductal Pancreático/diagnóstico , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Línea Celular Tumoral , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Femenino , Humanos , Factor Inhibidor de Leucemia/antagonistas & inhibidores , Factor Inhibidor de Leucemia/sangre , Masculino , Espectrometría de Masas , Ratones , Neoplasias Pancreáticas/diagnóstico , Comunicación Paracrina/efectos de los fármacos , Receptores OSM-LIF/deficiencia , Receptores OSM-LIF/genética , Receptores OSM-LIF/metabolismo , Microambiente Tumoral
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.
Hum Mol Genet ; 26(9): 1716-1731, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334964

RESUMEN

Congenital anomalies of the kidneys and urinary tract (CAKUT) are the most common cause of chronic kidney disease in children. As CAKUT is a genetically heterogeneous disorder and most cases are genetically unexplained, we aimed to identify new CAKUT causing genes. Using whole-exome sequencing and trio-based de novo analysis, we identified a novel heterozygous de novo frameshift variant in the leukemia inhibitory factor receptor (LIFR) gene causing instability of the mRNA in a patient presenting with bilateral CAKUT and requiring kidney transplantation at one year of age. LIFR encodes a transmembrane receptor utilized by IL-6 family cytokines, mainly by the leukemia inhibitory factor (LIF). Mutational analysis of 121 further patients with severe CAKUT yielded two rare heterozygous LIFR missense variants predicted to be pathogenic in three unrelated patients. LIFR mutants showed decreased half-life and cell membrane localization resulting in reduced LIF-stimulated STAT3 phosphorylation. LIFR showed high expression in human fetal kidney and the human ureter, and was also expressed in the developing murine urogenital system. Lifr knockout mice displayed urinary tract malformations including hydronephrosis, hydroureter, ureter ectopia, and, consistently, reduced ureteral lumen and muscular hypertrophy, similar to the phenotypes observed in patients carrying LIFR variants. Additionally, a form of cryptorchidism was detected in all Lifr-/- mice and the patient carrying the LIFR frameshift mutation. Altogether, we demonstrate heterozygous novel or rare LIFR mutations in 3.3% of CAKUT patients, and provide evidence that Lifr deficiency and deactivating LIFR mutations cause highly similar anomalies of the urogenital tract in mice and humans.


Asunto(s)
Receptores OSM-LIF/genética , Receptores OSM-LIF/metabolismo , Anomalías Urogenitales/genética , Adolescente , Adulto , Animales , Niño , Preescolar , Análisis Mutacional de ADN , Exoma , Femenino , Heterocigoto , Humanos , Lactante , Riñón/anomalías , Riñón/patología , Factor Inhibidor de Leucemia/genética , Factor Inhibidor de Leucemia/metabolismo , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/genética , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Masculino , Ratones , Ratones Noqueados , Mutación , Análisis de Secuencia de ADN , Uréter/anomalías , Uréter/patología , Sistema Urinario/patología
5.
Cancer Sci ; 109(6): 1764-1774, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29603493

RESUMEN

Breast cancer (BC) is an aggressive malignant disease in women worldwide with a high tendency to metastasize. However, important biomarkers for BC metastasis remain largely undefined. In the present study, we identified that long non-coding RNA-CTD-2108O9.1 is downregulated in BC tissues and cells and acts as a metastatic inhibitor of BC. Mechanistic investigation determined that lncRNA-CTD-2108O9.1 represses metastasis by targeting leukemia inhibitory factor receptor (LIFR), which is designated as a metastasis suppressor in BC. Our study characterizes a significant tumor suppressor active in BC metastasis repression through the known metastasis inhibitor LIFR.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , ARN Largo no Codificante/genética , Receptores OSM-LIF/genética , Animales , Neoplasias de la Mama/patología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/genética , Femenino , Humanos , Metástasis Linfática , Células MCF-7 , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Interferencia de ARN , Trasplante Heterólogo
6.
Biochem Biophys Res Commun ; 505(1): 274-281, 2018 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-30245131

RESUMEN

Using Tandem Mass Tags (TMT) labeling and LC-MS/MS analysis of peptides from two cell lines (CNE2 and its radioresistant subclone CNE2-IR), we identified 754 proteins differentially expressed in CNE2-IR compared to CNE2. MAP2K6 was identified as a candidate radioresistance-related protein kinase. In vitro functional analysis revealed that over-expression of MAP2K6 significantly enhanced cell survival and colony formation following irradiation in NPC cells. Further, knockdown of MAP2K6 in radioresistant NPC cells led to decreased colony formation and increased apoptotic cells following irradiation. However, the effect of MAP2K6 in regulating the radioresistance in NPC cells did not seem to depend on p38/MAPK activity. Importantly, MAP2K6 might be required for leukemia inhibitory factor receptor (LIFR)-regulated radioresistance, as the expression levels of MAP2K6 affected LIFR/p70S6K signaling activation in NPC cells. Further, MAP2K6 kinase activity is required to activate LIFR/p70S6K signaling and to confer pro-survival effect on NPC cells. In conclusion, MAP2K6 might be an important regulator of LIFR-induced radioresistance in NPC.


Asunto(s)
MAP Quinasa Quinasa 6/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Receptores OSM-LIF/metabolismo , Apoptosis/genética , Apoptosis/efectos de la radiación , Línea Celular Tumoral , Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Humanos , MAP Quinasa Quinasa 6/genética , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , Proteoma/genética , Interferencia de ARN , Tolerancia a Radiación/genética , Tolerancia a Radiación/efectos de la radiación , Radiación Ionizante , Receptores OSM-LIF/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/genética , Transducción de Señal/efectos de la radiación
7.
Biol Reprod ; 96(2): 313-326, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28203817

RESUMEN

The precise timing of progesterone signaling through its cognate receptor, the progesterone receptor (PGR), is critical for the establishment and maintenance of pregnancy. Loss of PGR expression in the murine uterine epithelium during the preimplantation period is a marker for uterine receptivity and embryo attachment. We hypothesized that the decrease in progesterone receptor A (PGRA) expression is necessary for successful embryo implantation. To test this hypothesis, a mouse model constitutively expressing PGRA (mPgrALsL/+) was generated. Expression of PGRA in all uterine compartments (Pgrcre) or uterine epithelium (Wnt7acre) resulted in infertility with defects in embryo attachment and stromal decidualization. Expression of critical PGRA target genes, indian hedgehog, and amphiregulin (Areg), was maintained through the window of receptivity while the estrogen receptor target gene, the leukemia inhibitory factor (Lif), a key regulator of embryo receptivity, was decreased. Transcriptomic and cistromic analyses of the mouse uterus at day 4.5 of pregnancy identified an altered group of genes regulating molecular transport in the control of fluid and ion levels within the uterine interstitial space. Additionally, LIF and its cognate receptor, the leukemia inhibitory factor receptor (LIFR), exhibited PGR-binding events in regions upstream of the transcriptional start sites, suggesting PGRA is inhibiting transcription at these loci. Therefore, downregulation of the PGRA isoform at the window of receptivity is necessary for the attenuation of hedgehog signaling, transcriptional activation of LIF signaling, and modulation of solutes and fluid, producing a receptive environment for the attaching embryo.


Asunto(s)
Implantación del Embrión , Endometrio , Progesterona/metabolismo , Receptores de Progesterona/metabolismo , Alelos , Animales , Clonación Molecular , Regulación hacia Abajo , Femenino , Regulación de la Expresión Génica/fisiología , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Masculino , Ratones Transgénicos , Receptores OSM-LIF/genética , Receptores OSM-LIF/metabolismo , Receptores de Progesterona/genética , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
8.
Biochim Biophys Acta ; 1850(4): 708-21, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25486623

RESUMEN

BACKGROUND: To study the role of miR-143 during embryo implantation in rat. METHODS: MiR-143 expression in rat early pregnancy was detected by Northern blot. The relation between miR-143 and Lifr predicted and confirmed by bioinformatics method, dual-luciferase activity assay, Western blot and immunohistochemistry. The role of miR-143 was detected by MTS, Edu and ranswell chamber assays. RESULTS: The expression level of miR-143 on gestation day 5-8 (g.d. 5-8) was higher than on g.d. 3-4 in uteri of pregnant rat. MiR-143 was mainly localized in the superficial stroma/primary decidual zone, luminal and glandular epithelia. The expression of miR-143 was not significantly influenced by pseudopregnancy, but the activation of delayed implantation and experimentally induced decidualization significantly promoted miR-143 expression. Over-expression of miR-143 in human endometrial stromal cells (ESCs) inhibited cell proliferation, migration and invasion. Knockdown of miR-143 promoted cell proliferation and invasion. The results of recombinant luciferase reporters showed that miR-143 could bind to the 3¢-untranslated region (UTR) of leukemia inhibitory factor receptor (Lifr) to inhibit Lifr translation. CONCLUSIONS: Uterine miR-143 may be involved in the successful pregnancy, especially during the process of blastocyst implantation through regulating Lifr. GENERAL SIGNIFICANCE: This study may have the potential to provide new insights into the understanding of miR-143 function during embryo implantation.


Asunto(s)
Implantación del Embrión , MicroARNs/fisiología , Animales , Movimiento Celular , Proliferación Celular , Estradiol/farmacología , Femenino , Humanos , MicroARNs/análisis , Embarazo , Ratas , Ratas Sprague-Dawley , Receptores OSM-LIF/genética , Útero/metabolismo
9.
J Cell Biochem ; 117(1): 49-58, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26060100

RESUMEN

Activation of cytokine signaling via the leukemia inhibitory factor receptor (LIFR) plays an integral role in hematopoiesis, osteogenesis, and placental development, along with mediating neurotrophic mechanisms. However, the regulatory control of the LIFR gene has remained largely unexplored. Here, we characterize the LIFR gene as a novel target of the RUNX1 transcription factor. The RUNX1 transcription factor is an essential regulator of hematopoiesis and is a frequent target of point mutations and chromosomal alterations in leukemia. RUNX1 regulates hematopoiesis through its control of genes important for hematopoietic cell growth, proliferation, and differentiation, including a number of cytokines and cytokine receptors. LIFR is regulated by two alternate promoters: a placental-specific and a ubiquitously active general promoter. We show that both of these promoters are regulated by RUNX1. However, in myeloid cells LIFR expression is driven solely by the general LIFR promoter with our data indicating that the placental promoter is epigenetically silenced in these cells. While RUNX1 activates the LIFR general promoter, the oncogenic RUNX1-ETO fusion protein generated by the t(8;21) translocation commonly associated with acute myeloid leukemia represses promoter activity. The data presented here establish LIFR as a transcriptional target of RUNX1 and suggest that disruption of RUNX1 activity in myeloid cells may result in altered LIFR signaling in these cells.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Receptores OSM-LIF/metabolismo , Western Blotting , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular Tumoral , Proliferación Celular/genética , Proliferación Celular/fisiología , Inmunoprecipitación de Cromatina , Aberraciones Cromosómicas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Humanos , Células Mieloides/metabolismo , Mutación Puntual/genética , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , Unión Proteica/fisiología , Receptores de Citocinas/genética , Receptores de Citocinas/metabolismo , Receptores OSM-LIF/genética
10.
Mol Cell ; 31(5): 737-48, 2008 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-18775332

RESUMEN

gp130 is a shared receptor for at least nine cytokines and can signal either as a homodimer or as a heterodimer with Leukemia Inhibitory Factor Receptor (LIF-R). Here, we biophysically and structurally characterize the full-length, transmembrane form of a quaternary cytokine receptor complex consisting of gp130, LIF-R, the cytokine Ciliary Neurotrophic Factor (CNTF), and its alpha receptor (CNTF-Ralpha). Thermodynamic analysis indicates that, unlike the cooperative assembly of the symmetric gp130/Interleukin-6/IL-6Ralpha hexameric complex, CNTF/CNTF-Ralpha heterodimerizes gp130 and LIF-R via noncooperative energetics to form an asymmetric 1:1:1:1 complex. Single particle electron microscopic analysis of the full-length gp130/LIF-R/CNTF-Ralpha/CNTF quaternary complex elucidates an asymmetric structural arrangement, in which the receptor extracellular and transmembrane segments join as a continuous, rigid unit, poised to sensitively transduce ligand engagement to the membrane-proximal intracellular signaling regions. These studies also enumerate the organizing principles for assembly of the "tall" class of gp130 family cytokine receptor complexes including LIF, IL-27, IL-12, and others.


Asunto(s)
Receptor gp130 de Citocinas/química , Complejos Multiproteicos/química , Estructura Cuaternaria de Proteína , Receptores OSM-LIF/química , Transducción de Señal/fisiología , Animales , Factor Neurotrófico Ciliar/química , Factor Neurotrófico Ciliar/genética , Factor Neurotrófico Ciliar/metabolismo , Cristalografía por Rayos X , Receptor gp130 de Citocinas/genética , Receptor gp130 de Citocinas/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Complejos Multiproteicos/ultraestructura , Receptor de Factor Neurotrófico Ciliar/química , Receptor de Factor Neurotrófico Ciliar/genética , Receptor de Factor Neurotrófico Ciliar/metabolismo , Receptores OSM-LIF/genética , Receptores OSM-LIF/metabolismo , Termodinámica
11.
J Biol Chem ; 289(1): 529-39, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24247238

RESUMEN

Rhabdomyosarcoma (RMS) is the most common soft tissue sarcoma found in children and young adults. It is characterized by the expression of a number of skeletal muscle-specific proteins, including MyoD and muscle α-actin. However, unlike normal myoblasts, RMS cells differentiate poorly both in vivo and in culture. As microRNAs are known to regulate tumorigenesis, intensive efforts have been made to identify microRNAs that are involved in RMS development. In this work, we found that miR-203 was frequently down-regulated by promoter hypermethylation in both RMS cell lines and RMS biopsies and could be reactivated by DNA-demethylating agents. Re-expression of miR-203 in RMS cells inhibited their migration and proliferation and promoted terminal myogenic differentiation. Mechanistically, miR-203 exerts its tumor-suppressive effect by directly targeting p63 and leukemia inhibitory factor receptor in RMS cells, which promotes myogenic differentiation by inhibiting the Notch and the JAK1/STAT1/STAT3 pathways, respectively. Our work reveals that miR-203 functions as a tumor suppressor in RMS development.


Asunto(s)
Metilación de ADN , ADN de Neoplasias/metabolismo , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , MicroARNs/biosíntesis , Regiones Promotoras Genéticas , Rabdomiosarcoma/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular Tumoral , ADN de Neoplasias/genética , Femenino , Humanos , Janus Quinasa 1/genética , Janus Quinasa 1/metabolismo , Masculino , Ratones , Ratones Desnudos , MicroARNs/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Receptores OSM-LIF/genética , Receptores OSM-LIF/metabolismo , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
12.
Reproduction ; 150(4): 395-403, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26336147

RESUMEN

Subordinate follicles (SFs) of bovine follicular waves undergo atresia due to declining FSH concentrations; however, the signalling mechanisms have not been fully deciphered. We used an FSH-induced co-dominance model to determine the effect of FSH on signalling pathways in granulosa cells of the second-largest follicles (SF in control cows and co-dominant follicle (co-DF2) in FSH-treated cows). The SF was smaller than DF in control cows while diameters of co-DF1 and co-DF2 in FSH-treated cows were similar. The presence of cleaved CASP3 protein confirmed that granulosa cells of SFs, but not of DFs and co-DFs, were apoptotic. To determine the effect of FSH on molecular characteristics of the second-largest follicles, we generated relative variables for the second largest follicle in each cow. For this, variables of SF or co-DF2 were divided by the variables of the largest follicle DF or co-DF1 in each cow. There was higher transcript abundance of MAPK1/3 and AKT1/2/3 but lower abundance of phosphorylated MAPK3/1 in SF than co-DF2 granulosa cells. Abundance of mRNA and phosphorylated protein of STAT3 was higher in granulosa cells of control SF than FSH-treated co-DF2. SF granulosa cells had higher levels of LIFR and IL6ST transcripts, the two receptors involved in STAT3 activation. Further, lower transcript abundance of interleukin 6 receptor (IL6R), another receptor involved in STAT3 activation, indicated that STAT3 activation in SF granulosa cells could be mainly due to leukemia inhibitory factor (LIF) signalling. These results indicate that atresia due to lack of FSH is associated with activated LIF-STAT3 signalling in SF granulosa cells, as FSH treatment reversed such activation.


Asunto(s)
Hormona Folículo Estimulante/farmacología , Factor Inhibidor de Leucemia/biosíntesis , Folículo Ovárico/efectos de los fármacos , Factor de Transcripción STAT3/biosíntesis , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/biosíntesis , Caspasa 3/genética , Bovinos , Femenino , Células de la Granulosa/metabolismo , Factor Inhibidor de Leucemia/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Oncogénica v-akt/efectos de los fármacos , Folículo Ovárico/ultraestructura , Receptores de Interleucina-6/biosíntesis , Receptores de Interleucina-6/genética , Receptores OSM-LIF/genética , Factor de Transcripción STAT3/genética , Transducción de Señal/efectos de los fármacos
13.
Zygote ; 21(2): 203-13, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22892066

RESUMEN

Summary The objective of this study was to evaluate the effect of supplementation of recombinant leukaemia inhibitory factor (LIF) in culture media on blastocyst development, total cell number and blastocyst hatching rates and the reverse transcription-polymerase chain reaction analysis of preimplantation buffalo embryos to determine whether they contain the LIF-encoding mRNA and its beta receptor (LIFRß) genes in different stages of preimplantation buffalo embryos. Cumulus-oocyte complexes retrieved from slaughterhouse buffalo ovaries were matured in vitro and fertilized using frozen buffalo semen. After 18 h of co-incubation with sperm, the presumptive zygotes were cultured in modified synthetic oviductal fluid without (control) or with rhLIF (100 ng/ml). There was no significant difference in the overall cleavage rate up to morula stage however the development of blastocysts, hatching rate and total cell numbers were significantly higher in the LIF-treated group than control. Transcripts for LIFRß were detected from immature, in vitro-matured oocytes and in the embryos up to blastocyst stage, while transcripts for the LIF were detected from 8-16-cell stage up to blastocyst, which indicated that embryo-derived LIF can act in an autocrine manner on differentiation process and blastocyst formation. This study indicated that the addition of LIF to the embryo culture medium improved development of blastocysts, functional (hatching) and morphological (number of cells) quality of the blastocysts produced in vitro. The stage-specific expression pattern of LIF and LIFRß mRNA transcripts in buffalo embryos indicated that LIF might play an important role in the preimplantation development and subsequent implantation of buffalo embryos.


Asunto(s)
Blastocisto/metabolismo , Implantación del Embrión , Embrión de Mamíferos/citología , Factor Inhibidor de Leucemia/genética , ARN Mensajero/genética , Receptores OSM-LIF/genética , Proteínas Recombinantes/genética , Secuencia de Aminoácidos , Animales , Blastocisto/citología , Búfalos , Embrión de Mamíferos/metabolismo , Femenino , Fertilización In Vitro , Técnicas para Inmunoenzimas , Factor Inhibidor de Leucemia/metabolismo , Datos de Secuencia Molecular , Filogenia , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores OSM-LIF/metabolismo , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido
14.
Cancer Biol Ther ; 24(1): 2271638, 2023 12 31.
Artículo en Inglés | MEDLINE | ID: mdl-37927213

RESUMEN

The poly(rC) binding protein 1 gene (PCBP1) encodes the heterogeneous nuclear ribonucleoprotein E1 (hnRNPE1), a nucleic acid-binding protein that plays a tumor-suppressive role in the mammary epithelium by regulating phenotypic plasticity and cell fate. Following the loss of PCBP1 function, the FAM3C gene (encoding the Interleukin-like EMT inducer, or "ILEI" protein) and the leukemia inhibitory factor receptor (LIFR) gene are upregulated. Interaction between FAM3C and LIFR in the extracellular space induces phosphorylation of signal transducer and activator of transcription 3 (pSTAT3). Overexpression and/or hyperactivity of STAT3 has been detected in 40% of breast cancer cases and is associated with a poor prognosis. Herein, we characterize feed-forward regulation of LIFR expression in response to FAM3C/LIFR/STAT3 signaling in mammary epithelial cells. We show that PCBP1 upregulates LIFR transcription through activity at the LIFR promoter, and that FAM3C participates in transcriptional regulation of LIFR. Additionally, our bioinformatic analysis reveals a signature of transcriptional regulation associated with FAM3C/LIFR interaction and identifies the TWIST1 transcription factor as a downstream effector that participates in the maintenance of LIFR expression. Finally, we characterize the effect of LIFR expression in cell-based experiments that demonstrate the promotion of invasion, migration, and self-renewal of breast cancer stem cells (BCSCs), consistent with previous studies linking LIFR expression to tumor initiation and metastasis in mammary epithelial cells.


Asunto(s)
Neoplasias de la Mama , Proteínas de Unión al ADN , Proteínas de Unión al ARN , Femenino , Humanos , Neoplasias de la Mama/patología , Línea Celular Tumoral , Autorrenovación de las Células/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/genética , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Proteínas de Neoplasias/genética , Receptores OSM-LIF/genética , Receptores OSM-LIF/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Invasividad Neoplásica
15.
J Neurosci ; 31(31): 11376-86, 2011 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-21813697

RESUMEN

In response to illness, animals subvert normal homeostasis and divert their energy utilization to fight infection. An important and unexplored feature of this response is the suppression of physical activity and foraging behavior in the setting of negative energy balance. Inflammatory signaling in the hypothalamus mediates the febrile and anorectic responses to disease, but the mechanism by which locomotor activity (LMA) is suppressed has not been described. Lateral hypothalamic orexin (Ox) neurons link energy status with LMA, and deficiencies in Ox signaling lead to hypoactivity and hypophagia. In the present work, we examine the effect of endotoxin-induced inflammation on Ox neuron biology and LMA in rats. Our results demonstrate a vital role for diminished Ox signaling in mediating inflammation-induced lethargy. This work defines a specific population of inflammation-sensitive, arousal-associated Ox neurons and identifies a proximal neural target for inflammatory signaling to Ox neurons, while eliminating several others.


Asunto(s)
Inflamación/complicaciones , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Letargia/tratamiento farmacológico , Letargia/etiología , Neuronas/metabolismo , Neuropéptidos/metabolismo , Análisis de Varianza , Animales , Adaptación a la Oscuridad/efectos de los fármacos , Modelos Animales de Enfermedad , Vías de Administración de Medicamentos , Ensayo de Inmunoadsorción Enzimática/métodos , Privación de Alimentos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Inflamación/inducido químicamente , Inyecciones Intraventriculares/métodos , Interleucina-1beta/farmacología , Interleucina-6/sangre , Péptidos y Proteínas de Señalización Intracelular/farmacología , Ventrículos Laterales/efectos de los fármacos , Ventrículos Laterales/fisiología , Letargia/patología , Masculino , Hormonas Estimuladoras de los Melanocitos/farmacología , Ratones , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/fisiología , Inhibidor NF-kappaB alfa , Trasplante de Neoplasias/métodos , Neuronas/efectos de los fármacos , Neuropéptidos/farmacología , Neurotensina/genética , Orexinas , Fotoperiodo , Polisacáridos/efectos adversos , Proteínas Proto-Oncogénicas c-fos/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344 , Ratas Sprague-Dawley , Receptores de Corticotropina/antagonistas & inhibidores , Receptores de Interleucina-11/genética , Receptores de Interleucina-11/metabolismo , Receptores OSM-LIF/genética , Receptores OSM-LIF/metabolismo
16.
Breast Cancer Res ; 14(6): 326, 2012 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-23216692

RESUMEN

The proto-oncogenes YAP and TAZ have previously gained much attention as downstream effectors of Hippo tumour suppressor signalling. While the regulation of YAP/TAZ by MST/LATS kinases is reasonably well understood, the nature of factors functioning upstream of MST/LATS is yet to be elucidated in detail. A recent paper by Ma and co-workers defines a novel role for leukemia inhibitory factor receptor (LIFR) signalling upstream of the Hippo-YAP pathway in breast cancer metastasis. Moreover, a whole genome in vivo RNA interference screen by Lippmann and colleagues identified LIFR as a breast tumour suppressor. Here, we discuss the implications of these studies for breast cancer research and treatment.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Neoplasias de la Mama/genética , Genes Supresores de Tumor , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Receptores OSM-LIF/genética , Aciltransferasas , Femenino , Vía de Señalización Hippo , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Transducción de Señal/genética , Factores de Transcripción/genética , Proteínas Señalizadoras YAP
17.
Clin Genet ; 82(1): 12-21, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22300393

RESUMEN

Stüve-Wiedemann syndrome (SWS) is a severe congenital skeletal dysplasia associated with life threatening dysautonomic manifestations. Newborns affected with this condition exhibit distinctive shortening and bowing of the long bones with reduced bone volume. The majority of affected newborns die early due to neuromuscular complications namely hyperthermia, apnea, and swallowing difficulties. In this review, we provide an overall picture on the clinical, including long-term management, molecular and cellular aspects of SWS and discuss briefly other related bent bone dysplasias.


Asunto(s)
Enfermedades del Desarrollo Óseo/genética , Exostosis Múltiple Hereditaria/genética , Factores de Transcripción NFI/genética , Osteocondrodisplasias/genética , Receptores OSM-LIF/genética , Enfermedades del Desarrollo Óseo/metabolismo , Enfermedades del Desarrollo Óseo/patología , Preescolar , Trastornos de Deglución/genética , Trastornos de Deglución/metabolismo , Trastornos de Deglución/patología , Exostosis Múltiple Hereditaria/metabolismo , Exostosis Múltiple Hereditaria/patología , Humanos , Lactante , Recién Nacido , Mutación , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patología , Disautonomías Primarias/genética , Disautonomías Primarias/metabolismo , Disautonomías Primarias/patología
18.
J Bone Miner Res ; 37(2): 185-201, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34477239

RESUMEN

Breast cancer cells frequently home to the bone marrow, where they encounter signals that promote survival and quiescence or stimulate their proliferation. The interleukin-6 (IL-6) cytokines signal through the co-receptor glycoprotein130 (gp130) and are abundantly secreted within the bone microenvironment. Breast cancer cell expression of leukemia inhibitory factor (LIF) receptor (LIFR)/STAT3 signaling promotes tumor dormancy in the bone, but it is unclear which, if any of the cytokines that signal through LIFR, including LIF, oncostatin M (OSM), and ciliary neurotrophic factor (CNTF), promote tumor dormancy and which signaling pathways are induced. We first confirmed that LIF, OSM, and CNTF and their receptor components were expressed across a panel of breast cancer cell lines, although expression was lower in estrogen receptor-negative (ER- ) bone metastatic clones compared with parental cell lines. In estrogen receptor-positive (ER+ ) cells, OSM robustly stimulated phosphorylation of known gp130 signaling targets STAT3, ERK, and AKT, while CNTF activated STAT3 signaling. In ER- breast cancer cells, OSM alone stimulated AKT and ERK signaling. Overexpression of OSM, but not CNTF, reduced dormancy gene expression and increased ER+ breast cancer bone dissemination. Reverse-phase protein array revealed distinct and overlapping pathways stimulated by OSM, LIF, and CNTF with known roles in breast cancer progression and metastasis. In breast cancer patients, downregulation of the cytokines or receptors was associated with reduced relapse-free survival, but OSM was significantly elevated in patients with invasive disease and distant metastasis. Together these data indicate that the gp130 cytokines induce multiple signaling cascades in breast cancer cells, with a potential pro-tumorigenic role for OSM and pro-dormancy role for CNTF. © 2021 American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Neoplasias de la Mama , Receptor gp130 de Citocinas/metabolismo , Citocinas , Neoplasias de la Mama/genética , Citocinas/metabolismo , Femenino , Humanos , Interleucina-6/metabolismo , Receptores de Citocinas/metabolismo , Receptores OSM-LIF/genética , Receptores OSM-LIF/metabolismo , Transducción de Señal , Microambiente Tumoral
19.
Cells ; 11(21)2022 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-36359879

RESUMEN

Pancreatic cancer is a leading cause of cancer mortality and is projected to become the second-most common cause of cancer mortality in the next decade. While gene-wide association studies and next generation sequencing analyses have identified molecular patterns and transcriptome profiles with prognostic relevance, therapeutic opportunities remain limited. Among the genes that are upregulated in pancreatic ductal adenocarcinomas (PDAC), the leukaemia inhibitory factor (LIF), a cytokine belonging to IL-6 family, has emerged as potential therapeutic candidate. LIF is aberrantly secreted by tumour cells and promotes tumour progression in pancreatic and other solid tumours through aberrant activation of the LIF receptor (LIFR) and downstream signalling that involves the JAK1/STAT3 pathway. Since there are no LIFR antagonists available for clinical use, we developed an in silico strategy to identify potential LIFR antagonists and drug repositioning with regard to LIFR antagonists. The results of these studies allowed the identification of mifepristone, a progesterone/glucocorticoid antagonist, clinically used in medical abortion, as a potent LIFR antagonist. Computational studies revealed that mifepristone binding partially overlapped the LIFR binding site. LIF and LIFR are expressed by human PDAC tissues and PDAC cell lines, including MIA-PaCa-2 and PANC-1 cells. Exposure of these cell lines to mifepristone reverses cell proliferation, migration and epithelial mesenchymal transition induced by LIF in a concentration-dependent manner. Mifepristone inhibits LIFR signalling and reverses STAT3 phosphorylation induced by LIF. Together, these data support the repositioning of mifepristone as a potential therapeutic agent in the treatment of PDAC.


Asunto(s)
Adenocarcinoma , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Embarazo , Femenino , Humanos , Receptores OSM-LIF/genética , Mifepristona/farmacología , Mifepristona/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Reposicionamiento de Medicamentos , Carcinoma Ductal Pancreático/patología , Antagonistas de Hormonas/farmacología , Neoplasias Pancreáticas
20.
Endocrinology ; 162(11)2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34402888

RESUMEN

Recent studies have demonstrated that the formation of an implantation chamber composed of a uterine crypt, an implantation-competent blastocyst, and uterine glands is a critical step in blastocyst implantation in mice. Leukemia inhibitory factor (LIF) activates signal transducer and activator of transcription 3 (STAT3) precursors via uterine LIF receptors (LIFRs), allowing successful blastocyst implantation. Our recent study revealed that the role of epithelial STAT3 is different from that of stromal STAT3. However, both are essential for blastocyst attachment, suggesting the different roles of epithelial and stromal LIFR in blastocyst implantation. However, how epithelial and stromal LIFR regulate the blastocyst implantation process remains unclear. To investigate the roles of LIFR in the uterine epithelium and stroma, we generated Lifr-floxed/lactoferrin (Ltf)-iCre (Lifr eKO) and Lifr-floxed/antimüllerian hormone receptor type 2 (Amhr2)-Cre (Lifr sKO) mice with deleted epithelial and stromal LIFR, respectively. Surprisingly, fertility and blastocyst implantation in the Lifr sKO mice were normal despite stromal STAT3 inactivation. In contrast, blastocyst attachment failed, and no implantation chambers were formed in the Lifr eKO mice with epithelial inactivation of STAT3. In addition, normal responsiveness to ovarian hormones was observed in the peri-implantation uteri of the Lifr eKO mice. These results indicate that the epithelial LIFR-STAT3 pathway initiates the formation of implantation chambers, leading to complete blastocyst attachment, and that stromal STAT3 regulates blastocyst attachment without stromal LIFR control. Thus, uterine epithelial LIFR is critical to implantation chamber formation and blastocyst attachment.


Asunto(s)
Implantación del Embrión/genética , Epitelio/metabolismo , Receptores OSM-LIF/fisiología , Útero/metabolismo , Animales , Blastocisto/fisiología , Decidua/fisiología , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Embarazo , Receptores OSM-LIF/genética , Receptores OSM-LIF/metabolismo , Útero/citología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda