Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Genes (Basel) ; 12(9)2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34573352

RESUMEN

The function of the immune system extends from defense against external pathogens to the recognition and elimination of mutated or dying cells, aiding elimination of malignant potential and/or maintaining homeostasis. The many cell types of the immune system secrete a broad range of factors to enable cellular signaling that is vital to physiological processes. Additionally, in the ovary, follicular selection and maturation, as well as ovulation, are directly regulated by the nearby immune cells. Additionally, ovulation and rupture of the follicle have been observed to resemble a local inflammatory response. Cells of the cumulus-oocyte complex (COC) show evolving gene expression profiles throughout the oocytes' lifespan, including genes associated with immunological processes. Analysis of these genes allows the identification of useful molecular markers, as well as highlighting gene functions and interactions in these cells. Cumulus cells were obtained from hormonally stimulated patients undergoing an in vitro fertilization procedure and studied under long-term culture conditions. The microarray technique made it possible to compare the level of CCs' gene expression on the 1st, 7th, 15th and 30th day of cultivation. Additionally, RNA microarray analysis was performed to map gene expression in these cells, associated with immunological processes and associated cytokine signaling. Subsequently, the use of DAVID software allowed us to identify the "defense response to other organism", "defense response", "defense response to virus", "cytokine secretion", "cytokine production" and "cytokine-mediated signaling pathway" GO BP terms, as well as allowing further analysis of the most differentially expressed genes associated with these processes. Of the 122 genes involved, 121 were upregulated and only one was downregulated. The seven most upregulated genes related to the abovementioned terms were ANXA3, IFIT1, HLA-DPA1, MX1, KRT8, HLA-DRA and KRT18. Therefore, genes involved in immunological defense processes are upregulated in CC cultures and could serve as useful molecular markers of growth and development in the COC, as well as the proliferation of granulosa and cumulus cells.


Asunto(s)
Células del Cúmulo/inmunología , Citocinas/metabolismo , Inmunidad/genética , Oocitos/inmunología , Ovulación/inmunología , Adulto , Proliferación Celular/genética , Células Cultivadas , Células del Cúmulo/metabolismo , Femenino , Fertilización In Vitro , Perfilación de la Expresión Génica , Humanos , Oocitos/metabolismo , Ovulación/genética , Inducción de la Ovulación , Cultivo Primario de Células , Transducción de Señal/genética , Transducción de Señal/inmunología , Regulación hacia Arriba/inmunología
2.
J Biol Chem ; 295(41): 13958-13964, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-32587093

RESUMEN

The recently emerged severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the causative agent of the devastating COVID-19 lung disease pandemic. Here, we tested the inhibitory activities of the antiviral interferons of type I (IFN-α) and type III (IFN-λ) against SARS-CoV-2 and compared them with those against SARS-CoV-1, which emerged in 2003. Using two mammalian epithelial cell lines (human Calu-3 and simian Vero E6), we found that both IFNs dose-dependently inhibit SARS-CoV-2. In contrast, SARS-CoV-1 was restricted only by IFN-α in these cell lines. SARS-CoV-2 generally exhibited a broader IFN sensitivity than SARS-CoV-1. Moreover, ruxolitinib, an inhibitor of IFN-triggered Janus kinase/signal transducer and activator of transcription signaling, boosted SARS-CoV-2 replication in the IFN-competent Calu-3 cells. We conclude that SARS-CoV-2 is sensitive to exogenously added IFNs. This finding suggests that type I and especially the less adverse effect-prone type III IFN are good candidates for the management of COVID-19.


Asunto(s)
Antivirales/farmacología , Betacoronavirus/efectos de los fármacos , Interferón Tipo I/farmacología , Interferones/farmacología , Animales , Betacoronavirus/aislamiento & purificación , Betacoronavirus/fisiología , COVID-19 , Línea Celular , Chlorocebus aethiops , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Humanos , Quinasas Janus/metabolismo , Nitrilos , Pandemias , Neumonía Viral/patología , Neumonía Viral/virología , Pirazoles/farmacología , Pirimidinas , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , SARS-CoV-2 , Transducción de Señal/efectos de los fármacos , Células Vero , Replicación Viral/efectos de los fármacos , Interferón lambda
3.
J Biol Chem ; 293(45): 17631-17645, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30242126

RESUMEN

Upon repeated exposure to endotoxin or lipopolysaccharide (LPS), myeloid cells enter a refractory state called endotoxin tolerance as a homeostatic mechanism. In innate immune cells, LPS is recognized by co-receptors Toll-like receptor 4 (TLR4) and CD-14 to initiate an inflammatory response for subsequent cytokine production. One such cytokine, interleukin (IL)-27, is produced by myeloid cells in response to bacterial infection. In monocytes, IL-27 has proinflammatory functions such as up-regulating TLR4 expression for enhanced LPS-mediated cytokine production; alternatively, IL-27 induces inhibitory functions in activated macrophages. This study investigated the effects of IL-27 on the induction of endotoxin tolerance in models of human monocytes compared with macrophages. Our data demonstrate that IL-27 inhibits endotoxin tolerance by up-regulating cell surface TLR4 expression and soluble CD14 production to mediate stability of the surface LPS-TLR4-CD14 complex in THP-1 cells. In contrast, elevated basal expression of membrane-bound CD14 in phorbol 12-myristate 13-acetate (PMA)-THP-1 cells, primary monocytes, and primary macrophages may promote CD14-mediated endocytosis and be responsible for the preservation of an endotoxin-tolerized state in the presence of IL-27. Overall, the efficacy of IL-27 in inhibiting endotoxin tolerance in human THP-1 monocytes and PMA-THP-1 macrophages is affected by membrane-bound and soluble CD14 expression.


Asunto(s)
Tolerancia Inmunológica/efectos de los fármacos , Interleucinas/inmunología , Receptores de Lipopolisacáridos/inmunología , Lipopolisacáridos/toxicidad , Macrófagos/inmunología , Modelos Inmunológicos , Monocitos/inmunología , Endocitosis/efectos de los fármacos , Endocitosis/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/inmunología , Humanos , Activación de Macrófagos/efectos de los fármacos , Células THP-1 , Acetato de Tetradecanoilforbol/farmacología , Receptor Toll-Like 4/inmunología
4.
J Biol Chem ; 293(6): 1957-1975, 2018 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-29284679

RESUMEN

Alzheimer's disease (AD) is a disabling and highly prevalent neurodegenerative condition, for which there are no effective therapies. Soluble oligomers of the amyloid-ß peptide (AßOs) are thought to be proximal neurotoxins involved in early neuronal oxidative stress and synapse damage, ultimately leading to neurodegeneration and memory impairment in AD. The aim of the current study was to evaluate the neuroprotective potential of mesenchymal stem cells (MSCs) against the deleterious impact of AßOs on hippocampal neurons. To this end, we established transwell cocultures of rat hippocampal neurons and MSCs. We show that MSCs and MSC-derived extracellular vesicles protect neurons against AßO-induced oxidative stress and synapse damage, revealed by loss of pre- and postsynaptic markers. Protection by MSCs entails three complementary mechanisms: 1) internalization and degradation of AßOs; 2) release of extracellular vesicles containing active catalase; and 3) selective secretion of interleukin-6, interleukin-10, and vascular endothelial growth factor to the medium. Results support the notion that MSCs may represent a promising alternative for cell-based therapies in AD.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Vesículas Extracelulares/metabolismo , Hipocampo/citología , Células Madre Mesenquimatosas/citología , Neuronas/metabolismo , Estrés Oxidativo , Sinapsis/metabolismo , Enfermedad de Alzheimer/genética , Péptidos beta-Amiloides/química , Animales , Células Cultivadas , Técnicas de Cocultivo , Vesículas Extracelulares/genética , Hipocampo/metabolismo , Humanos , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Neuronas/citología , Ratas , Ratas Wistar , Factor A de Crecimiento Endotelial Vascular/metabolismo
5.
J Biol Chem ; 291(42): 22030-22042, 2016 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-27590344

RESUMEN

Expression of DAPK1, a critical regulator of autophagy and apoptosis, is lost in a wide variety of tumors, although the mechanisms are unclear. A transcription factor complex consisting of ATF6 (an endoplasmic reticulum-resident factor) and C/EBP-ß is required for the IFN-γ-induced expression of DAPK1 IFN-γ-induced proteolytic processing of ATF6 and phosphorylation of C/EBP-ß are obligatory for the formation of this transcriptional complex. We report that defects in this pathway fail to control growth of chronic lymphocytic leukemia (CLL). Consistent with these observations, IFN-γ and chemotherapeutics failed to activate autophagy in CLL patient samples lacking ATF6 and/or C/EBP-ß. Together, these results identify a molecular basis for the loss of DAPK1 expression in CLL.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Autofagia , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteínas Quinasas Asociadas a Muerte Celular/biosíntesis , Regulación Enzimológica de la Expresión Génica , Regulación Leucémica de la Expresión Génica , Leucemia Linfocítica Crónica de Células B/metabolismo , Proteínas de Neoplasias/metabolismo , Factor de Transcripción Activador 6/genética , Proteína beta Potenciadora de Unión a CCAAT/genética , Línea Celular Transformada , Proteínas Quinasas Asociadas a Muerte Celular/genética , Femenino , Humanos , Interferón gamma/farmacología , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/genética , Masculino , Proteínas de Neoplasias/genética
6.
J Biol Chem ; 290(40): 24561-73, 2015 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-26296889

RESUMEN

Lineage specification in the hematopoietic system depends on the expression of lineage specific transcription factors. However, the role of hematopoietic cytokines in this process has been controversial and little is known about the intracellular signaling mechanisms by which cytokines instruct lineage choice. G-CSF and M-CSF are two lineage-specific cytokines that play a dominant role in granulopoiesis and monopoiesis, respectively. We show here that a G-CSFR mutant in which tyrosine 729 had been mutated to phenylalanine (Y729F) promoted monocyte rather than neutrophil development in myeloid precursors, which was associated with prolonged activation of Erk1/2 and augmented activation of downstream targets c-Fos and Egr1. Inhibition of Erk1/2 activation or knockdown of c-Fos or Egr1 largely rescued neutrophil development in cells expressing G-CSFR Y729F. We also show that M-CSF, but not G-CSF, stimulated strong and sustained activation of Erk1/2 in mouse lineage marker negative (Lin(-)) bone marrow cells. Significantly, inhibition of Erk1/2 signaling in these cells favored neutrophil over monocyte development in response to M-CSF. Thus, prolonged Erk1/2 activation resulted in monocyte development following G-CSF induction whereas inhibition of Erk1/2 signaling promoted neutrophil development at the expense of monocyte formation in response to M-CSF. These results reveal an important mechanism by which G-CSF and M-CSF instruct neutrophil versus monocyte lineage choice, i.e. differential activation of Erk1/2 pathway.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/metabolismo , Sistema de Señalización de MAP Quinasas , Factor Estimulante de Colonias de Macrófagos/metabolismo , Monocitos/enzimología , Neutrófilos/enzimología , Animales , Apoptosis , Células de la Médula Ósea/citología , Línea Celular , Linaje de la Célula , Citocinas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Ratones , Mutación , Mielopoyesis/fisiología , Interferencia de ARN , Transducción de Señal , Tirosina/química
7.
J Biol Chem ; 290(37): 22338-51, 2015 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-26183780

RESUMEN

The common γ molecule (γc) is a shared signaling receptor subunit used by six γc-cytokines. These cytokines play crucial roles in the differentiation of the mature immune system and are involved in many human diseases. Moreover, recent studies suggest that multiple γc-cytokines are pathogenically involved in a single disease, thus making the shared γc-molecule a logical target for therapeutic intervention. However, the current therapeutic strategies seem to lack options to treat such cases, partly because of the lack of appropriate neutralizing antibodies recognizing the γc and, more importantly, because of the inherent and practical limitations in the use of monoclonal antibodies. By targeting the binding interface of the γc and cytokines, we successfully designed peptides that not only inhibit multiple γc-cytokines but with a selectable target spectrum. Notably, the lead peptide inhibited three γc-cytokines without affecting the other three or non-γc-cytokines. Biological and mutational analyses of our peptide provide new insights to our current understanding on the structural aspect of the binding of γc-cytokines the γc-molecule. Furthermore, we provide evidence that our peptide, when conjugated to polyethylene glycol to gain stability in vivo, efficiently blocks the action of one of the target cytokines in animal models. Collectively, our technology can be expanded to target various combinations of γc-cytokines and thereby will provide a novel strategy to the current anti-cytokine therapies against immune, inflammatory, and malignant diseases.


Asunto(s)
Citocinas/antagonistas & inhibidores , Subunidad gamma Común de Receptores de Interleucina/antagonistas & inhibidores , Péptidos/farmacología , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Femenino , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Masculino , Péptidos/química , Péptidos/genética
8.
J Biol Chem ; 290(11): 7195-207, 2015 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-25635047

RESUMEN

Prolyl-4-hydroxylase (PHD) proteins are key in sensing tissue hypoxia. In nucleus pulposus (NP) cells, our previous work demonstrated that PHD isoforms have a differential contribution in controlling hypoxia-inducible factor (HIF)-α degradation and activity. Recently we have shown that a regulatory relationship exists between PHD3 and inflammatory cytokines in NP cells. With respect to PHD2, the most abundant PHD isoform in NP cells, very little is known concerning its function and regulation under inflammatory conditions that characterize intervertebral disc degeneration. Here, we show that PHD2 is a potent regulator of the catabolic activities of TNF-α; silencing of PHD2 significantly decreased TNF-α-induced expression of catabolic markers including SDC4, MMP-3, MMP-13, and ADAMTS5, as well as several inflammatory cytokines and chemokines, while partially restoring aggrecan and collagen II expression. Use of NF-κB reporters with ShPHD2, SiHIF-1α, as well as p65(-/-), PHD2(-/-), and PHD3(-/-) cells, shows that PHD2 serves as a co-activator of NF-κB/p65 signaling in HIF-1-independent fashion. Immunoprecipitation of endogenous and exogenously expressed tagged proteins, as well as fluorescence microscopy, indicates that following TNF-α treatment, PHD2 interacts and co-localizes with p65. Conversely, loss of function experiments using lentivirally delivered Sh-p65, Sh-IKKß, and NF-κB inhibitor confirmed that cytokine-dependent PHD2 expression in NP cells requires NF-κB signaling. These findings clearly demonstrate that PHD2 forms a regulatory circuit with TNF-α via NF-κB and thereby plays an important role in enhancing activity of this cytokine. We propose that during disc degeneration PHD2 may offer a therapeutic target to mitigate the deleterious actions of TNF-α, a key proinflammatory cytokine.


Asunto(s)
Interleucina-1beta/inmunología , Disco Intervertebral/citología , FN-kappa B/inmunología , Procolágeno-Prolina Dioxigenasa/inmunología , Factor de Transcripción ReIA/genética , Activación Transcripcional , Factor de Necrosis Tumoral alfa/inmunología , Animales , Células Cultivadas , Regulación de la Expresión Génica , Silenciador del Gen , Células HEK293 , Humanos , Prolina Dioxigenasas del Factor Inducible por Hipoxia , Degeneración del Disco Intervertebral/genética , Degeneración del Disco Intervertebral/inmunología , FN-kappa B/genética , Procolágeno-Prolina Dioxigenasa/genética , Ratas Wistar , Transducción de Señal , Factor de Transcripción ReIA/inmunología
9.
J Biol Chem ; 290(1): 359-70, 2015 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-25371211

RESUMEN

IL-23, composed of the cytokine subunit p19 and the soluble α receptor subunit p40, binds to a receptor complex consisting of the IL-23 receptor (IL-23R) and the IL-12 receptor ß1 (IL-12Rß1). Complex formation was hypothesized to follow the "site I-II-III" architectural paradigm, with site I of p19 being required for binding to p40, whereas sites II and III of p19 mediate binding to IL-12Rß1 and IL-23R, respectively. Here we show that the binding mode of p19 to p40 and of p19 to IL-23R follow the canonical site I and III paradigm but that interaction of IL-23 to IL-12Rß1 is independent of site II in p19. Instead, binding of IL-23 to the cytokine binding module of IL-12Rß1 is mediated by domains 1 and 2 of p40 via corresponding site II amino acids of IL-12Rß1. Moreover, domains 2 and 3 of p40 were sufficient for complex formation with p19 and to induce binding of p19 to IL-23R. The Fc-tagged fusion protein of p40_D2D3/p19 did, however, not act as a competitive IL-23 antagonist but, at higher concentrations, induced proliferation via IL-23R but independent of IL-12Rß1. On the basis of our experimental validation, we propose a non-canonical topology of the IL-23·IL-23R·IL-12Rß1 complex. Furthermore, our data help to explain why p40 is an antagonist of IL-23 and IL-12 signaling and show that site II of p19 is dispensable for IL-23 signaling.


Asunto(s)
Sudunidad beta 1 del Receptor de Interleucina-12/química , Subunidad p40 de la Interleucina-12/química , Interleucina-23/química , Receptores de Interleucina-12/química , Receptores de Interleucina/química , Animales , Sitios de Unión , Células CHO , Células COS , Línea Celular , Chlorocebus aethiops , Cricetulus , Expresión Génica , Humanos , Sudunidad beta 1 del Receptor de Interleucina-12/genética , Sudunidad beta 1 del Receptor de Interleucina-12/metabolismo , Subunidad p40 de la Interleucina-12/genética , Subunidad p40 de la Interleucina-12/metabolismo , Interleucina-23/genética , Interleucina-23/metabolismo , Ratones , Modelos Moleculares , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Receptores de Interleucina-12/genética , Receptores de Interleucina-12/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
10.
J Biol Chem ; 289(26): 18442-50, 2014 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-24802752

RESUMEN

Ciliary neurotrophic factor (CNTF) is a neurotrophic factor with therapeutic potential for neurodegenerative diseases. Moreover, therapeutic application of CNTF reduced body weight in mice and humans. CNTF binds to high or low affinity receptor complexes consisting of CNTFR·gp130·LIFR or IL-6R·gp130·LIFR, respectively. Clinical studies of the CNTF derivative Axokine revealed intolerance at higher concentrations, which may rely on the low-affinity binding of CNTF to the IL-6R. Here, we aimed to generate a CNTFR-selective CNTF variant (CV). CV-1 contained the single amino acid exchange R28E. Arg(28) is in close proximity to the CNTFR binding site. Using molecular modeling, we hypothesized that Arg(28) might contribute to IL-6R/CNTFR plasticity of CNTF. CV-2 to CV-5 were generated by transferring parts of the CNTFR-binding site from cardiotrophin-like cytokine to CNTF. Cardiotrophin-like cytokine selectively signals via the CNTFR·gp130·LIFR complex, albeit with a much lower affinity compared with CNTF. As shown by immunoprecipitation, all CNTF variants retained the ability to bind to CNTFR. CV-1, CV-2, and CV-5, however, lost the ability to bind to IL-6R. Although all variants induced cytokine-dependent cellular proliferation and STAT3 phosphorylation via CNTFR·gp130·LIFR, only CV-3 induced STAT3 phosphorylation via IL-6R·gp130·LIFR. Quantification of CNTF-dependent proliferation of CNTFR·gp130·LIFR expressing cells indicated that only CV-1 was as biologically active as CNTF. Thus, the CNTFR-selective CV-1 will allow discriminating between CNTFR- and IL-6R-mediated effects in vivo.


Asunto(s)
Sustitución de Aminoácidos , Factor Neurotrófico Ciliar/genética , Receptor gp130 de Citocinas/metabolismo , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/metabolismo , Receptor de Factor Neurotrófico Ciliar/metabolismo , Receptores de Interleucina-6/metabolismo , Factor Neurotrófico Ciliar/metabolismo , Receptor gp130 de Citocinas/genética , Humanos , Interleucina-6/metabolismo , Subunidad alfa del Receptor del Factor Inhibidor de Leucemia/genética , Mutación Missense , Fosforilación , Receptor de Factor Neurotrófico Ciliar/genética , Receptores de Interleucina-6/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal
11.
Int J Biochem Cell Biol ; 46: 113-23, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24275094

RESUMEN

Atherosclerosis is an inflammatory disorder of the vasculature regulated by cytokines. Amongst the cytokines, IL-33 attenuates the development of atherosclerosis in mouse model systems via several mechanisms, including inhibition of macrophage foam cell formation and promotion of a Th1 to Th2 shift. Proteases produced by macrophages, such as matrix metalloproteinases and members of ADAMTS (a disintegrin and metalloproteinase with thrombospondin motifs) family, play potential roles in regulating atherosclerotic plaque stability. Despite such importance, the action of IL-33 on the expression of such proteases has not been analyzed. We have therefore investigated the effect of IL-33 on the expression of ADAMTS-1, -4 and -5 in human macrophages. Immunohistochemical analysis showed that these three proteases were expressed in human atherosclerotic lesions, particularly by macrophages and, to a lesser extent, by smooth muscle cells and endothelial cells. The expression of ADAMTS-1, -4 and -5 in human macrophages was specifically inhibited by IL-33. The action of IL-33 on the expression of these ADAMTS members was mediated through its receptor ST2. IL-33 activated ERK1/2, JNK1/2 and c-Jun, but not p38 MAPK or Akt, in human macrophages. RNA interference assays using a combination of adenoviral encoding small hairpin RNA and small interfering RNA showed a requirement of ERK1/2, JNK1/2, c-Jun, PI3Kγ and PI3Kδ, but not p38α, in the IL-33-inhibited expression of these ADAMTS isoforms. These studies provide novel insights into the expression of ADAMTS-1, -4 and -5 in human atherosclerotic lesions and the regulation of their expression in human macrophages by the key anti-atherogenic cytokine IL-33.


Asunto(s)
Citocinas/metabolismo , Desintegrinas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Interleucinas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Metaloproteasas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Trombospondinas/metabolismo , Animales , Aterosclerosis/metabolismo , Humanos , Macrófagos/metabolismo , Ratones , Transducción de Señal
12.
Cancer Lett ; 344(1): 47-53, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24157812

RESUMEN

Invasive breast tumor cells generate three splice variants of the metastasis gene osteopontin, while non-invasive breast cells express only the unspliced form or no osteopontin at all. One role for osteopontin in tumor progression is the support of anchorage-independence. Here we show that the full-length gene product, osteopontin-a, induces a gene expression profile that is associated with tissue remodeling and directed movement/sprouting. This occurs via signals through STAT1 and STAT3 to sn-glycero-3-phosphocholine. Osteopontin-a upregulates the levels of glucose in breast cancer cells, likely through STAT3 and its transcriptional targets apolipoprotein D and IGFBP5. The splice variants osteopontin-a and osteopontin-c may synergize, with each form activating signal transduction pathways that are distinct from the other. The elevated glucose is used by osteopontin-c dependent signals to generate chemical energy (Shi et al. submitted for publication). The splice variant-specific metabolic effects of osteopontin add a novel aspect to the pro-metastatic functions of this molecule.


Asunto(s)
Neoplasias de la Mama/metabolismo , Glucosa/metabolismo , Homeostasis/fisiología , Invasividad Neoplásica , Osteopontina/metabolismo , Neoplasias de la Mama/patología , Adhesión Celular , Humanos , Immunoblotting , Células MCF-7 , Metabolómica , Isoformas de Proteínas , Transducción de Señal/fisiología , Transfección
13.
J Biol Chem ; 289(1): 65-73, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24280222

RESUMEN

Growth hormone (GH) signaling is required for promoting longitudinal body growth, stem cell activation, differentiation, and survival and for regulation of metabolism. Failure to adequately regulate GH signaling leads to disease: excessive GH signaling has been connected to cancer, and GH insensitivity has been reported in cachexia patients. Since its discovery in 1989, the receptor has served a pivotal role as the prototype cytokine receptor both structurally and functionally. Phosphorylation and ubiquitylation regulate the GH receptor (GHR) at the cell surface: two ubiquitin ligases (SCF(ßTrCP2) and CHIP) determine the GH responsiveness of cells by controlling its endocytosis, whereas JAK2 initiates the JAK/STAT pathway. We used blue native electrophoresis to identify phosphorylated and ubiquitylated receptor intermediates. We show that GHRs occur as ∼500-kDa complexes that dimerize into active ∼900-kDa complexes upon GH binding. The dimerized complexes act as platforms for transient interaction with JAK2 and ubiquitin ligases. If GH and receptors are made in the same cell (autocrine mode), only limited numbers of ∼900-kDa complexes are formed. The experiments reveal the dynamic changes in post-translational modifications during GH-induced signaling events and show that relatively simple cytokine receptors like GHRs are able to form higher order protein complexes. Insight in the complex formation of cytokine receptors is crucially important for engineering cytokines that control ligand-induced cell responses and for generating a new class of therapeutic agents for a wide range of diseases.


Asunto(s)
Complejos Multiproteicos/metabolismo , Multimerización de Proteína/fisiología , Receptores de Somatotropina/metabolismo , Transducción de Señal/fisiología , Animales , Comunicación Autocrina/fisiología , Células HEK293 , Hormona de Crecimiento Humana/genética , Hormona de Crecimiento Humana/metabolismo , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Ratones , Complejos Multiproteicos/genética , Fosforilación/fisiología , Receptores de Somatotropina/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/fisiología , Proteínas con Repetición de beta-Transducina/genética , Proteínas con Repetición de beta-Transducina/metabolismo
14.
J Biol Chem ; 288(49): 35159-69, 2013 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-24151074

RESUMEN

Satellite cell self-renewal is an essential process to maintaining the robustness of skeletal muscle regenerative capacity. However, extrinsic factors that regulate self-renewal of satellite cells are not well understood. Here, we demonstrate that TWEAK cytokine reduces the proportion of Pax7(+)/MyoD(-) cells (an index of self-renewal) on myofiber explants and represses multiple components of Notch signaling in satellite cell cultures. The number of Pax7(+) cells is significantly increased in skeletal muscle of TWEAK knock-out (KO) mice compared with wild-type in response to injury. Furthermore, Notch signaling is significantly elevated in cultured satellite cells and in regenerating myofibers of TWEAK-KO mice. Forced activation of Notch signaling through overexpression of the Notch1 intracellular domain (N1ICD) rescued the TWEAK-mediated inhibition of satellite cell self-renewal. TWEAK also activates the NF-κB transcription factor in satellite cells and inhibition of NF-κB significantly improved the number of Pax7(+) cells in TWEAK-treated cultures. Furthermore, our results demonstrate that a reciprocal interaction between NF-κB and Notch signaling governs the inhibitory effect of TWEAK on satellite cell self-renewal. Collectively, our study demonstrates that TWEAK suppresses satellite cell self-renewal through activating NF-κB and repressing Notch signaling.


Asunto(s)
FN-kappa B/metabolismo , Receptor Notch1/metabolismo , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/metabolismo , Factores de Necrosis Tumoral/metabolismo , Animales , Apoptosis/fisiología , Proliferación Celular , Células Cultivadas , Citocina TWEAK , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína MioD/genética , Proteína MioD/metabolismo , FN-kappa B/genética , Factor de Transcripción PAX7/genética , Factor de Transcripción PAX7/metabolismo , Estructura Terciaria de Proteína , Receptor Notch1/química , Receptor Notch1/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Regeneración/genética , Regeneración/fisiología , Transducción de Señal , Factores de Necrosis Tumoral/deficiencia , Factores de Necrosis Tumoral/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA