Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Stroke ; 50(7): 1850-1858, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31167620

RESUMEN

Background and Purpose- Ischemic stroke is one of the leading causes of morbidity and mortality worldwide and a major cause of long-term disability. Recently, long noncoding RNAs have been revealed, which are tightly associated with several human diseases. However, the functions of long noncoding RNAs in ischemic stroke still remain largely unknown. In the current study, for the first time, we investigated the role of long noncoding RNA Nespas in ischemic stroke. Methods- We used in vivo models of middle cerebral artery occlusion and in vitro models of oxygen-glucose deprivation to illustrate the effect of long noncoding RNA Nespas on ischemic stroke. Results- We found expression of Nespas was significantly increased in ischemic cerebral tissues and oxygen-glucose deprivation-treated BV2 cells in a time-dependent manner. Silencing of Nespas aggravated middle cerebral artery occlusion operation-induced IR injury and cell death. In addition, proinflammatory cytokine production and NF-κB (nuclear factor-κB) signaling activation were inhibited by Nespas overexpression. TAK1 (transforming growth factor-ß-activated kinase 1) was found to directly interact with Nespas, and TAK1 activation was significantly suppressed by Nespas. At last, we found Nespas-inhibited TRIM8 (tripartite motif 8)-induced K63-linked polyubiquitination of TAK1. Conclusions- We showed that Nespas played anti-inflammatory and antiapoptotic roles in cultured microglial cells after oxygen-glucose deprivation stimulation and in mice after ischemic stroke by inhibiting TRIM8-related K63-linked polyubiquitination of TAK1.


Asunto(s)
Isquemia Encefálica/patología , Muerte Celular , Silenciador del Gen , Inflamación/patología , Microglía/patología , ARN Largo no Codificante/genética , Accidente Cerebrovascular/patología , Animales , Células Cultivadas , Citocinas/metabolismo , Humanos , Infarto de la Arteria Cerebral Media/patología , Quinasas Quinasa Quinasa PAM/biosíntesis , Quinasas Quinasa Quinasa PAM/genética , Ratones , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Ubiquitina-Proteína Ligasas/biosíntesis , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación/genética
2.
Neurobiol Dis ; 103: 133-143, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28396258

RESUMEN

OBJECTIVE: Dual leucine zipper kinase (DLK/MA3K12) has been reported involved in apoptosis and neuronal degeneration during neural development and traumatic brain injury. This study was designed to investigate the role of DLK with its adaptor protein JNK interacting protein-3 (JIP3), and its downstream MA2K7/JNK signaling pathway in early brain injury (EBI) after subarachnoid hemorrhage (SAH) in a rat model. DESIGN: Controlled in vivo laboratory study. SETTING: Animal research laboratory. SUBJECTS: Two hundred and twenty-three adult male Sprague-Dawley rats weighing 280-320g. INTERVENTIONS: SAH was induced by endovascular perforation in rats. The SAH grade, neurological score, and brain water content were measured at 24 and 72h after SAH. Immunofluorescence staining was used to detect the cells that expressed DLK. The terminal deoxynucleotid transferase-deoxyuridine triphosphate (dUTP) nick end labeling (TUNEL) was used to detect the neuronal apoptosis. In mechanism research, the expression of DLK, JIP3, phosphorylated-JNK (p-JNK)/JNK, and cleaved caspase-3 (CC-3) were analyzed by western blot at 24h after SAH. The DLK small interfering RNA (siRNA), JIP3 siRNA, MA2K7 siRNA and recombinant DLK protein which injected intracerebroventricularly were given as the interventions. MEASUREMENTS AND MAIN RESULTS: The DLK expression was increased in the left cortex neurons and peaked at 24h after SAH. DLK siRNA attenuated brain edema, reduced neuronal apoptosis, and improved the neurobehavioral functions after SAH, but the recombinant DLK protein deteriorated neurobehavioral functions and brain edema. DLK siRNA decreased and recombinant DLK protein increased the expression of MA2K7/p-JNK/CC-3 at 24h after SAH. The JIP3 siRNA reduced the level of JIP3/MA2K7/p-JNK/CC-3, combined DLK siRNA and JIP3 siRNA further decreased the expression of DLK/MA2K7/p-JNK/CC-3, and MA2K7 siRNA lowered the amount of MA2K7/p-JNK/CC-3 at 24h after SAH. CONCLUSIONS: As a negative role, DLK was involved in EBI after SAH, possibly mediated by its adaptor protein JIP3 and MA2K7/JNK signaling pathways. To reduce the level of DLK may be a new target as intervention for SAH.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Lesiones Encefálicas/metabolismo , Silenciador del Gen/fisiología , Quinasas Quinasa Quinasa PAM/biosíntesis , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas del Tejido Nervioso/biosíntesis , Hemorragia Subaracnoidea/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis/fisiología , Lesiones Encefálicas/genética , Lesiones Encefálicas/patología , Quinasas Quinasa Quinasa PAM/genética , Masculino , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Sprague-Dawley , Hemorragia Subaracnoidea/genética , Hemorragia Subaracnoidea/patología
3.
IUBMB Life ; 68(2): 145-55, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26748475

RESUMEN

Inflammation of infiltrated macrophages in adipose tissue is a key contributor to the initiation of adipose insulin resistance. These macrophages are exposed to high local concentrations of free fatty acids (FFAs) and can be proinflammatory activated by saturated fatty acids (SFAs). However, the regulatory mechanisms on SFA-induced macrophage inflammation are still elusive. Peroxisome proliferator-activated receptor γ coactivator-1ß (PGC-1ß) is a member of the PGC-1 family of transcriptional coactivators and has been reported to play a key role in SFAs metabolism and in the regulation of inflammatory signaling. However, it remains unclear whether PGC-1ß is involved in SFA-induced macrophage inflammation. In this study, we found that PGC-1ß expression was significantly decreased in response to palmitic acid (PA) in macrophages in a dose dependent manner. PGC-1ß inhibited PA induced TNFα, MCP-1, and IL-1ß mRNA and protein expressions. Furthermore, PGC-1ß significantly antagonized PA induced macrophage nuclear factor-κB (NF-κB) p65 and JUN N-terminal kinase activation. Mechanistically, we revealed that TGF-ß-activated kinase 1 (TAK1) and its adaptor protein TAK1 binding protein 1 (TAB1) played a dominant role in the regulatory effects of PGC-1ß. We confirmed that PGC-1ß inhibited downstream inflammatory signals via binding with TAB1 and thus preventing TAB1/TAK1 binding and TAK1 activation. Finally, we showed that PGC-1ß overexpression in PA treated macrophages improved adipocytes PI3K-Akt insulin signaling in a paracrine fashion. Collectively, our results uncovered a novel mechanism on how macrophage inflammation induced by SFAs was regulated and suggest a potential target in the treatment of obesity induced insulin resistance.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Tejido Adiposo/metabolismo , Inflamación/genética , Quinasas Quinasa Quinasa PAM/genética , Factores de Transcripción/genética , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Tejido Adiposo/efectos de los fármacos , Animales , Quimiocina CCL2/biosíntesis , Regulación de la Expresión Génica , Humanos , Inflamación/inducido químicamente , Inflamación/patología , Resistencia a la Insulina/genética , Interleucina-1beta/biosíntesis , Quinasas Quinasa Quinasa PAM/biosíntesis , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Ácido Palmítico/toxicidad , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Factores de Transcripción/biosíntesis , Activación Transcripcional/genética , Factor de Necrosis Tumoral alfa/biosíntesis
4.
J Biol Chem ; 289(1): 299-311, 2014 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-24275658

RESUMEN

The induction of the intrinsic antiviral defense in mammals relies on the accumulation of foreign genetic material. As such, complete engagement of this response is limited to replication-competent viruses. Interferon regulatory factors (IRFs) are mediators of this defense with shared enhancer elements but display a spectrum of transcriptional potential. Here we describe a mechanism designed to enhance this response should a pathogen not be successfully inhibited. We find that activation of IRF7 results in the induction of MAP3K8 and restructuring of the antiviral transcriptome. MAP3K8 mediates the phosphorylation and repression of IRF3 homodimers to promote greater transcriptional activity through utilization of IRF3:IRF7 heterodimers. Among the genes influenced by the MAP3K8/IRF7 signaling axis are members of the SP100 gene family that serve as general transcriptional enhancers of the antiviral defense. We propose that this feed forward loop serves to reinforce the cellular response and is reserved for imminent threats to the host.


Asunto(s)
Factor 3 Regulador del Interferón/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Quinasas Quinasa Quinasa PAM/biosíntesis , Sistema de Señalización de MAP Quinasas , Proteínas Proto-Oncogénicas/biosíntesis , Vesiculovirus/metabolismo , Animales , Línea Celular , Inducción Enzimática/genética , Humanos , Factor 3 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/genética , Quinasas Quinasa Quinasa PAM/genética , Ratones , Multimerización de Proteína/genética , Proteínas Proto-Oncogénicas/genética , Infecciones por Rhabdoviridae/genética , Infecciones por Rhabdoviridae/metabolismo , Proteínas S100/genética , Proteínas S100/metabolismo , Vesiculovirus/genética
5.
J Cell Biochem ; 116(12): 2778-85, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25959272

RESUMEN

Although evidence suggests that the RAF/MEK/ERK pathway plays an important role in triple negative breast cancer (TNBC), resistance to MEK inhibitors has been observed in TNBC cells. Different mechanisms have been hypothesized to be involved in this phenomenon, including receptor tyrosine kinase-dependent activation of the PI3K/AKT pathway. In this study, we analyzed the effects of the MEK1/2 inhibitor selumetinib in combination with the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor gefitinib in a panel of TNBC cell lines that showed different levels of sensitivity to single-agent selumetinib: SUM-149 and MDA-MB-231 cells resulted to be sensitive, whereas SUM-159, MDA-MB-468 and HCC70 cells were relatively resistant to the drug. Treatment of TNBC cells with selumetinib produced an increase of the phosphorylation of the EGFR both in selumetinib-sensitive SUM-149, MDA-MB-231 and in selumetinib-resistant MDA-MB-468 TNBC cells. The combination of selumetinib and gefitinib resulted in a synergistic growth inhibitory effect in all the TNBC cell lines, although the IC50 was not reached in SUM-159 and MDA-MB-468 cells. This effect was associated with an almost complete suppression of ERK1/2 activation and a reduction of selumetinib-induced AKT phosphorylation. In addition, in selumetinib-sensitive TNBC cells the combination of selumetinib and gefitinib induced a significant G0/G1 cell cycle arrest and apoptosis. Taken together, our data demonstrated that blockade of the EGFR might efficiently increase the antitumor activity of selumetinib in a subgroup of TNBC and that this phenomenon might be related to the effects of such combination on both ERK1/2 and AKT activation.


Asunto(s)
Bencimidazoles/administración & dosificación , Receptores ErbB/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinazolinas/administración & dosificación , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Sinergismo Farmacológico , Receptores ErbB/biosíntesis , Femenino , Gefitinib , Humanos , Quinasas Quinasa Quinasa PAM/biosíntesis , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Oncogénica v-akt/genética , Inhibidores de Proteínas Quinasas/administración & dosificación , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
6.
BMC Cancer ; 14: 182, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24628919

RESUMEN

BACKGROUND: MLK3 gene mutations were described to occur in about 20% of microsatellite unstable gastrointestinal cancers and to harbor oncogenic activity. In particular, mutation P252H, located in the kinase domain, was found to have a strong transforming potential, and to promote the growth of highly invasive tumors when subcutaneously injected in nude mice. Nevertheless, the molecular mechanism underlying the oncogenic activity of P252H mutant remained elusive. METHODS: In this work, we performed Illumina Whole Genome arrays on three biological replicas of human HEK293 cells stably transfected with the wild-type MLK3, the P252H mutation and with the empty vector (Mock) in order to identify the putative signaling pathways associated with P252H mutation. RESULTS: Our microarray results showed that mutant MLK3 deregulates several important colorectal cancer- associated signaling pathways such as WNT, MAPK, NOTCH, TGF-beta and p53, helping to narrow down the number of potential MLK3 targets responsible for its oncogenic effects. A more detailed analysis of the alterations affecting the WNT signaling pathway revealed a down-regulation of molecules involved in the canonical pathway, such as DVL2, LEF1, CCND1 and c-Myc, and an up-regulation of DKK, a well-known negative regulator of canonical WNT signaling, in MLK3 mutant cells. Additionally, FZD6 and FZD10 genes, known to act as negative regulators of the canonical WNT signaling cascade and as positive regulators of the planar cell polarity (PCP) pathway, a non-canonic WNT pathway, were found to be up-regulated in P252H cells. CONCLUSION: The results provide an overall view of the expression profile associated with mutant MLK3, and they support the functional role of mutant MLK3 by showing a deregulation of several signaling pathways known to play important roles in the development and progression of colorectal cancer. The results also suggest that mutant MLK3 may be a novel modulator of WNT signaling, and pinpoint the activation of PCP pathway as a possible mechanism underlying the invasive potential of MLK3 mutant cells.


Asunto(s)
Neoplasias Colorrectales/genética , Quinasas Quinasa Quinasa PAM/genética , Transducción de Señal/genética , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Estudio de Asociación del Genoma Completo , Células HEK293 , Humanos , Quinasas Quinasa Quinasa PAM/biosíntesis , Mutagénesis Sitio-Dirigida , Proteina Quinasa Quinasa Quinasa 11 Activada por Mitógeno
7.
Biochem J ; 452(2): 359-65, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23557442

RESUMEN

Activation of PKR (double-stranded-RNA-dependent protein kinase) by DNA plasmids decreases translation, and limits the amount of recombinant protein produced by transiently transfected HEK (human embryonic kidney)-293 cells. Co-expression with Ebola virus VP35 (virus protein 35), which blocked plasmid activation of PKR, substantially increased production of recombinant TPL-2 (tumour progression locus 2)-ABIN-2 [A20-binding inhibitor of NF-κB (nuclear factor κB) 2]-NF-κB1 p105 complex. VP35 also increased expression of other co-transfected proteins, suggesting that VP35 could be employed generally to boost recombinant protein production by HEK-293 cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Ebolavirus/fisiología , Quinasas Quinasa Quinasa PAM/biosíntesis , Subunidad p50 de NF-kappa B/biosíntesis , Proteínas Proto-Oncogénicas/biosíntesis , Regulación hacia Arriba/genética , Proteínas Reguladoras y Accesorias Virales/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Ebolavirus/genética , Células HEK293 , Humanos , Quinasas Quinasa Quinasa PAM/genética , Complejos Multiproteicos/biosíntesis , Complejos Multiproteicos/genética , Subunidad p50 de NF-kappa B/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Transfección , Proteínas Reguladoras y Accesorias Virales/genética
8.
Br J Cancer ; 107(1): 129-36, 2012 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-22644295

RESUMEN

BACKGROUND: Tumour cell metastasis involves cell adhesion and invasion, processes that depend on signal transduction, which can be influenced by the tumour microenvironment. N-6 polyunsaturated fatty acids, found both in the diet and in response to inflammatory responses, are important components of this microenvironment. METHODS: We used short hairpin RNA (shRNA) knockdown of TGF-ß-activated kinase-1 (TAK1) in human tumour cells to examine its involvement in fatty acid-stimulated cell adhesion and invasion in vitro. An in vivo model of metastasis was developed in which cells, stably expressing firefly luciferase and either a control shRNA or a TAK1-specific shRNA, were injected into the mammary fat pads of mice fed diets, rich in n-6 polyunsaturated fatty acids. Tumour growth and spontaneous metastasis were monitored with in vivo and in situ imaging of bioluminescence. RESULTS: Arachidonic acid activated TAK1 and downstream kinases in MDA-MB-435 breast cancer cells and led to increased adhesion and invasion. Knockdown of TAK1 blocked this activation and inhibited both cell adhesion and invasion in vitro. Tumour growth at the site of injection was not affected by TAK1 knockdown, but both the incidence and extent of metastasis to the lung were significantly reduced in mice injected with TAK1 knockdown cells compared with mice carrying control tumour cells. CONCLUSION: These data demonstrate the importance of TAK1 signalling in tumour metastasis in vivo and suggest an opportunity for antimetastatic therapies.


Asunto(s)
Adhesión Celular/efectos de los fármacos , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Invasividad Neoplásica/prevención & control , Metástasis de la Neoplasia/prevención & control , Animales , Ácido Araquidónico , Neoplasias de la Mama , Línea Celular Tumoral , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Quinasas Quinasa Quinasa PAM/biosíntesis , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/farmacología , Ratones , Trasplante de Neoplasias , ARN Interferente Pequeño/farmacología
9.
Biochem Biophys Res Commun ; 420(3): 611-5, 2012 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-22445893

RESUMEN

The growth of thyroid cells is tightly regulated by thyroid stimulating hormone (TSH) through the cyclic adenosine 3', 5'-monophosphate (cAMP) signaling pathway by potentiating the mitogenic activity of insulin and insulin-like growth factors (IGFs). However, we recently reported that thyroglobulin (Tg), a major product of the thyroid, also induces the growth of thyroid cells cultured in 0.2% serum in the absence of TSH and insulin. In this report, we demonstrate that Tg induced phosphorylation of molecules of the c-Raf/MEK/ERK pathway of the mitogen-activated protein kinase (MAPK). The MEK-1/2 inhibitor PD98059 suppressed Tg-induced phosphorylation of ERK1/2 and reduced bromodeoxyuridine (BrdU) incorporation. Tg also induced expression of the essential transcriptional factors c-Myc, c-Fos and c-Jun and phosphorylation of the retinoblastoma (Rb) protein. The present results, together with the previous report, suggest that Tg utilizes multiple signaling cascades to induce thyroid cell growth independent of TSH/cAMP stimulation.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/biosíntesis , Quinasas Quinasa Quinasa PAM/biosíntesis , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Tiroglobulina/farmacología , Glándula Tiroides/efectos de los fármacos , Animales , Línea Celular , Medio de Cultivo Libre de Suero/farmacología , Replicación del ADN/efectos de los fármacos , Activación Enzimática , Flavonoides/farmacología , Expresión Génica/efectos de los fármacos , Insulina/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Ratas , Glándula Tiroides/citología , Glándula Tiroides/enzimología , Tirotropina/farmacología
10.
Blood ; 126(24): 2649-52, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26468227

Asunto(s)
Antineoplásicos/uso terapéutico , Imidazoles/uso terapéutico , Sarcoma de Células de Langerhans/tratamiento farmacológico , Terapia Molecular Dirigida , Proteínas de Neoplasias/antagonistas & inhibidores , Oximas/uso terapéutico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Azetidinas/administración & dosificación , Ciclofosfamida/administración & dosificación , Doxorrubicina/administración & dosificación , Resistencia a Antineoplásicos , Sustitución de Medicamentos , Humanos , Indoles/administración & dosificación , Sarcoma de Células de Langerhans/diagnóstico por imagen , Sarcoma de Células de Langerhans/genética , Sarcoma de Células de Langerhans/patología , Ganglios Linfáticos/diagnóstico por imagen , Ganglios Linfáticos/patología , Quinasas Quinasa Quinasa PAM/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/biosíntesis , Quinasas Quinasa Quinasa PAM/genética , Masculino , Persona de Mediana Edad , Imagen Multimodal , Mutación Missense , Proteínas de Neoplasias/genética , Piperidinas/administración & dosificación , Mutación Puntual , Tomografía de Emisión de Positrones , Prednisona/administración & dosificación , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Recurrencia , Sulfonamidas/administración & dosificación , Tomografía Computarizada por Rayos X , Vemurafenib , Vincristina/administración & dosificación
11.
Appl Microbiol Biotechnol ; 95(2): 531-40, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22639140

RESUMEN

Furfural is one of the major inhibitors generated during sugar production from cellulosic materials and, as an aldehyde, inhibits various cellular activities of microorganisms used, leading to prolonged lag time during ethanologenic fermentation. Since Saccharomyces cerevisiae strains tolerant to furfural are of great economic benefit in producing bioethanol, much effort to obtain more efficient strains continues to be made. In this study, we examined the furfural tolerance of transposon mutant strains (Tn 1-5) with enhanced ethanol tolerance and found that one of them (Tn 2), in which SSK2 is downregulated at the transcriptional level, displayed improved furfural tolerance. Such phenotype was abolished by complementation of the entire open reading frame of SSK2, which encodes a mitogen-activated protein (MAP) kinase kinase kinase of the high osmolarity glycerol (HOG) signaling pathway, suggesting an inhibitory effect of SSK2 in coping with furfural stress. Tn 2 showed a significant decrease in the intracellular level of reactive oxygen species (ROS) and early and high activation of Hog1p, a MAP kinase integral to the HOG pathway in response to furfural. The transcriptional levels of CTT1 and GLR1, two of known Hog1p downstream target genes whose protein products are involved in reducing ROS, were increased by 43 % and 56 % respectively compared with a control strain, probably resulting in the ROS decrease. Tn 2 also showed a shortened lag time during fermentation in the presence of furfural, resulting from efficient conversion of furfural to non-toxic (or less toxic) furfuryl alcohol. Taken together, the enhanced furfural tolerance of Tn 2 is suggested to be conferred by the combined effect of an early event of less ROS accumulation and a late event of efficient detoxification of furfural.


Asunto(s)
Antifúngicos/farmacología , Elementos Transponibles de ADN , Furaldehído/farmacología , Quinasas Quinasa Quinasa PAM/biosíntesis , Mutagénesis Insercional , Proteínas de Saccharomyces cerevisiae/biosíntesis , Saccharomyces cerevisiae/efectos de los fármacos , Saccharomyces cerevisiae/genética , Biotransformación , Celulosa/metabolismo , Citosol/química , Regulación hacia Abajo , Etanol/metabolismo , Expresión Génica , Quinasas Quinasa Quinasa PAM/genética , Especies Reactivas de Oxígeno/análisis , Saccharomyces cerevisiae/crecimiento & desarrollo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Transducción de Señal , Transcripción Genética
12.
Biochem Biophys Res Commun ; 408(3): 363-8, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21333626

RESUMEN

Cyclosporine A (CsA) is an immunosuppressive drug commonly used for maintaining chronic immune suppression in organ transplant recipients. It is known that patients receiving CsA manifest increased growth of aggressive non-melanoma skin cancers. However, the underlying mechanism by which CsA augments tumor growth is not fully understood. Here, we show that CsA augments the growth of A431 epidermoid carcinoma xenograft tumors by activating tumor growth factor ß-activated kinase1 (TAK1). The activation of TAK1 by CsA occurs at multiple levels by kinases ZMP, AMPK and IRAK. TAK1 forms heterodimeric complexes with TAK binding protein 1 and 2 (TAB1/TAB2) which in term activate nuclear factor κB (NFκB) and p38 MAP kinase. Transcriptional activation of NFκB is evidenced by IKKß-mediated phosphorylation-dependent degradation of IκB and consequent nuclear translocation of p65. This also leads to enhancement in the expression of its transcriptional target genes cyclin D1, Bcl2 and COX-2. Similarly, activation of p38 leads to enhanced inflammation-related signaling shown by increased phosphorylation of MAPKAPK2 and which in turn phosphorylates its substrate HSP27. Activation of both NFκB and p38 MAP kinase provide mitogenic stimuli to augment the growth of SCCs.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Carcinógenos/farmacología , Carcinoma de Células Escamosas/enzimología , Ciclosporina/farmacología , Inmunosupresores/farmacología , Quinasas Quinasa Quinasa PAM/biosíntesis , Animales , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Femenino , Humanos , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Histopathology ; 59(3): 556-61, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22034893

RESUMEN

AIMS: Subclassification of undifferentiated pleomorphic sarcoma/malignant fibrous histiocytoma (UPS/MFH) into distinct biological cohorts based on the expression patterns of molecular markers can identify patient subsets with especially unfavourable clinical outcomes. Identification of molecular prognosticators amenable for drug targeting can facilitate rational development of UPS/MFH tailored therapies. The aim was to evaluate expression of c-Met pathway components in a large cohort of UPS/MFH samples. METHODS AND RESULTS: An immunohistochemical analysis for hepatocyte growth factor (HGF), c-Met, phospho-c-Met (pc-Met), phospho-mitogen-activated protein kinase kinase (MAPKK) also known as mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) kinase (p-MEK) and phospho-protein kinase B (p-AKT) was performed on a clinically annotated tissue microarray of 158 UPS/MFH samples. Univariable and multivariable analyses were conducted to evaluate the correlation of molecular variables with UPS/MFH disease specific survival. All evaluated markers were expressed in UPS/MFH to varying levels. Most importantly, strong HGF, pc-Met, p-MEK and p-AKT expression correlated significantly with dismal patient outcome on univariable statistical analysis. Expression of p-MEK and p-AKT remained statistically significant independent prognosticators on multivariable analysis. CONCLUSIONS: c-Met pathway components and especially p-MEK and p-AKT are potential prognostic biomarkers for UPS/MFH; their inclusion in future molecular-based staging systems should be evaluated. Furthermore, novel approaches targeting HGF, c-Met, MEK/extracellular-regulated kinase (ERK) and/or AKT should be considered for a subset of UPS/MFH patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Histiocitoma Fibroso Maligno/metabolismo , Proteínas Proto-Oncogénicas c-met/biosíntesis , Neoplasias de los Tejidos Blandos/metabolismo , Histiocitoma Fibroso Maligno/patología , Humanos , Inmunohistoquímica , Quinasas Quinasa Quinasa PAM/biosíntesis , Pronóstico , Modelos de Riesgos Proporcionales , Proteínas Proto-Oncogénicas c-akt/biosíntesis , Neoplasias de los Tejidos Blandos/patología , Análisis de Matrices Tisulares
14.
Nat Med ; 6(5): 556-63, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10802712

RESUMEN

The transforming-growth-factor-beta-activated kinase TAK1 is a member of the mitogen-activated protein kinase kinase kinase family, which couples extracellular stimuli to gene transcription. The in vivo function of TAK1 is not understood. Here, we investigated the potential involvement of TAK1 in cardiac hypertrophy. In adult mouse myocardium, TAK1 kinase activity was upregulated 7 days after aortic banding, a mechanical load that induces hypertrophy and expression of transforming growth factor beta. An activating mutation of TAK1 expressed in myocardium of transgenic mice was sufficient to produce p38 mitogen-activated protein kinase phosphorylation in vivo, cardiac hypertrophy, interstitial fibrosis, severe myocardial dysfunction, 'fetal' gene induction, apoptosis and early lethality. Thus, TAK1 activity is induced as a delayed response to mechanical stress, and can suffice to elicit myocardial hypertrophy and fulminant heart failure.


Asunto(s)
Presión Sanguínea , Gasto Cardíaco Bajo/etiología , Cardiomegalia/etiología , Quinasas Quinasa Quinasa PAM/biosíntesis , Factor de Transcripción Activador 6 , Animales , Aorta/cirugía , Proteínas de Unión al ADN/metabolismo , Diástole , Regulación hacia Abajo , Quinasas Quinasa Quinasa PAM/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Nucleares/metabolismo , Factor de Respuesta Sérica , Transducción de Señal , Sístole , Factores de Transcripción , Factor de Crecimiento Transformador beta/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos
15.
Clin Exp Pharmacol Physiol ; 38(4): 278-84, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21314842

RESUMEN

1. Cardiac troponin I-interacting kinase (TNNI3K) is a novel cardiac-specific kinase gene. Quantitative real-time reverse transcription polymerase chain reaction analysis showed a significant increase in TNNI3K mRNA expression in hypertrophic cardiomyocytes induced by endothelin-1 (ET-1). The aim of the present study was to investigate the effects of TNNI3K on neonate rat cardiomyocyte hypertrophy induced by ET-1. 2. Adenoviruses were amplified in 293A cells. To determine a reasonable adenovirus infection dose cardiomyocytes were infected with an adenovirus carrying human TNNI3K (Ad-TNNI3K) at varying multiplicity of infection (MOI) and the expression of TNNI3K was analysed by western blot. 3. Cardiomyocytes were infected with either a control adenovirus carrying green fluorescent protein (Ad-GFP) or Ad-TNNI3K. Compared with Ad-GFP, the Ad-TNNI3K induced an increase in sarcomere organization, cell surface area, (3) H-leucine incorporation and ß-MHC re-expression. This type of hypertrophic phenomenon is similar to that observed in Ad-GFP-infected hypertrophic cardiomyocytes induced by ET-1. To determine the functional role of TNNI3K in ET-1-induced hypertrophic cardiomyocytes, the cells were infected with Ad-GFP or Ad-TNNI3K. Ad-TNNI3K induced an increase in sarcomere organization, cell surface area and (3) H-leucine incorporation compared with Ad-GFP. 4. These results suggest that TNNI3K overexpression induces cardiomyocytes hypertrophy and accelerates hypertrophy in hypertrophic cardiomyocytes. Therefore, TNNI3K might be an interesting target for the clinical treatment of hypertrophy.


Asunto(s)
Cardiomegalia/enzimología , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Miocitos Cardíacos/enzimología , Adenoviridae/genética , Animales , Células Cultivadas , Endotelina-1/farmacología , Expresión Génica/efectos de los fármacos , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Humanos , Leucina/metabolismo , Quinasas Quinasa Quinasa PAM/biosíntesis , Proteínas Serina-Treonina Quinasas , Ratas , Ratas Sprague-Dawley , Sarcómeros/efectos de los fármacos , Sarcómeros/enzimología , Miosinas Ventriculares/biosíntesis , Miosinas Ventriculares/genética
16.
Am J Dermatopathol ; 33(7): 681-6, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21915030

RESUMEN

The c-Jun amino-terminal kinase (JNK) pathway seems to play important roles in the pathogenesis of several tumors, but its significance in extramammary Paget disease (EMPD) has not been investigated yet. The purpose of the study was to investigate the potential contribution of the JNK-associated molecules, such as hematopoietic progenitor kinase 1 (HPK1), mitogen-activated protein/extracellular signal-related protein kinase kinase kinase1 (MEKK1), transforming growth factor-ß activated kinase 1 (TAK1), and phosphomitogen-activated protein kinase kinase 4 (p-MKK4) to the development of EMPD. Thirty-five paraffin-embedded EMPD specimens were subjected to immunohistochemical staining for HPK1, MEKK1, TAK1, and p-MKK4. All the 35 EMPD, including 13 dermal invasive EMPD and 2 lymph node metastasis, showed cytoplasmic overexpression of HPK1, MEKK1, and p-MKK4. The expression (%positive cells) of HPK1, MEKK1, and p-MKK4 in EMPD (92.3% ± 8.6%, 92.9% ± 8.6%, and 92.7% ± 7.4%, respectively) were significantly higher than in normal eccrine sweat gland cells (51.6% ± 10.4%, 44.7% ± 11.7%, 0% ± 0%). In addition, the expression of HPK1-, MEKK1-, and p-MKK4 in invasive EMPD was significantly higher than in noninvasive EMPD. Meanwhile, the expression of TAK1 was basically low and no significantly different between EMPD and normal controls. In conclusion, these results indicate that JNK pathway may play a role in the pathogenesis of EMPD.


Asunto(s)
MAP Quinasa Quinasa 4/biosíntesis , Quinasa 1 de Quinasa de Quinasa MAP/biosíntesis , Enfermedad de Paget Extramamaria/enzimología , Enfermedad de Paget Extramamaria/patología , Proteínas Serina-Treonina Quinasas/biosíntesis , Adulto , Anciano , Biomarcadores de Tumor/análisis , Femenino , Humanos , Inmunohistoquímica , Quinasas Quinasa Quinasa PAM/biosíntesis , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico
17.
Cell Cycle ; 20(3): 320-331, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33461373

RESUMEN

Triple-negative breast cancer (TNBC) is the most aggressive histological subtype of breast cancer and is characterized by poor outcomes and a lack of specific-targeted therapies. Transforming growth factor-ß (TGF-ß) acts as the key cytokine in the epithelial-mesenchymal transition (EMT) and the metastasis of TNBC. However, the regulatory mechanisms of the TGF-ß signaling pathway remain largely unknown. In this study, we identified that the USP1/WDR48 complex could effectively enhance TGF-ß-mediated EMT and migration of TNBC cells. Furthermore, lower phosphorylation of Smad2/3, Erk, Jnk, and p38 was noted on the suppression of the expression of endogenous USP1 or WDR48. Moreover, the USP1-WDR48 complex was found to downregulate the polyubiquitination of TAK1 and mediate its in vitro stability. Therefore, our findings have shed a light on the novel role of the USP1/WDR48 complex in promoting TGF-ß-induced EMT and migration in TNBC via in vitro stabilization of TAK1.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Quinasas Quinasa Quinasa PAM/biosíntesis , Factor de Crecimiento Transformador beta/biosíntesis , Neoplasias de la Mama Triple Negativas/metabolismo , Proteasas Ubiquitina-Específicas/biosíntesis , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Quinasas Quinasa Quinasa PAM/genética , Estabilidad Proteica , Factor de Crecimiento Transformador beta/genética , Neoplasias de la Mama Triple Negativas/genética , Proteasas Ubiquitina-Específicas/genética
18.
J Am Heart Assoc ; 10(4): e014311, 2021 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-33522247

RESUMEN

Background Heart pathological hypertrophy has been recognized as a predisposing risk factor for heart failure and arrhythmia. DUSP (dual-specificity phosphatase) 26 is a member of the DUSP family of proteins, which has a significant effect on nonalcoholic fatty liver disease, neuroblastoma, glioma, and so on. However, the involvement of DUSP26 in cardiac hypertrophy remains unclear. Methods and Results Our study showed that DUSP26 expression was significantly increased in mouse hearts in response to pressure overload as well as in angiotensin II-treated cardiomyocytes. Cardiac-specific overexpression of DUSP26 mice showed attenuated cardiac hypertrophy and fibrosis, while deficiency of DUSP26 in mouse hearts resulted in increased cardiac hypertrophy and deteriorated cardiac function. Similar effects were also observed in cellular hypertrophy induced by angiotensin II. Importantly, we showed that DUSP26 bound to transforming growth factor-ß activated kinase 1 and inhibited transforming growth factor-ß activated kinase 1 phosphorylation, which led to suppression of the mitogen-activated protein kinase signaling pathway. In addition, transforming growth factor-ß activated kinase 1-specific inhibitor inhibited cardiomyocyte hypertrophy induced by angiotensin II and attenuated the exaggerated hypertrophic response in DUSP26 conditional knockout mice. Conclusions Taken together, DUSP26 was induced in cardiac hypertrophy and protected against pressure overload induced cardiac hypertrophy by modulating transforming growth factor-ß activated kinase 1-p38/ c-Jun N-terminal kinase-signaling axis. Therefore, DUSP26 may provide a therapeutic target for treatment of cardiac hypertrophy and heart failure.


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Fosfatasas de Especificidad Dual/farmacología , Regulación de la Expresión Génica , Quinasas Quinasa Quinasa PAM/genética , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/farmacología , Miocitos Cardíacos/metabolismo , Animales , Cardiomegalia/genética , Cardiomegalia/metabolismo , Modelos Animales de Enfermedad , Ecocardiografía , Quinasas Quinasa Quinasa PAM/biosíntesis , Ratones , Ratones Noqueados , Miocitos Cardíacos/patología , ARN/genética , Transducción de Señal
19.
Inflammation ; 43(4): 1548-1560, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32388658

RESUMEN

The lncRNA nuclear enriched abundant transcript 1 (NEAT1) promotes sepsis-inflammatory responses and acute kidney injury (AKI), but little known about the underlying mechanisms. This study aims to investigate the roles of NEAT1 in regulating macrophage polarization and its potential for alleviating inflammatory responses during sepsis pathogenesis. Mouse RAW264.7 macrophages were treated with lipopolysaccharide (LPS) as a cellular inflammatory model. NEAT1 shRNA, miR-125a-5p mimics, and TRAF6-overexpressing vector were used to transfect RAW264.7 cells. NEAT1, miR-125a-5p, and mRNA levels of functional genes were detected by quantitative RT-PCR. Protein abundances were analyzed by western blotting. Macrophage polarization was evaluated by flow cytometry. The bindings of miR-125a-5p with NEAT1 or TRAF6 gene were validated by dual luciferase reporter assay. LPS treatment promoted NEAT1 and suppressed miR-125a-5p expression in mouse macrophage cells. NEAT1 silencing by shRNAs promoted macrophage M2 polarization under LPS treatment, which upregulated miR-125a-5p expression, repressed TRAF6 expression and TAK1 protein phosphorylation in macrophages. These cellular and molecular changes induced by NEAT1 shRNAs were abrogated by miR-125a-5p inhibitors. Moreover, miR-125a-5p mimics suppressed TRAF6 expression and TAK1 protein phosphorylation in LPS-treated macrophages, thus causing macrophage M2 polarization under LPS treatment. TRAF6 overexpression abrogated the miR-125a-5p mimics-induced macrophage M2 polarization. miR-125a-5p could directly bind to NEAT1 or TRAF6 gene in macrophages. lncRNA NEAT1 knockdown ameliorates LPS-induced inflammation by promoting macrophage M2 polarization via miR-125a-5p/TRAF6/TAK1 axis.


Asunto(s)
Polaridad Celular/fisiología , Quinasas Quinasa Quinasa PAM/biosíntesis , Macrófagos/metabolismo , MicroARNs/biosíntesis , ARN Largo no Codificante/biosíntesis , Factor 6 Asociado a Receptor de TNF/biosíntesis , Animales , Polaridad Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Células HEK293 , Humanos , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/prevención & control , Lipopolisacáridos/toxicidad , Macrófagos/efectos de los fármacos , Ratones , Células RAW 264.7 , ARN Largo no Codificante/antagonistas & inhibidores
20.
Mol Cancer Ther ; 19(1): 247-257, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31562256

RESUMEN

YAP and TAZ are central determinants of malignancy; however, their functions remain still undruggable. We identified TGFß-activated kinase 1 (TAK1) as a central hub integrating the most relevant signals sustaining pancreatic cancer aggressiveness and chemoresistance. Glycogen synthase kinase (GSK)3 is known to stabilize TAK1, and its inhibition causes a reduction in TAK1 levels. Here, we hypothesized that TAK1 could sustain YAP/TAZ program, and thus, modulation of TAK1 expression through the inhibition of GSK3 could impair YAP/TAZ functions in pancreatic cancer.Differentially expressed transcripts between pancreatic cancer cells expressing scramble or TAK1-specific shRNA were annotated for functional interrelatedness by ingenuity pathway analysis. TAK1 expression was modulated by using different GSK3 inhibitors, including LY2090314. In vivo activity of LY2090314 alone or in combination with nab-paclitaxel was evaluated in an orthotopic nude mouse model.Differential gene expression profiling revealed significant association of TAK1 expression with HIPPO and ubiquitination pathways. We measured a significant downregulation of YAP/TAZ and their regulated genes in shTAK1 cells. TAK1 prevented YAP/TAZ proteasomal degradation in a kinase independent manner, through a complex with TRAF6, thereby fostering their K63-ubiquitination versus K48-ubiquitination. Pharmacologic modulation of TAK1 by using GSK3 inhibitors significantly decreased YAP/TAZ levels and suppressed their target genes and oncogenic functions. In vivo, LY2090314 plus nab-paclitaxel significantly prolonged mice survival duration.Our study demonstrates a unique role for TAK1 in controlling YAP/TAZ in pancreatic cancer. LY2090314 is a novel agent that warrants further clinical development in combination with nab-paclitaxel for the treatment of pancreatic cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Quinasas Quinasa Quinasa PAM/biosíntesis , Neoplasias Pancreáticas/metabolismo , Transactivadores/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Albúminas/administración & dosificación , Albúminas/farmacología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Femenino , Compuestos Heterocíclicos con 3 Anillos/administración & dosificación , Compuestos Heterocíclicos con 3 Anillos/farmacología , Xenoinjertos , Humanos , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Maleimidas/administración & dosificación , Maleimidas/farmacología , Ratones , Ratones Desnudos , Paclitaxel/administración & dosificación , Paclitaxel/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Distribución Aleatoria , Transactivadores/genética , Transactivadores/metabolismo , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Proteínas Señalizadoras YAP
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA