Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 284
Filtrar
1.
FASEB J ; 34(7): 9547-9562, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32501591

RESUMEN

Circulating plasma TRAIL levels are suppressed in patients with cardiovascular and diabetic diseases. To identify novel targets in vascular metabolic diseases, genome-wide transcriptome of aortic tissue from Trail-/- versus Trail+/+ mice were interrogated. We found 861 genes differentially expressed with TRAIL deletion. Gene enrichment analyses showed many of these genes were related to inflammation, cell-to-cell cytoskeletal interactions, and transcriptional modulation. We identified vascular protective and pathological gene clusters, with Ifi205 as the most significantly reduced vascular protective gene, whereas Glut1, the most significantly increased pathological gene with TRAIL deletion. We hypothesized that therapeutic targets could be devised from such integrated analysis and validated our findings from vascular tissues of diabetic mice. From the differentially expressed gene targets, enriched transcription factor (TF) and microRNA binding motifs were identified. The top two TFs were Elk1 and Sp1, with enrichment to eight gene targets common to both. miR-520d-3p and miR-377-3p were the top enriched microRNAs with TRAIL deletion; with four overlapping genes enriched for both microRNAs. Our findings offer an alternate in silico approach for therapeutic target identification and present a deeper understanding of gene signatures and pathways altered with TRAIL suppression in the vasculature.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Angiopatías Diabéticas/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Transcriptoma , Animales , Biología Computacional , Angiopatías Diabéticas/etiología , Angiopatías Diabéticas/patología , Humanos , Ratones , Ratones Noqueados , MicroARNs/genética
2.
Proc Natl Acad Sci U S A ; 114(22): 5755-5760, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28500273

RESUMEN

Signal transduction networks allow eukaryotic cells to make decisions based on information about intracellular state and the environment. Biochemical noise significantly diminishes the fidelity of signaling: networks examined to date seem to transmit less than 1 bit of information. It is unclear how networks that control critical cell-fate decisions (e.g., cell division and apoptosis) can function with such low levels of information transfer. Here, we use theory, experiments, and numerical analysis to demonstrate an inherent trade-off between the information transferred in individual cells and the information available to control population-level responses. Noise in receptor-mediated apoptosis reduces information transfer to approximately 1 bit at the single-cell level but allows 3-4 bits of information to be transmitted at the population level. For processes such as eukaryotic chemotaxis, in which single cells are the functional unit, we find high levels of information transmission at a single-cell level. Thus, low levels of information transfer are unlikely to represent a physical limit. Instead, we propose that signaling networks exploit noise at the single-cell level to increase population-level information transfer, allowing extracellular ligands, whose levels are also subject to noise, to incrementally regulate phenotypic changes. This is particularly critical for discrete changes in fate (e.g., life vs. death) for which the key variable is the fraction of cells engaged. Our findings provide a framework for rationalizing the high levels of noise in metazoan signaling networks and have implications for the development of drugs that target these networks in the treatment of cancer and other diseases.


Asunto(s)
Modelos Biológicos , Transducción de Señal/fisiología , Fenómenos Biofísicos , Comunicación Celular , Simulación por Computador , Células HeLa , Humanos , Teoría de la Información , Canales Iónicos/efectos de los fármacos , Canales Iónicos/fisiología , Transducción de Señal/efectos de los fármacos , Biología de Sistemas , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología
3.
Nucleic Acids Res ; 45(10): 5930-5944, 2017 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-28459998

RESUMEN

Glioblastoma (GBM) is still one of the most lethal forms of brain tumor despite of the improvements in treatments. TRAIL (TNF-related apoptosis-inducing ligand) is a promising anticancer agent that can be potentially used as an alternative or complementary therapy because of its specific antitumor activity. To define the novel pathways that regulate susceptibility to TRAIL in GBM cells, we performed a genome-wide expression profiling of microRNAs in GBM cell lines with the distinct sensitivity to TRAIL-induced apoptosis. We found that the expression pattern of miR-7 is closely correlated with sensitivity of GBM cells to TRAIL. Furthermore, our gain and loss of function experiments showed that miR-7 is a potential sensitizer for TRAIL-induced apoptosis in GBM cells. In the mechanistic study, we identified XIAP is a direct downstream gene of miR-7. Additionally, this regulatory axis could also exert in other types of tumor cells like hepatocellular carcinoma cells. More importantly, in the xenograft model, enforced expression of miR-7 in TRAIL-overexpressed mesenchymal stem cells increased apoptosis and suppressed tumor growth in an exosome dependent manner. In conclusion, we identify that miR-7 is a critical sensitizer for TRAIL-induced apoptosis, thus making it as a promising therapeutic candidate for TRAIL resistance in GBM cells.


Asunto(s)
Apoptosis/fisiología , Neoplasias Encefálicas/patología , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , MicroARNs/genética , Proteínas de Neoplasias/fisiología , ARN Neoplásico/genética , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Animales , Apoptosis/genética , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Exosomas/genética , Perfilación de la Expresión Génica , Técnicas de Transferencia de Gen , Estudio de Asociación del Genoma Completo , Glioblastoma/genética , Células HEK293 , Células Hep G2 , Humanos , Masculino , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/administración & dosificación , MicroARNs/biosíntesis , MicroARNs/uso terapéutico , ARN/administración & dosificación , ARN/uso terapéutico , ARN Neoplásico/administración & dosificación , ARN Neoplásico/biosíntesis , ARN Neoplásico/uso terapéutico , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Proteína Inhibidora de la Apoptosis Ligada a X/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Adv Exp Med Biol ; 1152: 65-73, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31456180

RESUMEN

The term "autophagy", which means "self (auto) - eating (phagy)", describes a catabolic process that is evolutionarially conserved among all eukaryotes. Although autophagy is mainly accepted as a cell survival mechanism, it also modulates the process known as "type II cell death". AKT/mTOR pathway is an upstream activator of autophagy and it is tightly regulated by the ATG (autophagy-related genes) signaling cascade. In addition, wide ranging cell signaling pathways and non-coding RNAs played essential roles in the control of autophagy. Autophagy is closely related to pathological processes such as neurodegenerative diseases and cancer as well as physiological conditions. After the Nobel Prize in Physiology or Medicine 2016 was awarded to Yoshinori Ohsumi "for his discoveries of mechanisms for autophagy", there was an explosion in the field of autophagy and molecular biologists started to pay considerable attention to the mechanistic insights related to autophagy in different diseases. Since autophagy behaved dualistically, both as a cell death and a cell survival mechanism, it opened new horizons for a deeper analysis of cell type and context dependent behavior of autophagy in different types of cancers. There are numerous studies showing that the induction of autophagy mechanism will promote survival of cancer cells. Since autophagy is mainly a mechanism to keep the cells alive, it may protect breast cancer cells against stress conditions such as starvation and hypoxia. For these reasons, autophagy was noted to be instrumental in metastasis and drug resistance. In this chapter we have emphasized on role of role of autophagy in breast cancer. Additionally we have partitioned this chapter into exciting role of microRNAs in modulation of autophagy in breast cancer. We have also comprehensively summarized how TRAIL-mediated signaling and autophagy operated in breast cancer cells.


Asunto(s)
Autofagia , Neoplasias de la Mama/patología , MicroARNs/genética , Transducción de Señal , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Progresión de la Enfermedad , Humanos
5.
Proc Natl Acad Sci U S A ; 112(28): 8650-5, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26124099

RESUMEN

Nonsmall cell lung cancer (NSCLC) is one of the leading causes of death worldwide. TNF-related apoptosis-inducing ligand (TRAIL) has been shown to induce apoptosis in malignant cells without inducing significant toxicity in normal cells. However, several carcinomas, including lung cancer, remain resistant to TRAIL. MicroRNAs (miRNAs) are small noncoding RNAs of ∼ 24 nt that block mRNA translation and/or negatively regulate its stability. They are often aberrantly expressed in cancer and have been implicated in increasing susceptibility or resistance to TRAIL-induced apoptosis by inhibiting key functional proteins. Here we show that miR-148a is down-regulated in cells with acquired TRAIL-resistance compared with TRAIL-sensitive cells. Enforced expression of miR-148a sensitized cells to TRAIL and reduced lung tumorigenesis in vitro and in vivo through the down-modulation of matrix metalloproteinase 15 (MMP15) and Rho-associated kinase 1 (ROCK1). These findings suggest that miR-148a acts as a tumor suppressor and might have therapeutic application in the treatment of NSCLC.


Asunto(s)
Apoptosis/fisiología , Carcinogénesis , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , MicroARNs/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Metilación de ADN , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo
6.
Biochim Biophys Acta ; 1863(8): 2104-14, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27208428

RESUMEN

Transforming growth factor-beta (TGF-ß) induces the epithelial to mesenchymal transition (EMT) in breast epithelial cells and plays an important role in mammary morphogenesis and breast cancer. In non-transformed breast epithelial cells TGF-ß antagonizes epidermal growth factor (EGF) action and induces growth inhibition. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been reported to participate in lumen formation during morphogenesis of human breast epithelial cells. Our previous work indicated that sensitivity of human breast epithelial cells to TRAIL can be modulated through the activation of the epidermal growth factor receptor-1 (EGFR). Here, we show that TGF-ß opposes EGF-mediated sensitization to TRAIL-induced caspase-8 activation and apoptosis in non-transformed breast epithelial cells. Death-inducing signalling complex (DISC) formation by TRAIL was significantly reduced in cells treated with TGF-ß. TGF-ß treatment activates cytoprotective autophagy and down-regulates TRAIL-R2 expression at the cell surface by promoting the intracellular accumulation of this receptor. Lastly, we demonstrate that EMT is not involved in the inhibitory effect of TGF-ß on apoptosis by TRAIL. Together, the data reveal a fine regulation by EGF and TGF-ß of sensitivity of human breast epithelial cells to TRAIL which may be relevant during morphogenesis.


Asunto(s)
Apoptosis/fisiología , Mama/citología , Factor de Crecimiento Epidérmico/fisiología , Células Epiteliales/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Factor de Crecimiento Transformador beta1/fisiología , Antígenos CD , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Autofagia/fisiología , Cadherinas/metabolismo , Células Cultivadas , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/efectos de los fármacos , Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/citología , Transición Epitelial-Mesenquimal/fisiología , Femenino , Células HeLa , Humanos , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteínas Recombinantes/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Factor de Crecimiento Transformador beta1/farmacología
7.
Cell Mol Life Sci ; 73(10): 2017-27, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26910728

RESUMEN

The TNF-related apoptosis inducing ligand TRAIL is a member of the TNF superfamily that has been firstly studied and evaluated for its anti-cancer activity, and the insights into its biology have already led to the identification of several TRAIL-based anticancer strategies with strong clinical therapeutic potentials. Nonetheless, the TRAIL system is far more complex and it can lead to a wider range of biological effects other than the ability of inducing apoptosis in cancer cells. By virtue of the different receptors and the different signalling pathways involved, TRAIL plays indeed a role in the regulation of different processes of the innate and adaptive immune system and this feature makes it an intriguing molecule under consideration in the development/progression/treatment of several immunological disorders. In this context, central nervous system represents a peculiar anatomic site where, despite its "status" of immune-privileged site, both innate and adaptive inflammatory responses occur and are involved in several pathological conditions. A number of studies have evaluated the role of TRAIL and of TRAIL-related pathways as pro-inflammatory or protective stimuli, depending on the specific pathological condition, confirming a twofold nature of this molecule. In this light, the aim of this review is to summarize the main preclinical evidences of the potential/involvement of TRAIL molecule and TRAIL pathways for the treatment of central nervous system disorders and the key suggestions coming from their assessment in preclinical models as proof of concept for future clinical studies.


Asunto(s)
Enfermedades del Sistema Nervioso Central/fisiopatología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Enfermedad de Alzheimer/fisiopatología , Animales , Biomarcadores/metabolismo , Isquemia Encefálica/fisiopatología , Supervivencia Celular , Trastornos del Conocimiento/fisiopatología , Humanos , Inflamación/fisiopatología , Esclerosis Múltiple/fisiopatología , Neoplasias/fisiopatología , Transducción de Señal , Accidente Cerebrovascular/fisiopatología
8.
J Biol Chem ; 289(31): 21544-61, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24939851

RESUMEN

Death receptor 5 (DR5) is a death domain-containing transmembrane receptor that triggers cell death upon binding to its ligand, TNF-related apoptosis-inducing ligand (TRAIL), and a combination of TRAIL and agents that increase the expression of DR5 is expected to be a novel anticancer therapy. In this report, we demonstrate that the stress response gene ATF3 is required for endoplasmic reticulum stress-mediated DR5 induction upon zerumbone (ZER) and celecoxib (CCB) in human p53-deficient colorectal cancer cells. Both agents activated PERK-eIF2α kinases and induced the expression of activating transcription factor 4 (ATF4)-CCAAT enhancer-binding protein (C/EBP) homologous protein, which were remarkably suppressed by reactive oxygen species scavengers. In the absence of ATF3, the induction of DR5 mRNA and protein was abrogated significantly, and this was associated with reduced cell death by cotreatment of TRAIL with ZER or CCB. By contrast, exogenous expression of ATF3 caused a more rapid and elevated expression of DR5, resulting in enhanced sensitivity to apoptotic cell death by TRAIL/ZER or TRAIL/CCB. A reporter assay demonstrated that at least two ATF/cAMP response element motifs as well as C/EBP homologous protein motif at the proximal region of the human DR5 gene promoter were required for ZER-induced DR5 gene transcription. Taken together, our results provide novel insights into the role of ATF3 as an essential transcription factor for p53-independent DR5 induction upon both ZER and CCB treatment, and this may be a useful biomarker for TRAIL-based anticancer therapy.


Asunto(s)
Factor de Transcripción Activador 3/fisiología , Apoptosis/fisiología , Neoplasias del Colon/patología , Retículo Endoplásmico/fisiología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Factor de Transcripción Activador 3/genética , Apoptosis/efectos de los fármacos , Secuencia de Bases , Celecoxib , Línea Celular Tumoral , Cartilla de ADN , Humanos , Regiones Promotoras Genéticas , Pirazoles/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sesquiterpenos/farmacología , Sulfonamidas/farmacología
9.
Biochem Biophys Res Commun ; 463(4): 894-9, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26074143

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), either alone or in combination with other anti-cancer agents, is a promising new strategy for the treatment of cancer. However, aberrant PI3K/Akt/mTOR survival signaling may confer TRAIL resistance by altering the balance between pro- and anti-apoptotic proteins. In the present study, we showed that the Akt/mTOR inhibitor RAD001 (everolimus) induced cell death in a dose-dependent manner and enhanced TRAIL-induced apoptosis in human leukemic Jurkat T cells, which show PI3K/Akt/mTOR pathway activation and basal expression levels of death receptor (DR) 5 (TRAIL-R2). Investigation of the effect of RAD001 treatment on the expression of TRAIL receptors (TRAIL-Rs) in Jurkat T cells showed that RAD001 significantly upregulated DR5 by up to 51.22%, but not other TRAIL-Rs such as DR4 (TRAIL-R1), decoy receptor (DcR) 1 (TRAIL-R3), and DcR2 (TRAIL-R4). Pretreatment with DR5:Fc chimera abrogated the RAD001-induced increase of TRAIL cytotoxicity, indicating that the upregulation of DR5 by RAD001 plays a role in enhancing the susceptibility of Jurkat T cells to TRAIL. Our results indicate that combination treatment with RAD001 and TRAIL may be a novel therapeutic strategy in leukemia.


Asunto(s)
Antineoplásicos/farmacología , Leucemia/patología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Sirolimus/análogos & derivados , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Regulación hacia Arriba/efectos de los fármacos , Apoptosis/fisiología , Relación Dosis-Respuesta a Droga , Everolimus , Humanos , Células Jurkat , Leucemia/fisiopatología , Sirolimus/farmacología
10.
Cancer Cell ; 12(1): 66-80, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17613437

RESUMEN

Cells expressing oncogenic c-Myc are sensitized to TNF superfamily proteins. c-Myc also is an important factor in determining whether a cell is sensitive to TRAIL-induced apoptosis, and it is well established that the mitochondrial pathway is essential for apoptosis induced by c-Myc. We investigated whether c-Myc action on the mitochondria is required for TRAIL sensitivity and found that Myc sensitized cells with defective intrinsic signaling to TRAIL. TRAIL induced expression of antiapoptotic Mcl-1 and cIAP2 through activation of NF-kappaB. Both Myc and the multikinase inhibitor sorafenib block NF-kappaB. Combining sorafenib with TRAIL in vivo showed dramatic efficacy in TRAIL-resistant tumor xenografts. We propose the combination of TRAIL with sorafenib holds promise for further development.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/fisiología , Bencenosulfonatos/farmacología , Proteínas Inhibidoras de la Apoptosis/fisiología , Proteínas de Neoplasias/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Proteínas Proto-Oncogénicas c-myc/fisiología , Piridinas/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Animales , Apoptosis/efectos de los fármacos , Proteína 3 que Contiene Repeticiones IAP de Baculovirus , Línea Celular Tumoral , Femenino , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Ratones , Ratones Desnudos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Proteínas de Neoplasias/genética , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Proteínas Proto-Oncogénicas c-bcl-2/genética , Sorafenib , Transcripción Genética/fisiología , Ubiquitina-Proteína Ligasas
11.
Mol Cell Proteomics ; 12(5): 1436-50, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23319142

RESUMEN

The extracellular ligand-induced extrinsic pathway of apoptosis is executed via caspase protease cascades that activate downstream effectors by means of site-directed proteolysis. Here we identify proteome changes upon the induction of apoptosis by the cytokine tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in a Jurkat T cell line. We detected caspase-dependent cleavage substrates by quantifying protein intensities before and after TRAIL induction in SDS gel slices. Apoptotic protein cleavage events are identified by a characteristic stable isotope labeling with amino acids in cell culture (SILAC) ratio pattern across gel slices that results from differential migration of the cleaved and uncleaved proteins. We applied a statistical test to define apoptotic substrates in the proteome. Our approach identified more than 650 of these cleaved proteins in response to TRAIL-induced apoptosis, including many previously unknown substrates and cleavage sites. Inhibitor treatment combined with triple SILAC demonstrated that the detected cleavage events were caspase dependent. Proteins located in the lumina of organelles such as mitochondria and endoplasmic reticulum were significantly underrepresented in the substrate population. Interestingly, caspase cleavage is generally observed in not only one but several members of stable complexes, but often with lower stoichiometry. For instance, all five proteins of the condensin I complex were cleaved upon TRAIL treatment. The apoptotic substrate proteome data can be accessed and visualized in the MaxQB database and might prove useful for basic and clinical research into TRAIL-induced apoptosis. The technology described here is extensible to a wide range of other proteolytic cleavage events.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Relacionadas con la Autofagia , Secuencia de Consenso , Proteínas de Unión al ADN/metabolismo , Humanos , Células Jurkat , Complejos Multiproteicos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Proteoma/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo
12.
Nihon Rinsho ; 73(8): 1302-7, 2015 Aug.
Artículo en Japonés | MEDLINE | ID: mdl-26281682

RESUMEN

Apoptosis and autophagy usually function to eliminate damaged cells and damaged proteins, respectively. Dysfunction of these events induces oncogenesis and cancer development. Therefore, small compounds that activate apoptosis and autophagy are good candidates for anti-cancer chemotherapeutics to combat cancers. This review focuses on recent advances in apoptosis/autophagy and their relationship with tumorigenesis.


Asunto(s)
Antineoplásicos , Apoptosis/genética , Autofagia/genética , Descubrimiento de Drogas , Terapia Molecular Dirigida , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Autofagia/efectos de los fármacos , Autofagia/fisiología , Humanos , Proteínas Inhibidoras de la Apoptosis/fisiología , Ratones , Neoplasias/genética , Proteínas Proto-Oncogénicas c-bcl-2/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología
13.
Am J Respir Cell Mol Biol ; 51(1): 86-93, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24484417

RESUMEN

Allergic asthma is a complex disease characterized by acute inflammation of the airways that over time leads to the development of significant structural changes termed remodeling. TNF-related apoptosis-inducing ligand (TRAIL) has an important regulatory role in acute allergic airways inflammation through up-regulation of the E3 ubiquitin ligase Midline-1 (MID-1), which limits protein phosphatase 2A (PP2A) activity and downstream dephosphorylation of proinflammatory signaling molecules. The relevance of TRAIL in the development of airways remodeling has yet to be determined. In this study, the lungs of wild-type (WT) BALB/c and Tnfsf10 knockout (TRAIL-/-) mice were chronically exposed to ovalbumin (OVA) for 12 weeks to induce hallmark features of chronic allergic airways disease, including airways hyperreactivity (AHR), subepithelial collagen deposition, goblet cell hyperplasia, and smooth muscle hypertrophy. TRAIL-/- mice were largely protected from the development of AHR and peribronchial eosinophilia and had reduced levels of mast cells in the airways. This correlated with lower levels of cytokines, including IL-4, -5, -10, and -13, and with lower levels of proinflammatory chemokines from cultured cells isolated from the draining lymph nodes. TRAIL-/- mice were also protected from the characteristic features of airways remodeling, including peribronchial fibrosis, smooth muscle hypertrophy, and mucus hypersecretion, which correlated with reduced TGF-ß1 levels in the lungs. MID-1 expression was reduced in TRAIL-/- mice and up-regulated in allergic WT mice. Raising PP2A activity using 2-amino-4-(4-heptyloyphenol)-2-methylbutan-1-ol in allergic WT mice reduced eosinophilia, TGF-ß1, and peribronchial fibrosis. This study shows that TRAIL promotes airways remodeling in an OVA-induced model of chronic allergic airways disease. Targeting TRAIL and its downstream proinflammatory signaling pathway involving PP2A may be of therapeutic benefit in reducing the hallmark features of airways remodeling observed in chronic allergic airways inflammation.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/fisiología , Hiperreactividad Bronquial/patología , Músculo Liso/patología , Eosinofilia Pulmonar/patología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Animales , Apoptosis , Western Blotting , Hiperreactividad Bronquial/inducido químicamente , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/metabolismo , Citocinas/genética , Citocinas/metabolismo , Técnicas para Inmunoenzimas , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Moco/efectos de los fármacos , Moco/metabolismo , Músculo Liso/efectos de los fármacos , Músculo Liso/inmunología , Músculo Liso/metabolismo , Ovalbúmina/farmacología , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Proteínas/genética , Proteínas/metabolismo , Eosinofilia Pulmonar/inducido químicamente , Eosinofilia Pulmonar/inmunología , Eosinofilia Pulmonar/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Ubiquitina-Proteína Ligasas
14.
Cell Physiol Biochem ; 34(2): 506-18, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25116350

RESUMEN

BACKGROUND/AIM: P21, a multifunctional cell cycle-regulatory molecule, regulates apoptotic cell death. In this study we examined the effect of altered p21 expression on the sensitivity of acute myeloid leukemia cells in response to HDAC inhibitor SAHA treatment and investigated the underlying mechanism. METHODS: Stably transfected HL60 cell lines were established in RPMI-1640 with supplementation of G-418. Cell viability was measured by MTT assay. Western blot was applied to assess the protein expression levels of target genes. Cell apoptosis was monitored by AnnexinV-PE/7AAD assay. RESULTS: We showed HL60 cells that that didn't up-regulate p21 expression were more sensitive to SAHA-mediated apoptosis than NB4 and U937 cells that had increased p21 level. Enforced expression of p21 in HL60 cells reduced sensitivity to SAHA and blocked TRAIL-mediated apoptosis. Conversely, p21 silencing in NB4 cells enhanced SAHA-mediated apoptosis and lethality. Finally, we found that combined treatment with SAHA and rapamycin down-regulated p21 and enhanced apoptosis in AML cells. CONCLUSION: We conclude that up-regulated p21 expression mediates resistance to SAHA via inhibition of TRAIL apoptotic pathway. P21 may serve as a candidate biomarker to predict responsiveness or resistance to SAHA-based therapy in AML patients. In addition, rapamycin may be an effective agent to override p21-mediated resistance to SAHA in AML patients.


Asunto(s)
Apoptosis/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Leucemia Mieloide Aguda/patología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Regulación hacia Arriba , Secuencia de Bases , Western Blotting , Caspasa 8/metabolismo , Regulación hacia Abajo , Resistencia a Antineoplásicos , Células HL-60 , Humanos , Leucemia Mieloide Aguda/metabolismo , Interferencia de ARN , Sirolimus/farmacología
15.
Hepatology ; 58(4): 1362-74, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23703673

RESUMEN

UNLABELLED: Cholangiocarcinoma (CCA) cells paradoxically express the death ligand tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and thus rely on potent survival signals to circumvent cell death by TRAIL. Hedgehog (Hh) signaling is an important survival pathway in CCA. Herein, we further examine the mechanisms whereby Hh signaling mediates apoptosis resistance in CCA, revealing a pivotal role for the cell division regulating serine/threonine kinase polo-like kinase 2 (PLK2). We employed 50 human CCA samples (25 intrahepatic and 25 extrahepatic CCA) as well as human KMCH-1, Mz-CHA-1, and HUCCT-1 CCA cells for these studies. In vivo experiments were conducted using a syngeneic rat orthotopic CCA model. In human samples, polo-like kinase (PLK)1/2/3-immunoreactive cancer cells were present in the preponderance of intra- and extrahepatic CCA specimens. Inhibition of Hh signaling by cyclopamine reduced PLK2, but not PLK1 or PLK3, messenger RNA and protein expression in vehicle-treated and sonic Hh-treated CCA cells, confirming our previous microarray study. PLK2 regulation by Hh signaling appears to be direct, because the Hh transcription factors, glioma-associated oncogene 1 and 2, bind to the PLK2 promotor. Moreover, inhibition of PLK2 by the PLK inhibitor, BI 6727 (volasertib), or PLK2 knockdown was proapoptotic in CCA cells. BI 6727 administration or PLK2 knockdown decreased cellular protein levels of antiapoptotic myeloid cell leukemia 1 (Mcl-1), an effect reversed by the proteasome inhibitor, MG-132. Finally, BI 6727 administration reduced Mcl-1 protein expression in CCA cells, resulting in CCA cell apoptosis and tumor suppression in vivo. CONCLUSION: PLK2 appears to be an important mediator of Hh survival signaling. These results suggest PLK inhibitors to be of therapeutic value for treatment of human CCA.


Asunto(s)
Neoplasias de los Conductos Biliares/fisiopatología , Conductos Biliares Intrahepáticos , Colangiocarcinoma/fisiopatología , Proteínas Hedgehog/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Transducción de Señal/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Neoplasias de los Conductos Biliares/patología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Colangiocarcinoma/patología , Modelos Animales de Enfermedad , Inhibidores Enzimáticos/farmacología , Xenoinjertos , Humanos , Masculino , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Pteridinas/farmacología , Ratas , Ratas Endogámicas F344 , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología
16.
Mol Cell Biochem ; 392(1-2): 125-34, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24788726

RESUMEN

Renal cell carcinoma (RCC) is the most common types among kidney cancers. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) strongly induces apoptosis in RCC. However, TRAIL therapy also leads to hepatotoxicity. To improve the biosafety, we inserted miRNA response elements (MREs) of miR-138, miR-199, and miR-122 into an adenoviral vector, Ad-TRAIL-3MREs, to restrict TRAIL expression within RCC cells. Luciferase assays showed that MREs can regulate the expression of exogenous gene in RCC cells. Ad-TRAIL-3MREs selectively expressed TRAIL and induce apoptosis in RCC cells, but not in normal cells. MTT assays revealed that Ad-TRAIL-3MREs reduced viability of RCC cells without cytotoxicity to normal cells. Ad-TRAIL-3MREs suppressed the growth of ACHN tumors and exerted no hepatotoxicity in vivo. Collectively, we generated a TRAIL-expressing adenoviral vector under the regulation of MREs. This miRNA-based gene therapy may be a promising strategy for RCC treatment.


Asunto(s)
Carcinoma de Células Renales/patología , Neoplasias Renales/patología , MicroARNs/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Adenoviridae/genética , Secuencia de Bases , Carcinoma de Células Renales/metabolismo , Cartilla de ADN , Vectores Genéticos , Humanos , Neoplasias Renales/metabolismo , MicroARNs/genética , Reacción en Cadena de la Polimerasa , Ligando Inductor de Apoptosis Relacionado con TNF/genética
17.
Crit Rev Immunol ; 33(1): 23-40, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23510024

RESUMEN

Sepsis is the leading cause of death in most intensive care units, and the death of septic patients usually does not result from the initial septic event but rather from subsequent nosocomial infections. Patients who survive severe sepsis often display severely compromised immune function. Not only is there significant apoptosis of lymphoid and myeloid cells that depletes critical components of the immune system during sepsis, there is also decreased function of the remaining immune cells. Studies of animals and humans suggest the immune defects that occur during sepsis may be critical to pathogenesis and subsequent mortality. This review focuses on sepsis-induced alterations with the cluster differentiation (CD) 8 T-cell compartment that can affect the control of secondary heterologous infections. Understanding how a septic event directly influences CD8 T-cell populations through apoptotic death and homeostatic proliferation and indirectly by immune-mediated suppression will provide valuable starting points for developing new treatment options.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Tolerancia Inmunológica , Sepsis/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Animales , Apoptosis , Homeostasis , Humanos , Sepsis/mortalidad
18.
Nat Med ; 13(11): 1308-15, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17934471

RESUMEN

The role of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in immune responses mediated by T-helper 2 (T(H)2) lymphocytes is unknown. Here we characterize the development of allergic airway disease in TRAIL-deficient (Tnfsf10(-/-)) mice and in mice exposed to short interfering RNA targeting TRAIL. We show that TRAIL is abundantly expressed in the airway epithelium of allergic mice and that inhibition of signaling impairs production of the chemokine CCL20 and homing of myeloid dendritic cells and T cells expressing CCR6 and CD4 to the airways. Attenuated homing limits T(H)2 cytokine release, inflammation, airway hyperreactivity and expression of the transcriptional activator STAT6. Activation of STAT6 by interleukin-13 restores airway hyperreactivity in Tnfsf10(-/-) mice. Recombinant TRAIL induces pathognomic features of asthma and stimulates the production of CCL20 in primary human bronchial epithelium cells. TRAIL is also increased in sputum of asthmatics. The function of TRAIL in the airway epithelium identifies this molecule as a target for the treatment of asthma.


Asunto(s)
Quimiocina CCL20/fisiología , Activación de Linfocitos/inmunología , Hipersensibilidad Respiratoria/inmunología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Células Th2/inmunología , Animales , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/metabolismo , Quimiocina CCL20/antagonistas & inhibidores , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Interferencia de ARN , Hipersensibilidad Respiratoria/metabolismo , Hipersensibilidad Respiratoria/patología , Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Ligando Inductor de Apoptosis Relacionado con TNF/deficiencia , Ligando Inductor de Apoptosis Relacionado con TNF/genética
19.
Nat Med ; 13(9): 1070-7, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17767167

RESUMEN

Apo2L/TRAIL stimulates cancer cell death through the proapoptotic receptors DR4 and DR5, but the determinants of tumor susceptibility to this ligand are not fully defined. mRNA expression of the peptidyl O-glycosyltransferase GALNT14 correlated with Apo2L/TRAIL sensitivity in pancreatic carcinoma, non-small-cell lung carcinoma and melanoma cell lines, and up to 30% of samples from various human malignancies showed GALNT14 overexpression. RNA interference of GALNT14 reduced cellular Apo2L/TRAIL sensitivity, whereas overexpression increased responsiveness. Biochemical analysis of DR5 identified several ectodomain O-(N-acetyl galactosamine-galactose-sialic acid) structures. Sequence comparison predicted conserved extracellular DR4 and DR5 O-glycosylation sites; progressive mutation of the DR5 sites attenuated apoptotic signaling. O-glycosylation promoted ligand-stimulated clustering of DR4 and DR5, which mediated recruitment and activation of the apoptosis-initiating protease caspase-8. These results uncover a new link between death-receptor O-glycosylation and apoptotic signaling, providing potential predictive biomarkers for Apo2L/TRAIL-based cancer therapy.


Asunto(s)
Receptores de Muerte Celular/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Secuencia de Aminoácidos , Animales , Apoptosis , Carcinoma de Pulmón de Células no Pequeñas , Línea Celular Tumoral , Supervivencia Celular , Predisposición Genética a la Enfermedad , Glicosilación , Humanos , Neoplasias Pulmonares , Melanoma , Ratones , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Pancreáticas , ARN Mensajero/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/genética , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Trasplante Heterólogo
20.
Acta Biochim Biophys Sin (Shanghai) ; 46(6): 471-6, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24739635

RESUMEN

The apoptotic ligand TNF-related apoptosis-inducing ligand (TRAIL) is believed to be a promising candidate for cancer gene therapy, yet gene therapy strategies to tackle this disease systemically are often impaired by inefficient delivery of the vector to the tumor tissue. Mesenchymal stem cells (MSCs) have been shown to home to tumor sites and could potentially act as a shield and vehicle for an antitumor gene therapy vector. Here, we used an adenoviral vector expressing TRAIL to transduce MSCs and studied the apoptosis-inducing activity of these TRAIL-carrying MSCs on esophageal cancer cell Eca-109. Our results showed that, in vitro, TRAIL-expressing MSCs were able to inhibit proliferation and induce apoptosis in Eca-109 cells by an MTT assay, co-culture experiments and flow cytometry analysis. In vivo, TRAIL-expressing MSCs also displayed an ability to inhibit tumor growth in an Eca-109 xenograft mouse model. Together, our findings indicated that the gene therapy strategy of MSCs-based TRAIL gene delivery has a wide potential value for improving the treatment of esophageal cancer.


Asunto(s)
Adenoviridae/genética , Neoplasias Esofágicas/patología , Células Madre Mesenquimatosas/fisiología , Ligando Inductor de Apoptosis Relacionado con TNF/fisiología , Animales , Western Blotting , Línea Celular Tumoral , Técnicas de Cocultivo , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos BALB C
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA