Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Vis Exp ; (172)2021 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-34180908

RESUMEN

Patient-derived tumor organoids (PDOs) are expected to be a preclinical cancer model with better reproducibility of disease than traditional cell culture models. PDOs have been successfully generated from a variety of human tumors to recapitulate the architecture and function of tumor tissue accurately and efficiently. However, PDOs are unsuitable for an in vitro high-throughput assay system (HTS) or cell analysis using 96-well or 384-well plates when evaluating anticancer drugs because they are heterogeneous in size and form large clusters in culture. These cultures and assays use extracellular matrices, such as Matrigel, to create tumor tissue scaffolds. Therefore, PDOs have a low throughput and high cost, and it has been difficult to develop a suitable assay system. To address this issue, a simpler and more accurate HTS was established using PDOs to evaluate the potency of anticancer drugs and immunotherapy. An in vitro HTS was created that uses PDOs established from solid tumors cultured in 384-well plates. An HTS was also developed for assessment of antibody-dependent cellular cytotoxicity activity to represent the immune response using PDOs cultured in 96-well plates.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Organoides , Reproducibilidad de los Resultados
2.
Oncol Lett ; 21(5): 406, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33841567

RESUMEN

An in vitro assay system using patient-derived tumor models represents a promising preclinical cancer model that replicates the disease better than traditional cell culture models. Patient-derived tumor organoid (PDO) and patient-derived tumor xenograft (PDX) models have been previously established from different types of human tumors to recapitulate accurately and efficiently their tissue architecture and function. However, these models have low throughput and are challenging to construct. Thus, the present study aimed to establish a simple in vitro high-throughput assay system using PDO and PDX models. Furthermore, the current study aimed to evaluate different classes of anticancer drugs, including chemotherapeutic, molecular targeted and antibody drugs, using PDO and PDX models. First, an in vitro high-throughput assay system was constructed using PDO and PDX established from solid and hematopoietic tumors cultured in 384-well plates to evaluate anticancer agents. In addition, an in vitro evaluation system of the immune response was developed using PDO and PDX. Novel cancer immunotherapeutic agents with marked efficacy have been used against various types of tumor. Thus, there is an urgent need for in vitro functional potency assays that can simulate the complex interaction of immune cells with tumor cells and can rapidly test the efficacy of different immunotherapies or antibody drugs. An evaluation system for the antibody-dependent cellular cytotoxic activity of anti-epidermal growth factor receptor antibody and the cytotoxic activity of activated lymphocytes, such as cytotoxic T lymphocytes and natural killer cells, was constructed. Moreover, immune response assay systems with bispecific T-cell engagers were developed using effector cells. The present results demonstrated that in vitro assay systems using PDO and PDX may be suitable for evaluating anticancer agents and immunotherapy potency with high reproducibility and simplicity.

3.
Cells ; 8(5)2019 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-31137590

RESUMEN

Patient-derived tumor organoids (PDOs) represent a promising preclinical cancer model that better replicates disease, compared with traditional cell culture models. We have established PDOs from various human tumors to accurately and efficiently recapitulate the tissue architecture and function. Molecular targeted therapies with remarkable efficacy are currently in use against various tumors. Thus, there is a need for in vitro functional-potency assays that can be used to test the efficacy of molecular targeted drugs and model complex interactions between immune cells and tumor cells to evaluate the potential for cancer immunotherapy. This study represents an in vitro evaluation of different classes of molecular targeted drugs, including small-molecule inhibitors, monoclonal antibodies, and an antibody-drug conjugate, using lung PDOs. We evaluated epidermal growth factor receptor and human epidermal growth factor receptor 2 (HER2) inhibitors using a suitable high-throughput assay system. Next, the antibody-dependent cellular cytotoxicity (ADCC) activity of an anti-HER2 monoclonal antibody was evaluated to visualize the interactions of immune cells with PDOs during ADCC responses. Moreover, an evaluation system was developed for the immune checkpoint inhibitors, nivolumab and pembrolizumab, using PDOs. Our results demonstrate that the in vitro assay systems using PDOs were suitable for evaluating molecular targeted drugs under conditions that better reflect pathological conditions.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/uso terapéutico , Carcinoma Adenoescamoso/tratamiento farmacológico , Carcinoma de Células Escamosas/tratamiento farmacológico , Evaluación de Medicamentos/métodos , Neoplasias Pulmonares/tratamiento farmacológico , Terapia Molecular Dirigida , Organoides/efectos de los fármacos , Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Biopsia , Carcinoma Adenoescamoso/patología , Carcinoma Adenoescamoso/cirugía , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/cirugía , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Receptores ErbB/antagonistas & inhibidores , Humanos , L-Lactato Deshidrogenasa/análisis , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/cirugía , Receptor ErbB-2/antagonistas & inhibidores
4.
FEBS J ; 285(22): 4146-4164, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30281916

RESUMEN

Activating transcription factor 6 alpha (referred to as ATF6 hereafter) is an endoplasmic reticulum (ER)-resident glycoprotein and one of the three sensors of the unfolded protein response (UPR). Upon ER stress, ATF6 is exported to the Golgi complex where it is cleaved by the S1P and S2P proteases thus releasing ATF6 cytosolic fragment and leading to the transcription of ATF6 target genes. In this study, we performed a phenotypic small-interfering RNA (siRNA) screening to better characterize the ER mechanisms involved in ATF6 activation upon ER stress. This revealed that silencing of ER-degradation-enhancing alpha-mannosidase-like protein-1 (EDEM1) increased the bioavailability of ER stress-induced ATF6 export to the Golgi complex through the stabilization of the natively unstable ATF6 protein. Moreover, we characterized a somatic variant of EDEM1 (N198I) found in hepatocellular carcinoma that alters ATF6 signaling and might provide a selective advantage to the transforming cells. Hence, our work confirms the natively unstable nature of ATF6 and links this property to potentially associated pro-oncogenic functions.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Carcinoma Hepatocelular/patología , Estrés del Retículo Endoplásmico , Neoplasias Hepáticas/patología , Proteínas de la Membrana/metabolismo , Mutación , Respuesta de Proteína Desplegada , Factor de Transcripción Activador 6/genética , Apoptosis , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Proliferación Celular , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Proteínas de la Membrana/genética , Transducción de Señal , Células Tumorales Cultivadas
5.
Oncol Rep ; 40(2): 635-646, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29917168

RESUMEN

Patient-derived tumor xenograft models represent a promising preclinical cancer model that better replicates disease, compared with traditional cell culture; however, their use is low-throughput and costly. To overcome this limitation, patient-derived tumor organoids (PDOs) were established from human lung, ovarian and uterine tumor tissues, among others, to accurately and efficiently recapitulate the tissue architecture and function. PDOs were able to be cultured for >6 months, and formed cell clusters with similar morphologies to their source tumors. Comparative histological and comprehensive gene expression analyses proved that the characteristics of PDOs were similar to those of their source tumors, even following long-term expansion in culture. At present, 53 PDOs have been established by the Fukushima Translational Research Project, and were designated as Fukushima PDOs (F­PDOs). In addition, the in vivo tumorigenesis of certain F­PDOs was confirmed using a xenograft model. The present study represents a detailed analysis of three F­PDOs (termed REME9, 11 and 16) established from endometrial cancer tissues. These were used for cell growth inhibition experiments using anticancer agents. A suitable high-throughput assay system, with 96- or 384­well plates, was designed for each F­PDO, and the efficacy of the anticancer agents was subsequently evaluated. REME9 and 11 exhibited distinct responses and increased resistance to the drugs, as compared with conventional cancer cell lines (AN3 CA and RL95-2). REME9 and 11, which were established from tumors that originated in patients who did not respond to paclitaxel and carboplatin (the standard chemotherapy for endometrial cancer), exhibited high resistance (half-maximal inhibitory concentration >10 µM) to the two agents. Therefore, assay systems using F­PDOs may be utilized to evaluate anticancer agents using conditions that better reflect clinical conditions, compared with conventional methods using cancer cell lines, and to discover markers that identify the pharmacological effects of anticancer agents.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Endometriales/tratamiento farmacológico , Organoides/efectos de los fármacos , Animales , Carboplatino/farmacología , Carcinogénesis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Paclitaxel/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
J Toxicol Sci ; 42(6): 755-761, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29142174

RESUMEN

In recent years, human-induced pluripotent stem cell-derived cardiomyocytes (hiPS-CMs) have been widely used to develop evaluation systems for drug cardiotoxicity, including the arrhythmia caused by QT prolongation. To accurately assess the arrhythmogenic potential of drugs, associated with QT prolongation, we developed an evaluation system using hiPS-CMs and gene expression analysis. hiPS-CMs were treated with 8 arrhythmogenic and 17 non-arrhythmogenic drugs at several concentrations for 24 hr to comprehensively analyze gene expression. The results showed that 19 genes were upregulated in the arrhythmogenic drug-treated cells compared with their expression levels in the non-treated and non-arrhythmogenic drug-treated cells. The arrhythmogenic risks of the drugs were evaluated by scoring gene expression levels. The results indicated that arrhythmogenic risks could be inferred when cells were treated at a concentration 100 times higher than the maximum blood concentration of the drug. Thus, we succeeded in developing a system for evaluation of the arrhythmogenic potential of drugs using gene expression analysis.


Asunto(s)
Amlodipino/toxicidad , Arritmias Cardíacas/inducido químicamente , Bencimidazoles/toxicidad , Bisoprolol/toxicidad , Evaluación Preclínica de Medicamentos/métodos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Células Madre Pluripotentes Inducidas , Síndrome de QT Prolongado/inducido químicamente , Miocitos Cardíacos , Fenilpropionatos/toxicidad , Piridazinas/toxicidad , Tetrazoles/toxicidad , Transcriptoma/efectos de los fármacos , Compuestos de Bifenilo , Cardiotoxicidad , Células Cultivadas , Relación Dosis-Respuesta a Droga , Humanos , Linagliptina/toxicidad , Naftalenos/toxicidad , Piperazinas/toxicidad , Clorhidrato de Prasugrel/toxicidad , Sumatriptán/toxicidad , Regulación hacia Arriba/efectos de los fármacos
7.
Oncol Rep ; 37(1): 66-76, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27840973

RESUMEN

Epidermal growth factor receptor (EGFR) overexpression and EGFR-mediated signaling pathway dysregulation have been observed in tumors from patients with various cancers, especially non-small cell lung cancer. Thus, several anti-EGFR drugs have been developed for cancer therapy. For patients with known EGFR activating mutations (EGFR exon 19 in-frame deletions and exon 21 L858R substitution), treatment with an EGFR tyrosine kinase inhibitor (EGFR TKI; gefitinib, erlotinib or afatinib) represents standard first-line therapy. However, the clinical efficacy of these TKIs is ultimately limited by the development of acquired drug resistance such as by mutation of the gatekeeper T790 residue (T790M). To overcome this acquired drug resistance and develop novel anti-EGFR drugs, a cell-based assay system for EGFR TKI resistance mutant-selective inhibitors is required. We constructed a novel cell-based assay for the evaluation of EGFR TKI efficacy against EGFR mutation. To this end, we established non-tumorigenic immortalized breast epithelial cells that proliferate dependent on EGF (MCF 10A cells), which stably overexpress mutant EGFR. We found that the cells expressing EGFR containing the T790M mutation showed higher resistance against gefitinib, erlotinib and afatinib compared with cells expressing wild-type EGFR. In contrast, L858R mutant-expressing cells exhibited higher TKI sensitivity. The effect of T790M-selective inhibitors (osimertinib and rociletinib) on T790M mutant-expressing cells was significantly higher than gefitinib and erlotinib. Finally, when compared with commercially available isogenic MCF 10A cell lines carrying introduced mutations in EGFR, our EGFR mutant-overexpressing cells exhibited obviously higher responsiveness to EGFR TKIs depending on the underlying mutations because of the higher levels of EGFR phosphorylation in the EGFR mutant-overexpressing cells than in the isogenic cell lines. In conclusion, we successfully developed a novel cell-based assay for evaluating the efficacy of anti-EGFR drugs against EGFR mutation.


Asunto(s)
Antineoplásicos/aislamiento & purificación , Evaluación Preclínica de Medicamentos/métodos , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Inhibidores de Proteínas Quinasas/aislamiento & purificación , Afatinib , Antineoplásicos/uso terapéutico , Técnicas de Cultivo de Célula , Células Cultivadas , Resistencia a Antineoplásicos/efectos de los fármacos , Clorhidrato de Erlotinib/farmacología , Gefitinib , Humanos , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinazolinas/farmacología , Transfección
8.
PLoS One ; 9(11): e112714, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25383781

RESUMEN

BACKGROUND: Type 2 diabetes is characterized by pancreatic beta-cell dysfunction and is associated with low-grade inflammation. Recent observations suggest that apoptosis signal-regulating kinase 1 (ASK1) is involved in beta-cell death in response to different stressors. In this study, we tested whether ASK1 deficiency protects beta-cells from glucolipotoxic conditions and cytokines treatment or from glucose homeostasis alteration induced by endotoxemia. METHODOLOGY/PRINCIPAL FINDINGS: Insulin secretion was neither affected upon shRNA-mediated downregulation of ASK1 in MIN6 cells nor in islets from ASK1-deficient mice. ASK1 silencing in MIN6 cells and deletion in islets did not prevent the deleterious effect of glucolipotoxic conditions or cytokines on insulin secretion. However, it protected MIN6 cells from death induced by ER stress or palmitate and islets from short term caspase activation in response to cytokines. Moreover, endotoxemia induced by LPS infusion increased insulin secretion during hyperglycemic clamps but the response was similar in wild-type and ASK1-deficient mice. Finally, insulin sensitivity in the presence of LPS was not affected by ASK1-deficiency. CONCLUSIONS/SIGNIFICANCE: Our study demonstrates that ASK1 is not involved in beta-cell function and dysfunction but controls stress-induced beta-cell death.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Glucosa/metabolismo , Inflamación/patología , Células Secretoras de Insulina/metabolismo , MAP Quinasa Quinasa Quinasa 5/genética , MAP Quinasa Quinasa Quinasa 5/metabolismo , Animales , Células Cultivadas , Citocinas/farmacología , Diabetes Mellitus Tipo 2/metabolismo , Glucosa/farmacología , Humanos , Inflamación/metabolismo , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/patología , Lipopolisacáridos/farmacología , Ratones , Ratones Noqueados , Ácido Palmítico/farmacología , Estrés Fisiológico
9.
Mol Cell Biol ; 34(10): 1839-49, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24636989

RESUMEN

ATF6α, a membrane-anchored transcription factor from the endoplasmic reticulum (ER) that modulates the cellular response to stress as an effector of the unfolded-protein response (UPR), is a key player in the development of tumors of different origin. ATF6α activation has been linked to oncogenic transformation and tumor maintenance; however, the mechanism(s) underlying this phenomenon remains elusive. Here, using a phenotypic small interfering RNA (siRNA) screening, we identified a novel role for ATF6α in chemoresistance and defined the protein disulfide isomerase A5 (PDIA5) as necessary for ATF6α activation upon ER stress. PDIA5 contributed to disulfide bond rearrangement in ATF6α under stress conditions, thereby leading to ATF6α export from the ER and activation of its target genes. Further analysis of the mechanism demonstrated that PDIA5 promotes ATF6α packaging into coat protein complex II (COPII) vesicles and that the PDIA5/ATF6α activation loop is essential to confer chemoresistance on cancer cells. Genetic and pharmacological inhibition of the PDIA5/ATF6α axis restored sensitivity to the drug treatment. This work defines the mechanisms underlying the role of ATF6α activation in carcinogenesis and chemoresistance; furthermore, it identifies PDIA5 as a key regulator ATF6α-mediated cellular functions in cancer.


Asunto(s)
Factor de Transcripción Activador 6/metabolismo , Antineoplásicos/farmacología , Benzamidas/farmacología , Piperazinas/farmacología , Proteína Disulfuro Isomerasas/metabolismo , Pirimidinas/farmacología , Vesículas Cubiertas por Proteínas de Revestimiento/metabolismo , Supervivencia Celular/efectos de los fármacos , Cistina/metabolismo , Resistencia a Antineoplásicos , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico , Células HeLa , Proteínas de Choque Térmico/metabolismo , Humanos , Mesilato de Imatinib , Transporte de Proteínas , Respuesta de Proteína Desplegada
10.
Cancer Res ; 73(15): 4732-43, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23752693

RESUMEN

Growing evidence supports a role for the unfolded protein response (UPR) in carcinogenesis; however, the precise molecular mechanisms underlying this phenomenon remain elusive. Herein, we identified the circadian clock PER1 mRNA as a novel substrate of the endoribonuclease activity of the UPR sensor IRE1α. Analysis of the mechanism shows that IRE1α endoribonuclease activity decreased PER1 mRNA in tumor cells without affecting PER1 gene transcription. Inhibition of IRE1α signaling using either siRNA-mediated silencing or a dominant-negative strategy prevented PER1 mRNA decay, reduced tumorigenesis, and increased survival, features that were reversed upon PER1 silencing. Clinically, patients showing reduced survival have lower levels of PER1 mRNA expression and increased splicing of XBP1, a known IRE-α substrate, thereby pointing toward an increased IRE1α activity in these patients. Hence, we describe a novel mechanism connecting the UPR and circadian clock components in tumor cells, thereby highlighting the importance of this interplay in tumor development.


Asunto(s)
Endorribonucleasas/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Glioblastoma/metabolismo , Proteínas Circadianas Period/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Respuesta de Proteína Desplegada/fisiología , Animales , Secuencia de Bases , Endorribonucleasas/genética , Glioblastoma/genética , Humanos , Ratones , Datos de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Circadianas Period/genética , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Procesamiento Postranscripcional del ARN , ARN Mensajero , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Biol Chem ; 288(24): 17190-201, 2013 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-23629659

RESUMEN

In BCR-ABL-expressing cells, sphingolipid metabolism is altered. Because the first step of sphingolipid biosynthesis occurs in the endoplasmic reticulum (ER), our objective was to identify ABL targets in the ER. A phosphoproteomic analysis of canine pancreatic ER microsomes identified 49 high scoring phosphotyrosine-containing peptides. These were then categorized in silico and validated in vitro. We demonstrated that the ER-resident human protein serine palmitoyltransferase long chain-1 (SPTLC1), which is the first enzyme of sphingolipid biosynthesis, is phosphorylated at Tyr(164) by the tyrosine kinase ABL. Inhibition of BCR-ABL using either imatinib or shRNA-mediated silencing led to the activation of SPTLC1 and to increased apoptosis in both K562 and LAMA-84 cells. Finally, we demonstrated that mutation of Tyr(164) to Phe in SPTLC1 increased serine palmitoyltransferase activity. The Y164F mutation also promoted the remodeling of cellular sphingolipid content, thereby sensitizing K562 cells to apoptosis. Our observations provide a mechanistic explanation for imatinib-mediated cell death and a novel avenue for therapeutic strategies.


Asunto(s)
Supervivencia Celular , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Procesamiento Proteico-Postraduccional , Serina C-Palmitoiltransferasa/metabolismo , Sustitución de Aminoácidos , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Benzamidas/farmacología , Núcleo Celular/enzimología , Ceramidas/metabolismo , Perros , Resistencia a Antineoplásicos , Retículo Endoplásmico/metabolismo , Proteínas de Fusión bcr-abl/química , Proteínas de Fusión bcr-abl/metabolismo , Técnicas de Silenciamiento del Gen , Aparato de Golgi/enzimología , Humanos , Mesilato de Imatinib , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Microsomas/metabolismo , Fragmentos de Péptidos/química , Fosforilación , Fosfotirosina/metabolismo , Piperazinas/farmacología , Transporte de Proteínas , Proteoma/metabolismo , Pirimidinas/farmacología , ARN Interferente Pequeño/genética , Serina C-Palmitoiltransferasa/química , Serina C-Palmitoiltransferasa/genética , Transcripción Genética/efectos de los fármacos
12.
Mol Cancer Ther ; 11(12): 2610-20, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23041544

RESUMEN

The molecular mechanisms and cellular targets of sorafenib, a multikinase inhibitor used for the treatment of hepatocellular carcinoma (HCC), remain to be fully characterized. Recent studies have shown that sorafenib induces tumor cell death through the activation of endoplasmic reticulum stress signaling and/or autophagy in various cellular models. Using liver cancer-derived cell lines, we specifically show that the IRE1 and phosphorylated extracellular signal-regulated kinase arms of the unfolded protein response (UPR) become activated upon sorafenib treatment, whereas the ATF6 arm is inhibited. Our results also reveal that sorafenib treatment causes disruption to the secretory pathway, as witnessed by the fragmentation of the Golgi apparatus and the induction of autophagy. On the basis of these observations, we tested the relevance of the AAA⁺ ATPase p97/VCP as a potential functional target of sorafenib. Our results show that p97/VCP tyrosine phosphorylation is prevented upon sorafenib treatment, and that this can be correlated with enhanced membrane association. Moreover, we show that DBeQ, a recently discovered inhibitor of p97/VCP, enhances sorafenib-mediated toxicity in cultured cells. Our data show a novel mechanism for sorafenib-mediated cell death in HCC, which depends on the integrity of the secretory pathway; and we identify p97/VCP phosphorylation as a potential target for improved sorafenib treatment efficacy in patients.


Asunto(s)
Adenosina Trifosfatasas/antagonistas & inhibidores , Carcinoma Hepatocelular/tratamiento farmacológico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Neoplasias Hepáticas/tratamiento farmacológico , Niacinamida/análogos & derivados , Compuestos de Fenilurea/farmacología , Adenosina Trifosfatasas/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Células Hep G2 , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Terapia Molecular Dirigida , Niacinamida/farmacología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Quinazolinas/farmacología , Vías Secretoras/efectos de los fármacos , Sorafenib
13.
Cell Signal ; 24(8): 1548-55, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22481091

RESUMEN

The endoplasmic reticulum (ER) is the first compartment of secretory pathway. It plays a major role in ER chaperone-assisted folding and quality control, including post-translational modification such as disulfide bond formation of newly synthesized secretory proteins. Protein folding and assembly takes place in the ER, where redox conditions are distinctively different from the other organelles and are favorable for disulfide formation. These reactions generate the production of reactive oxygen species (ROS) as a byproduct of thiol/disulfide exchange reaction among ER oxidoreductin 1 (Ero1), protein disulfide isomerase (PDI) and ER client proteins, during the formation of disulfide bonds in nascent or incorrectly folded proteins. When uncontrolled, this phenomenon perturbs ER homeostasis, thus aggravating the accumulation of improperly folded or unfolded proteins in this compartment (ER stress). This results in the activation of an adaptive mechanism named the unfolded protein response (UPR). In mammalian cells, the UPR is mediated by three ER-resident membrane proteins (PERK, IRE1 and ATF6) and regulates the expression of the UPR target genes, which themselves encode ER chaperones, folding enzymes, pro-apoptotic proteins and antioxidants, with the objective of restoring ER homeostatic balance. In this review, we will describe redox dependent activation (ER) and amplification (cytosol) loops that control the UPR and the consequences these regulatory loops have on cell fate and physiology.


Asunto(s)
Proteínas/metabolismo , Transducción de Señal , Respuesta de Proteína Desplegada , Animales , Retículo Endoplásmico/metabolismo , Oxidación-Reducción , Desplegamiento Proteico , Proteínas/química
14.
J Biol Chem ; 286(52): 44855-68, 2011 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-22025610

RESUMEN

The protein-disulfide isomerase (PDI) family member anterior gradient 2 (AGR2) is reportedly overexpressed in numerous cancers and plays a role in cancer development. However, to date the molecular functions of AGR2 remain to be characterized. Herein we have identified AGR2 as bound to newly synthesized cargo proteins using a proteomics analysis of endoplasmic reticulum (ER) membrane-bound ribosomes. Nascent protein chains that translocate into the ER associate with specific ER luminal proteins, which in turn ensures proper folding and posttranslational modifications. Using both imaging and biochemical approaches, we confirmed that AGR2 localizes to the lumen of the ER and indirectly associates with ER membrane-bound ribosomes through nascent protein chains. We showed that AGR2 expression is controlled by the unfolded protein response and is in turn is involved in the maintenance of ER homeostasis. Remarkably, we have demonstrated that siRNA-mediated knockdown of AGR2 significantly alters the expression of components of the ER-associated degradation machinery and reduces the ability of cells to cope with acute ER stress, properties that might be relevant to the role of AGR2 in cancer development.


Asunto(s)
Estrés del Retículo Endoplásmico/fisiología , Retículo Endoplásmico/enzimología , Regulación de la Expresión Génica/fisiología , Homeostasis/fisiología , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Respuesta de Proteína Desplegada/fisiología , Animales , Células COS , Chlorocebus aethiops , Perros , Retículo Endoplásmico/genética , Células HEK293 , Humanos , Ratones , Mucoproteínas , Proteínas Oncogénicas , Proteínas/genética , Proteínas Proto-Oncogénicas/genética
15.
FASEB J ; 25(9): 3115-29, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21680894

RESUMEN

Activation of the bifunctional kinase/RNase enzyme IRE1α is part of an adaptive response triggered on accumulation of misfolded proteins in the endoplasmic reticulum (ER). To facilitate recovery of ER homeostasis, IRE1α molecules oligomerize, allowing for their transautophosphorylation and endoribonuclease activation. These, in turn, induce the activation of specific transcriptional and post-transcriptional programs. To identify novel and selective modulators of IRE1α activity, we investigated IRE1α oligomerization properties using IRE1α-derived peptides identified through an activity-based in vitro assay. We then used these peptides to probe IRE1α activity in vitro and in vivo using both cultured human hepatocellular carcinoma-derived HuH7 cells and Caenorhabditis elegans experimental systems. We identified a peptide derived from the kinase domain of human IRE1α, which promoted IRE1α oligomerization in vitro, enhanced its Xbp1 mRNA cleavage activity in vitro (1.7×) in cell culture (1.8×) and in vivo (1.3×), and attenuated both ER stress-mediated JNK activation and regulated IRE1-dependent mRNA decay (RIDD). This was accompanied by a 2.5-fold increase in survival on tunicamycin-induced ER stress and reduced apoptosis by 1.4-fold in cells expressing this peptide. Hence, targeted and selective activation of the catalytic properties of IRE1α may consequently define new strategies to protect cells from deleterious effects of ER stress signaling.


Asunto(s)
Retículo Endoplásmico/metabolismo , Endorribonucleasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Caenorhabditis elegans , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endorribonucleasas/genética , Regulación de la Expresión Génica , Humanos , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Modelos Moleculares , Péptidos , Conformación Proteica , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción del Factor Regulador X , Transducción de Señal , Estrés Fisiológico , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína 1 de Unión a la X-Box
16.
Biosci Biotechnol Biochem ; 67(4): 914-8, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12784641

RESUMEN

Trichothecene 3-O-acetyltransferase (encoded by Tri101) inactivates the virulence factor of the cereal pathogen Fusarium graminearum. Zearalenone hydrolase (encoded by zhd101) detoxifies the oestrogenic mycotoxin produced by the same pathogen. These genes were introduced into a model monocotyledon rice plant to evaluate their usefulness for decontamination of mycotoxins. The strong and constitutive rice Act1 promoter did not cause accumulation of TRI101 protein in transgenic rice plants. In contrast, the same promoter was suitable for transgenic production of ZHD101 protein; so far, five promising T0 plants have been generated. Low transgenic expression of Tri101 was suggested to be increased by addition of an omega enhancer sequence upstream of the start codon.


Asunto(s)
Grano Comestible/genética , Proteínas Fúngicas , Micotoxinas/antagonistas & inhibidores , Plantas Modificadas Genéticamente , Acetiltransferasas/genética , Acetiltransferasas/metabolismo , Grano Comestible/microbiología , Elementos de Facilitación Genéticos , Fusarium/química , Fusarium/virología , Hidrolasas/genética , Hidrolasas/metabolismo , Regiones Promotoras Genéticas , Transformación Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA