Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
1.
BMC Biol ; 22(1): 105, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702628

RESUMEN

BACKGROUND: Histone H3K4 tri-methylation (H3K4me3) catalyzed by Set1/COMPASS, is a prominent epigenetic mark found in promoter-proximal regions of actively transcribed genes. H3K4me3 relies on prior monoubiquitination at the histone H2B (H2Bub) by Rad6 and Bre1. Swd2/Cps35, a Set1/COMPASS component, has been proposed as a key player in facilitating H2Bub-dependent H3K4me3. However, a more comprehensive investigation regarding the relationship among Rad6, Swd2, and Set1 is required to further understand the mechanisms and functions of the H3K4 methylation. RESULTS: We investigated the genome-wide occupancy patterns of Rad6, Swd2, and Set1 under various genetic conditions, aiming to clarify the roles of Set1 and Rad6 for occupancy of Swd2. Swd2 peaks appear on both the 5' region and 3' region of genes, which are overlapped with its tightly bound two complexes, Set1 and cleavage and polyadenylation factor (CPF), respectively. In the absence of Rad6/H2Bub, Set1 predominantly localized to the 5' region of genes, while Swd2 lost all the chromatin binding. However, in the absence of Set1, Swd2 occupancy near the 5' region was impaired and rather increased in the 3' region. CONCLUSIONS: This study highlights that the catalytic activity of Rad6 is essential for all the ways of Swd2's binding to the transcribed genes and Set1 redistributes the Swd2 to the 5' region for accomplishments of H3K4me3 in the genome-wide level.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Histonas , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Histonas/metabolismo , Histonas/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Metilación , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Enzimas Ubiquitina-Conjugadoras/genética
2.
BMB Rep ; 56(11): 600-605, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37401237

RESUMEN

Intrahepatic cholangiocarcinoma (ICC) is a bile duct cancer and a rare malignant tumor with a poor prognosis owing to the lack of an early diagnosis and resistance to conventional chemotherapy. A combination of gemcitabine and cisplatin is the typically attempted first-line treatment approach. However, the underlying mechanism of resistance to chemotherapy is poorly understood. We addressed this by studying dynamics in the human ICC SCK cell line. Here, we report that the regulation of glucose and glutamine metabolism was a key factor in overcoming cisplatin resistance in SCK cells. RNA sequencing analysis revealed a high enrichment cell cycle-related gene set score in cisplatin-resistant SCK (SCK-R) cells compared to parental SCK (SCK WT) cells. Cell cycle progression correlates with increased nutrient requirement and cancer proliferation or metastasis. Commonly, cancer cells are dependent upon glucose and glutamine availability for survival and proliferation. Indeed, we observed the increased expression of GLUT (glucose transporter), ASCT2 (glutamine transporter), and cancer progression markers in SCK-R cells. Thus, we inhibited enhanced metabolic reprogramming in SCK-R cells through nutrient starvation. SCK-R cells were sensitized to cisplatin, especially under glucose starvation. Glutaminase-1 (GLS1), which is a mitochondrial enzyme involved in tumorigenesis and progression in cancer cells, was upregulated in SCK-R cells. Targeting GLS1 with the GLS1 inhibitor CB-839 (telaglenastat) effectively reduced the expression of cancer progression markers. Taken together, our study results suggest that a combination of GLUT inhibition, which mimics glucose starvation, and GLS1 inhibition could be a therapeutic strategy to increase the chemosensitivity of ICC. [BMB Reports 2023; 56(11): 600-605].


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Humanos , Cisplatino/farmacología , Cisplatino/uso terapéutico , Glutamina/metabolismo , Glucosa/metabolismo , Proliferación Celular , Colangiocarcinoma/tratamiento farmacológico , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Línea Celular Tumoral , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología , Neoplasias de los Conductos Biliares/tratamiento farmacológico
3.
Mol Cells ; 46(8): 476-485, 2023 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-37460253

RESUMEN

Gastric cancer stem-like cells (GCSCs) possess stem cell properties, such as self-renewal and tumorigenicity, which are known to induce high chemoresistance and metastasis. These characteristics of GCSCs are further enhanced by autophagy, worsening the prognosis of patients. Currently, the mechanisms involved in the induction of stemness in GCSCs during autophagy remain unclear. In this study, we compared the cellular responses of GCSCs with those of gastric cancer intestinal cells (GCICs) whose stemness is not induced by autophagy. In response to glucose starvation, the levels of ß-catenin and stemness-related genes were upregulated in GCSCs, while the levels of ß-catenin declined in GCICs. The pattern of deubiquitinase ubiquitin C-terminal hydrolase-L3 (UCH-L3) expression in GCSCs and GCICs was similar to that of ß-catenin expression depending on glucose deprivation. We also observed that inhibition of UCH-L3 activity reduced ß-catenin protein levels. The interaction between UCH-L3 and ß-catenin proteins was confirmed, and it reduced the ubiquitination of ß-catenin. Our results suggest that UCH-L3 induces the stabilization of ß-catenin, which is required to promote stemness during autophagy activation. Also, UCH-L3 expression was regulated by c-Fos, and the levels of c-Fos increased in response to autophagy activation. In summary, our findings suggest that the inhibition of UCH-L3 during nutrient deprivation could suppress stress resistance of GCSCs and increase the survival rates of gastric cancer patients.


Asunto(s)
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , beta Catenina/metabolismo , Células Madre Neoplásicas/metabolismo , Ubiquitinación
4.
Cancer Sci ; 114(1): 187-200, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36114756

RESUMEN

Retinoic acid receptor-related orphan receptor α (RORα) is a transcription factor involved in nuclear gene expression and a known tumor suppressor. RORα was the first identified substrate of lysine methylation-dependent degradation. However, the mechanisms of other post-translational modifications (PTMs) that occur in RORα remain largely unknown, especially in liver cancer. Arginine methylation is a common PTM in arginine residues of nonhistone and histone proteins and affects substrate protein function and fate. We found an analogous amino acid disposition containing R37 at the ROR N-terminus compared to histone H3 residue, which is arginine methylated. Here, we provide evidence that R37 methylation-dependent degradation is carried out by protein arginine methyltransferase 5 (PRMT5). Further, we discovered that PRMT5 regulated the interaction between the E3 ubiquitin ligase ITCH and RORα through RORα arginine methylation. Arginine methylation-dependent ubiquitination-mediated RORα degradation reduced downstream target gene activation. H2 O2 -induced reactive oxygen species (ROS) decreased PRMT5 protein levels, consequently increasing RORα protein levels in HepG2 liver cancer cells. In addition, ROS inhibited liver cancer progression by inducing apoptosis via PRMT5-mediated RORα methylation and the ITCH axis. Our results potentiate PRMT5 as an elimination target in cancer therapy, and this additional regulatory level within ROS signaling may help identify new targets for therapeutic intervention in liver cancer.


Asunto(s)
Arginina , Neoplasias Hepáticas , Humanos , Metilación , Especies Reactivas de Oxígeno/metabolismo , Arginina/metabolismo , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Neoplasias Hepáticas/genética
5.
FEBS J ; 290(4): 951-961, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35869661

RESUMEN

Cell identity is determined by the transcriptional regulation of a cell-type-specific gene group. The Paf1 complex (Paf1C), an RNA polymerase II-associating factor, is an important transcriptional regulator that not only participates in transcription elongation and termination but also affects transcription-coupled histone modifications and chromatin organisation. Recent studies have shown that Paf1C is involved in the expression of genes required for self-renewal and pluripotency in stem cells and tumorigenesis. In this review, we focused on the role of Paf1C as a critical transcriptional regulator in cell fate decisions. Paf1C affects the pluripotency of stem cells by regulating the expression of core transcription factors such as Oct4 and Nanog. In addition, Paf1C directly binds to the promoters or distant elements of target genes, thereby maintaining the pluripotency in embryonic stem cells derived from an early stage of the mammalian embryo. Paf1C is upregulated in cancer stem cells, as compared with that in cancer cells, suggesting that Paf1C may be a target for cancer therapy. Interestingly, Paf1C is involved in multiple developmental stages in Drosophila, zebrafish, mice and even humans, thereby displaying a trend for the correlation between Paf1C and cell fate. Thus, we propose that Paf1C is a critical contributor to cell differentiation, cell specification and its characteristics and could be employed as a therapeutic target in developmental diseases.


Asunto(s)
Células Madre Embrionarias , Factores de Transcripción , Animales , Humanos , Ratones , Núcleo Celular/metabolismo , Células Madre Embrionarias/metabolismo , Mamíferos/metabolismo , Proteínas Nucleares/genética , Factores de Transcripción/genética , Pez Cebra/metabolismo
6.
Int J Radiat Biol ; 96(11): 1413-1422, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32856972

RESUMEN

PURPOSE: Cancer stem cells (CSCs) are relatively resistant to radiation compared to their non-tumorigenic progeny. Ionizing radiation (IR) can expand the pool of CSCs that leads to more aggressive cancers, but the reason underlying CSC-induced cancer aggressiveness after radiation therapy remains unclear. To understand this, we investigated the phenotypic and molecular characteristics of sphere cells formed from IR-treated patient-derived xenograft (PDX) lung adenocarcinoma tumors. MATERIALS AND METHODS: After treatment with various modes of IR, we collected tumors from PDX mice and successfully obtained sphere cells. To compare tumorigenicity, we performed migration, invasion, and mouse transplantation assays with sphere cells from each group. To investigate the molecular features, we used a cDNA microarray and compared gene expression among groups. RESULTS AND CONCLUSIONS: Tumorigenicity assays revealed that sphere cells from 2- or 5-Gy IR-treated tumors more aggressive than sphere cells from non-IR treated tumors. Microarray results showed that SERPIB4 and CCL2 were upregulated in sphere cells from IR-treated tumors compared to that in sphere cells from non-IR treated tumors. Interestingly, these genes are related to immune reactions in cancer. Taken together, our results suggest that the aggressiveness of sphere cells obtained after IR treatment is related to resistance, and provide new opportunities for exploring targeted therapies to overcome common radioresistance.


Asunto(s)
Adenocarcinoma del Pulmón/patología , Transformación Celular Neoplásica , Esferoides Celulares/efectos de la radiación , Adenocarcinoma del Pulmón/radioterapia , Animales , Biomarcadores de Tumor/metabolismo , Humanos , Ratones , Esferoides Celulares/metabolismo , Esferoides Celulares/patología
7.
Biomaterials ; 255: 120151, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32505033

RESUMEN

Neoadjuvant radiotherapy has become an important therapeutic option for colorectal cancer (CRC) patients, whereas complete tumor response is observed only in 20-30% patients. Therefore, the development of diagnostic probe for radio-resistance is important to decide an optimal treatment timing and strategy for radiotherapy-resistant CRC patients. In this study, using the patient-derived xenograft (PDX) mouse model established with a radio-resistant CRC tumor tissue, we found low-density lipoprotein receptor-related protein-1 (LRP-1) as a radio-resistant marker protein induced by initial-dose radiation in radio-resistant CRC tumors. Simultaneously, we discovered a LRP-1 targeting peptide in a radio-resistant CRC PDX through in vivo peptide screening. We next engineered the theranostic agent made of human serum albumin nanoparticles (HSA NPs) containing 5-FU for chemo-radiotherapy and decorating LRP-1-targeting peptide for tumor localization, Cy7 fluorophore for diagnostic imaging. The nanoparticle-based theranostic agent accurately targeted the tumor designated by LRP-1 responding radiation and showed dramatically improved therapeutic efficacy in the radio-resistant PDX model. In conclusion, we have identified LRP-1 as a signature protein of radio-resistant CRC and successfully developed LRP-1-targeting HSA-NP containing 5-FU that is a novel theranostic tool for both diagnostic imaging and neoadjuvant therapy of CRC patients. This approach is clinically applicable to improve the effectiveness of neo-adjuvant radiotherapy and increase the ratio of complete tumor response in radio-resistant CRC.


Asunto(s)
Neoplasias Colorrectales , Nanopartículas , Receptores de Lipoproteína , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Humanos , Ratones , Terapia Neoadyuvante , Medicina de Precisión
8.
Invest New Drugs ; 38(6): 1696-1706, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32462369

RESUMEN

Inhibitor of apoptosis proteins (IAPs) are overexpressed in the majority of cancers and prevent apoptosis by inhibiting caspases. IAPs have therefore attracted considerable attention as potential targets for anticancer therapy. Here, we demonstrated that HM90822 (abbreviated HM822; a new synthetic IAP antagonist) induced apoptotic cell death via proteasome-dependent degradation of BIR2/3 domain-containing IAPs in human pancreatic cancer cells. HM822 inhibited the expression of XIAP and cIAP1/2 proteins in Panc-1 and BxPC-3 cells, which are sensitive to HM822. HM822 also induced IAP ubiquitination and promoted proteasome-dependent IAP degradation. However, cells expressing phospho-XIAP (Ser87) and AKT exhibited resistance to HM822. In other words, the overexpression of AKT-CA (constitutive active form for AKT) or AKT-WT induced resistance to HM822. In addition, in Panc-1 xenograft and orthotopic mouse models, we revealed that tumor growth was suppressed by the administration of HM822. Taken together, these results suggest that HM822 induces apoptosis through ubiquitin/proteasome-dependent degradation of BIR3 domain-containing IAPs. These findings suggest that phospho-XIAP and phospho-AKT may be used as biomarkers for predicting the efficacy of HM822 in pancreatic cancer patients.


Asunto(s)
Antineoplásicos/uso terapéutico , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Neoplasias Pancreáticas/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Femenino , Humanos , Proteínas Inhibidoras de la Apoptosis/genética , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Carga Tumoral/efectos de los fármacos , Ubiquitinación/efectos de los fármacos
9.
FEBS J ; 287(14): 2891-2902, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31967712

RESUMEN

Histone modifications affect transcription by changing the chromatin structure. In particular, histone H3 lysine 4 trimethylation (H3K4me3) is one of the most recognized epigenetic marks of active transcription. While many studies have provided evidence of the correlation between H3K4me3 and active transcription, details regarding the mechanism involved remain unclear. The first study on the broad H3K4me3 domain was reported in 2014; subsequently, the function of this domain has been studied in various cell types. In this review, we summarized the recent studies on the role of the broad H3K4me3 domain in transcription, development, memory formation, and several diseases, including cancer and autoimmune diseases. The broadest H3K4me3 domains are associated with increased transcriptional precision of cell-type-specific genes related to cell identity and other essential functions. The broad H3K4me3 domain regulates maternal zygotic activation in early mammalian development. In systemic autoimmune diseases, high expression of immune-responsive genes requires the presence of the broad H3K4me3 domain in the promoter-proximal regions. Transcriptional repression of tumor-suppressor genes is associated with the shortening of the broad H3K4me3 domains in cancer cells. Additionally, the broad H3K4me3 domain interacts with the super-enhancer to regulate cancer-associated genes. During memory formation, H3K4me3 breadth is regulated in the hippocampus CA1 neurons. Taken together, these findings indicate that H3K4me3 breadth is essential for the regulation of the transcriptional output across multiple cell types.


Asunto(s)
Metilación de ADN , Enfermedad/genética , Epigénesis Genética , Regulación de la Expresión Génica , Histonas/química , Transcripción Genética , Animales , Código de Histonas , Humanos
10.
Genes Genomics ; 42(3): 273-281, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31797314

RESUMEN

BACKGROUND: Streptomyces seoulensis has contributed to the discovery and initiation of extensive research into nickel superoxide dismutase (NiSOD), a unique type of superoxide dismutase found in actinomycetes. Still so far, there is no information about whole genome sequence of this strain. OBJECTIVE: To investigate complete genome sequence and perform bioinformatic analyses for genomic functions related with nickel-associated genes. METHODS: DNA was extracted using the Wizard Genomic DNA Purification Kit then sequenced using a Pacific Biosciences SMRT cell 8Pac V3 DNA Polymerase Binding Kit P6 with the PacBiov2 RSII platform. We assembled the PacBio long-reads with the HGAP3 pipeline. RESULTS: We obtained complete genome sequence of S. seoulensis, which comprises a 6,339,363 bp linear chromosome. While analyzing the genome to annotate the genomic function, we discovered the nickel-associated genes. We observed that the sodN gene encoding for NiSOD is located adjacent to the sodX gene, which encodes for the nickel-type superoxide dismutase maturation protease. In addition, several nickel-associated genes and gene clusters-nickel-responsive regulator, nickel uptake transporter, nickel-iron [NiFe]-hydrogenase and other putative genes were also detected. Strain specific genes were discovered through a comparative analysis of S. coelicolor and S. griseus. Further bioinformatic analyses revealed that this strain encodes at least 22 putative biosynthetic gene clusters, thereby implying that S. seoulensis has the potential to produce novel bioactive compounds. CONCLUSION: We annotated the genome and determined nickel-associated genes and gene clusters and discovered biosynthetic gene clusters for secondary metabolites implying that S. seoulensis produces novel types of bioactive compounds.


Asunto(s)
Genoma Bacteriano , Streptomyces/genética , Superóxido Dismutasa/genética , Genómica , Familia de Multigenes , Níquel/metabolismo , Secuenciación Completa del Genoma
11.
Arch Pharm Res ; 42(9): 815-823, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-30879173

RESUMEN

RH1 (2, 5-diaziridinyl-3-(hydroxymethy)-6-methyl-1, 4-benzoquinone) is a bioreductive anticancer drug. The mechanism underlying its therapeutic properties has not yet been elucidated. In this study, we aimed to determine whether RH1 exerts its anticancer effect via p53-mediated apoptosis and senescence in vitro and in vivo. RH1 displayed dose-dependent biphasic effects in vitro, i.e., it induced apoptosis at higher dose and senescence at lower dose accompanied by marked activation of p53. Thus, RH1 primarily induced cell death by apoptosis. The cytotoxicity of RH1 was inhibited in A549 cells treated with the p53-inhibitor pifithrin-α or transfected p53 siRNA and in human colon cancer HCT116 isogenic (p53-/-) cells. At sub-lethal doses of RH1, the cells survived and underwent senescence. The senescent cells showed flattened and enlarged morphology, and exhibited blue color in senescence-associated ß-galactosidase staining. These changes were found to be related to p53. RH1-induced senescence decreased in A549-E6 cells (suppressed p53 level) and HCT 116 p53-/- cells. The growth of A549 xenograft tumors in nude mice was significantly delayed by intraperitoneal injection of RH1, and senescent cells were observed in these xenograft tumors. These results suggest that the in vivo anticancer therapeutic effect of RH1 is mediated by senescence via p53 activation.


Asunto(s)
Antineoplásicos/farmacología , Aziridinas/farmacología , Benzoquinonas/farmacología , Senescencia Celular/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Células A549 , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Relación Estructura-Actividad , Proteína p53 Supresora de Tumor/genética
12.
Microbiologyopen ; 8(4): e00680, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-29949242

RESUMEN

Ribosome often stalls on mRNA sequences harboring consecutive proline codons. Elongation factor P (EF-P) is required for the stalled ribosome to continue translation and thus the absence of EF-P affects translation of the associated open reading frame. Here we report that EF-P controls translation of the mgtA gene encoding a Mg2+ -transporting ATPase from the intracellualr pathogen Salmonella enterica serovar Typhimurium. EF-P's effect on mgtA translation is dependent on the 550th and 551st proline codons in the coding region and thus substitution of those proline codons eliminates EF-P-mediated control of MgtA protein without affecting the Mg2+ -transporting activity of the mgtA gene. The Pro550 and Pro551-substituted mgtA gene promotes Salmonella's intramacrophage survival and mouse virulence, suggesting that EF-P-mediated translational control of the mgtA gene is required for Salmonella pathogenesis.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Proteínas de Transporte de Membrana/metabolismo , Factores de Elongación de Péptidos/metabolismo , Biosíntesis de Proteínas , Infecciones por Salmonella/microbiología , Salmonella typhimurium/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Secuencias de Aminoácidos , Animales , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Femenino , Eliminación de Gen , Humanos , Magnesio/metabolismo , Proteínas de Transporte de Membrana/química , Proteínas de Transporte de Membrana/genética , Ratones , Factores de Elongación de Péptidos/genética , Salmonella typhimurium/química , Salmonella typhimurium/genética
13.
Anticancer Res ; 38(1): 287-293, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29277785

RESUMEN

BACKGROUND/AIM: To maximize success rate for development of HER2-targeted therapeutics, patient-derived xenograft (PDX) models reflecting HER2-positive gastric cancer (HER2+ GC) patients were established. MATERIALS AND METHODS: GC tissues obtained from surgery of GC patients were implanted into immune-deficient mice, and tumor tissue of HER2+ PDXs were verified of the patient-mimic HER2 expression by immunohistochemistry and explored for the feasibility by testing with Herceptin, the approved therapeutics and novel HER2 antibody therapeutics being developed. RESULTS: We obtained 5 cases of HER2+ GC PDX models reflecting patient's GC tumor, consisting of 2 cases of HER2 3+ and 2 cases of HER2 2+. Novel HER2 antibody displayed significantly improved anti-cancer efficacy in combination with Herceptin. CONCLUSION: The HER2+ GC PDX models were successfully established to be utilized for preclinical evaluation of HER2-targeting drugs and combined therapies for GC treatment, as an ideal platform of personalized tools for precision therapy.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos Inmunológicos/uso terapéutico , Receptor ErbB-2/antagonistas & inhibidores , Neoplasias Gástricas/tratamiento farmacológico , Trastuzumab/uso terapéutico , Adenocarcinoma/patología , Anciano , Animales , Antineoplásicos Inmunológicos/farmacología , Femenino , Humanos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Medicina de Precisión , Receptor ErbB-2/inmunología , Receptor ErbB-2/metabolismo , Neoplasias Gástricas/patología , Trastuzumab/farmacología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
J Microbiol Biotechnol ; 28(2): 181-189, 2018 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-29169224

RESUMEN

Candida albicans is a major pathogenic fungus in humans, and meets at first the innate immune cells, such as macrophages, in its host. One important strategy of the host cell to kill C. albicans is to produce reactive oxygen species (ROS) by the macrophages. In response to ROS produced by the macrophages, C. albicans operates its defense mechanisms against them by expressing its oxidative stress response genes. Although there have been many research studies explaining the specific transcription factors and the expression of the oxidative stress genes in C. albicans, the regulation of the oxidative stress genes by chromatin structure is little known. Epigenetic regulation by the chromatin structure is very important for the regulation of eukaryotic gene expression, including the chromatin structure dynamics by histone modifications. Among various histone modifications, histone acetylation is reported for its direct relationship to the regulation of gene expression. Recent studies reported that histone acetyltransferases regulate genes to respond to the oxidative stress in C. albicans. In this review, we introduce all histone acetyltransferases that C. albicans contains and some papers that explain how histone acetyltransferases participate in the oxidative stress response in C. albicans.


Asunto(s)
Candida albicans/enzimología , Candida albicans/metabolismo , Histona Acetiltransferasas/metabolismo , Estrés Oxidativo/genética , Candida albicans/patogenicidad , Candidiasis/microbiología , Epigénesis Genética , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Regulación Fúngica de la Expresión Génica , Inestabilidad Genómica , Interacciones Huésped-Patógeno/fisiología , Macrófagos/metabolismo , Macrófagos/microbiología , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción/genética , Virulencia/genética
15.
Oncol Rep ; 38(4): 2259-2266, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28849069

RESUMEN

The incidence of hepatocellular carcinoma (HCC) has continued to increase worldwide, and advanced HCC is difficult to treat using the currently available therapeutics. Chemoradiotherapy with cisplatin (cis-diamminedichloroplatinum, CDDP) is expected to confer a curative benefit on HCC patients; however, its application is limited due to side-effects such as acute nephrotoxicity as well as the conventionally limited application of chemoradiotherapy for HCC. For the practical application of this drug in the clinical setting, we formulated a novel drug carrier-comprising bio-nanocapsule (BNC) and liposomal CDDP (BNC-LP-CDDP) that recognizes the human liver and releases CDDP. BNC-LP-CDDP showed selectively high cytotoxicity for HCC cells, and markedly reduced the survival fractions of HCC when combined with ionizing radiation (IR) treatment in in vitro assays. In particular, the treatment of mice bearing human HCC with BNC-LP-CDDP and 3 Gy IR showed 95.68% growth inhibition, whereas IR treatment alone showed 65.6% growth inhibition. Moreover, BNC-LP-CDDP led to the withdrawal of CDDP-induced nephrotoxicity. These results indicate that BNC-LP-CDDP in combination with IR markedly enhanced the chemo-radiotherapeutic efficacy and eliminated CDDP induced nephrotoxicity, thus, suggesting the potential for its clinical application as human HCC therapy.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/radioterapia , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/radioterapia , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Quimioradioterapia/métodos , Cisplatino/administración & dosificación , Cisplatino/química , Terapia Combinada , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/química , Humanos , Liposomas/administración & dosificación , Liposomas/química , Neoplasias Hepáticas/patología , Ratones , Nanocápsulas/administración & dosificación , Nanocápsulas/química , Radiación Ionizante , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Biomaterials ; 141: 136-148, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28688285

RESUMEN

Designing nanocarriers with active targeting has been increasingly emphasized as for an ideal delivery mechanism of anti-cancer therapeutic agents, but the actualization has been constrained by lack of reliable strategy ultimately applicable. Here, we designed and verified a strategy to achieve active targeting nanomedicine that works in a living body, utilizing animal models bearing a patient's tumor tissue and subjected to the same treatments that would be used in the clinic. The concept for this strategy was that a novel peptide probe and its counterpart protein, which responded to a therapy, were identified, and then the inherent ability of the peptide to target the designated tumor protein was used for active targeting in vivo. An initial dose of ionizing radiation was locally delivered to the gastric cancer (GC) tumor of a patient-derived xenograft mouse model, and phage-displayed peptide library was intravenously injected. The peptides tightly bound to the tumor were recovered, and the counterpart protein was subsequently identified. Peptide-conjugated liposomal drug showed dramatically improved therapeutic efficacy and possibility of diagnostic imaging with radiation. These results strongly suggested the potential of our strategy to achieve in vivo functional active targeting and to be applied clinically for human cancer treatment.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Liposomas/química , Péptidos/química , Neoplasias Gástricas/tratamiento farmacológico , Animales , Antibióticos Antineoplásicos/uso terapéutico , Doxorrubicina/uso terapéutico , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanomedicina , Nanopartículas/química , Biblioteca de Péptidos , Neoplasias Gástricas/patología
17.
Sci Rep ; 7: 42098, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28181542

RESUMEN

When a ribosome translates mRNA sequences, the ribosome often stalls at certain codons because it is hard to translate. Consecutive proline codons are such examples that induce ribosome stalling and elongation factor P (EF-P) is required for the stalled ribosome to continue translation at those consecutive proline codons. We found that EF-P is required for translation of the mgtB gene encoding a Mg2+ transporter in the mgtCBR virulence operon from the intracellular pathogen Salmonella enterica serovar Typhimurium. Salmonella lacking EF-P decreases MgtB protein levels in a manner dependent on consecutive proline codons located in the mgtB coding region despite increasing transcription of the mgtCBR operon via the mgtP open reading frame in the leader RNA, resulting in an altered ratio between MgtC and MgtB proteins within the operon. Substitution of the consecutive proline codons to alanine codons eliminates EF-P-mediated control of the mgtB gene during infection and thus contributes to Salmonella's survival inside macrophages where Salmonella experiences low levels of EF-P. This suggests that this pathogen utilizes a strategy to coordinate expression of virulence genes by an evolutionarily conserved translation factor.


Asunto(s)
Adenosina Trifosfatasas/biosíntesis , Proteínas Bacterianas/biosíntesis , Proteínas de Transporte de Catión/biosíntesis , Regulación Bacteriana de la Expresión Génica , Factores de Elongación de Péptidos/metabolismo , Biosíntesis de Proteínas , Salmonelosis Animal/microbiología , Salmonella typhimurium/crecimiento & desarrollo , Salmonella typhimurium/genética , Animales , Línea Celular , Perfilación de la Expresión Génica , Macrófagos/microbiología , Ratones Endogámicos C3H , Viabilidad Microbiana , Factores de Elongación de Péptidos/deficiencia , Virulencia
18.
Int J Biochem Cell Biol ; 83: 47-55, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27923747

RESUMEN

Tumor necrosis factor-related apoptosis-induced ligand (TRAIL) induces apoptosis selectively in cancer cells without affecting the majority of normal human cells. However, hepatocellular carcinoma (HCC) cells often display resistance to TRAIL-induced apoptosis. Ibulocydine (IB) is an isobutyrate ester pro-drug of a novel synthetic Cdk inhibitor that targets Cdk7 and Cdk9. In this study, we show that treatment with subtoxic doses of IB in combination with TRAIL displays potent cytotoxicity in TRAIL-resistant human HCC cells. Combination of IB and TRAIL was found to synergistically induce apoptosis through activation of caspases, which was blocked by a pan-caspase inhibitor (zVAD). Although the expression of Mcl-1 and survivin were reduced by IB plus TRAIL, overexpression of Mcl-1 and survivin did not block the cell death induced by co-treatment. Moreover, overexpression of Bcl-xL did not significantly interfere with the cell death induced by co-treatment of IB and TRAIL. Interestingly, the combination treatment induced cleavage of Bax, which was translocated to mitochondria upon induction of apoptosis. Furthermore, down-regulation of Bax by small interfering RNA effectively reduced the cell death and loss of mitochondrial membrane potential (MMP) caused by co-treatment with IB and TRAIL. Finally, pre-treatment of HCC cells with a calpain inhibitor effectively blocked IB plus TRAIL-induced cleavage of Bax and apoptosis. Collectively, our results demonstrate that IB increases the sensitivity of human HCC cells to TRAIL via mitochondria signaling pathway mediated by calpain-induced cleavage of Bax, suggesting that combined treatment with IB and TRAIL may offer an effective therapeutic strategy for human HCC.


Asunto(s)
Apoptosis/efectos de los fármacos , Calpaína/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Profármacos/farmacología , Nucleósidos de Pirimidina/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Apoptosis/fisiología , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Caspasas/metabolismo , Línea Celular Tumoral , Sinergismo Farmacológico , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Profármacos/administración & dosificación , Nucleósidos de Pirimidina/administración & dosificación , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Regulación hacia Arriba , Proteína X Asociada a bcl-2/antagonistas & inhibidores , Proteína X Asociada a bcl-2/genética , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
19.
Lung Cancer ; 101: 137-144, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27794402

RESUMEN

INTRODUCTION: Treatment of non-small cell lung cancer (NSCLC) is an important and often unmet medical need regardless of the disease stage at the time of first diagnosis. Antigen-specific immunotherapy may be a feasible therapeutic option if tumor associated antigens (TAAs) that can be targeted by the patient's immune system are identified. The study objective (NCT01837511) was to investigate the expression rates of MAGE-A3 and PRAME in tumors from East Asian NSCLC patients, and the associations between TAA expression and clinico-pathologic patient characteristics. METHODS: Archived formalin-fixed paraffin-embedded tumor tissue specimens were tested for MAGE-A3 and PRAME expression by quantitative reverse transcription polymerase chain reaction. Exploratory analyses of the impact of patient and tumor characteristics on antigen expression were performed by multivariate logistic regression analyses. RESULTS: A total of 377 specimens were tested and a valid expression result was obtained for 86.5% and 92.6% for MAGE-A3 and PRAME, respectively. Of the specimens with valid test results, 26.4% expressed MAGE-A3, 49.9% PRAME, 20.0% both and 57.5% expressed at least one TAA. The same pattern of associations between antigen expression and patient and tumor characteristics was found for both TAAs: higher rates of antigen-positive tumors were found in squamous cell carcinomas compared to adenocarcinomas, and for smokers compared to non-smokers. CONCLUSIONS: Expression of MAGE-A3 and PRAME suggests an association with tumor histology and the patient's smoking status. The rates of TAA-positive tumors found in these East and South East Asian NSCLC patients indicate that both antigens may serve as targets for antigen-specific immunotherapies.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/metabolismo , Prevalencia , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Asia Sudoriental/epidemiología , Biomarcadores de Tumor/análisis , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Asia Oriental/epidemiología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoterapia/métodos , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Estudios Retrospectivos , Fumar/epidemiología
20.
Oncotarget ; 7(47): 77348-77357, 2016 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-27764799

RESUMEN

We assessed the efficacy of the polymeric nanoparticle containing docetaxel (PNP-DTX) in preclinical mouse models and determined the maximum tolerated dose (MTD) through clinical study. Subcutaneous and orthotopic mouse models were dedicated. Tumor growth delay in orthotopic model and quantification of in vivo imaging in orthotopic model were evaluated. Phase I clinical study was a single-center, prospective, open-label trial in advanced solid tumors. PNP-DTX was injected intravenously and the starting dose was 20 mg/m2 escalated to 35 mg/m2, 45 mg/m2, 60 mg/m2 and 75 mg/m2. Pharmacokinetics, tumor response, toxicities were evaluated. Preclinical result revealed the more potent cytotoxic effect of PNP-DTX than docetaxel (DTX). However, there was no difference between PNP-DTX and DTX in subcutaneous model. Tubulin polymerization assay showed that PNP-DTX preserved original mode of action of DTX. For phase I clinical trial, 18 patients were analyzed. The dose of 75 mg/m2 was tentatively determined as the MTD and the most common toxicity was grade 4 neutropenia not lasting over 7days. The Cmax of 60 mg/m2 PNP-DTX and AUClast of 45 mg/m2 PNP-DTX were measured to be comparable to those of 75 mg/m2 DTX. Partial remission (PR) was achieved in 4 (22%) patients. The potency of PNP-DTX was revealed especially in orthotopic mouse model. The MTD of PNP-DTX could not be confirmed, but 75 mg/m2 was tentatively determined. The PNP-DTX of 45 mg/m2 had the same pharmacokinetic profile with that of 75 mg/m2 DTX.


Asunto(s)
Antineoplásicos/administración & dosificación , Nanopartículas , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Polímeros , Taxoides/administración & dosificación , Moduladores de Tubulina/administración & dosificación , Adulto , Anciano , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacocinética , Línea Celular Tumoral , Modelos Animales de Enfermedad , Docetaxel , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Humanos , Masculino , Dosis Máxima Tolerada , Ratones , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Taxoides/efectos adversos , Taxoides/farmacocinética , Resultado del Tratamiento , Moduladores de Tubulina/efectos adversos , Moduladores de Tubulina/farmacocinética , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA