Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Phys Chem Lett ; 15(29): 7398-7402, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-38995855

RESUMEN

Understanding the structural variations of conformational isomers in proteins is crucial for elucidating protein folding mechanisms. Here, we present a novel method for obtaining conformation-selective ultraviolet (UV)-UV hole burning (HB) spectra of ubiquitin ions ((Ubi+zH)+z, z = 7-10) produced via electrospray ionization. Our approach involves binding multiple N2 molecules to ubiquitin ions ((Ubi+zH)+z(N2)m, m = 1-55) within a cryogenic ion trap. Upon exposure to UV irradiation, efficient fragmentation of (Ubi+zH)+z(N2)m occurs, primarily yielding bare (Ubi+zH)+z ions as fragments. The significant mass difference between the parent and fragment ions facilitates the acquisition of UV-UV HB spectra, which reveal the presence of at least two distinct conformers. Molecular dynamics simulations suggest that these conformers correspond to A-state structures, differing only in the interactions of a tyrosine residue with neighboring residues. Our findings underscore UV-UV HB spectroscopy of protein ions as a powerful tool for exploring diverse protein isomers.


Asunto(s)
Iones , Simulación de Dinámica Molecular , Ubiquitina , Rayos Ultravioleta , Ubiquitina/química , Iones/química , Conformación Proteica , Espectrometría de Masa por Ionización de Electrospray
2.
Phys Chem Chem Phys ; 25(34): 22644-22649, 2023 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-37591819

RESUMEN

Anisotropic circular dichroism (CD) refers to the CD of oriented molecules, which varies with the direction of light propagation toward the molecules. Thus, anisotropic CD spectroscopy has been used to investigate the orientations of molecules in anisotropic media such as liquid crystals and thin films. However, it is unclear whether anisotropic CD results from isolated chromophores or their intermolecular interactions with other atoms or molecules that form anisotropically aligned structures. Herein, anisotropic CD of isolated chiral molecules was observed for the first time. The resonant two-photon ionization CD spectra of jet-cooled pseudoephedrine and styrene oxide indicated a difference between the CD values of the P/R and Q branches of the origin bands of the S0-S1 transition. This difference may have resulted from the anisotropic CD phenomena of these molecules, which are oriented via photoselection. Although jet-cooled molecules may have nearly random orientations, those excited to the P/R or Q branch become oriented because the transition probability to these branches depends on the molecular orientation relative to the direction of light propagation. These results demonstrate that the CD spectra of cold, isolated molecules, such as those in an interstellar medium, may exhibit anisotropic CD values.

3.
J Phys Chem A ; 126(26): 4295-4299, 2022 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-35758322

RESUMEN

Circular dichroism (CD) spectroscopy of jet-cooled molecules provides conformation-specific CD spectra. However, its widespread utilization has been limited by the weak CD effects and the low density of gas-phase molecules. Here, we developed a dual-beam method to improve the sensitivity and accuracy of gas-phase CD measurements. Circularly and linearly polarized pulses were generated from a single laser pulse and used to irradiate a single molecular-beam pulse to produce two ion peaks. The ion peaks induced by linearly polarized pulses were subtracted from those induced by circularly polarized pulses to correct the CD values for the pulse-to-pulse fluctuations in laser power and gas density. The resonant two-photon ionization CD spectrum of (1R,2R)-(-)-pseudoephedrine revealed that the standard deviations of CD values measured using the dual-beam method were three times lower than those measured using a single-beam method. The dual-beam method provides an effective, accurate, and easy-to-use tool to obtain gas-phase CD spectra.

4.
Clin Transl Immunology ; 11(1): e1364, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35003748

RESUMEN

OBJECTIVES: AXL-mediated activation of aberrant tyrosine kinase drives various oncogenic processes and facilitates an immunosuppressive microenvironment. We evaluated the anti-tumor and anti-metastatic activities of SKI-G-801, a small-molecule inhibitor of AXL, alone and in combination with anti-PD-1 therapy. METHODS: In vitro pAXL inhibition by SKI-G-801 was performed in both human and mouse cancer cell lines. Immunocompetent mouse models of tumor were established to measure anti-metastatic potential of SKI-G-801. Furthermore, SKI-G-801, anti-PD-1 or their combination was administered as an adjuvant or neoadjuvant in the 4T1 tumor model to assess their potential for clinical application. RESULTS: SKI-G-801 robustly inhibited pAXL expression in various cell lines. SKI-G-801 alone or in combination with anti-PD-1 potently inhibited metastasis in B16F10 melanoma, CT26 colon and 4T1 breast models. SKI-G-801 inhibited the growth of B16F10 and 4T1 tumor-bearing mice but not immune-deficient mice. An antibody depletion assay revealed that CD8+ T cells significantly contributed to SKI-G-801-mediated survival. Anti-PD-1 and combination group were observed the increased CD8+Ki67+ and effector T cells and M1 macrophage and decreased M2 macrophage, and granulocytic myeloid-derived suppressor cell (G-MDSC) compared to the control group. The neoadjuvant combination of SKI-G-801 and anti-PD-1 therapy achieved superior survival benefits by inducing more profound T-cell responses in the 4T1 syngeneic mouse model. CONCLUSION: SKI-G-801 significantly suppressed tumor metastasis and growth by enhancing anti-tumor immune responses. Our results suggest that SKI-G-801 has the potential to overcome anti-PD-1 therapy resistance and allow more patients to benefit from anti-PD-1 therapy.

5.
J Hematol Oncol ; 14(1): 148, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34530889

RESUMEN

BACKGROUND: Little is known about endogenous inhibitors of angiogenic growth factors. In this study, we identified a novel endogenous anti-angiogenic factor expressed in pericytes and clarified its underlying mechanism and clinical significance. METHODS: Herein, we found Kai1 knockout mice showed significantly enhanced angiogenesis. Then, we investigated the anti-angiogenic roll of Kai1 in vitro and in vivo. RESULTS: KAI1 was mainly expressed in pericytes rather than in endothelial cells. It localized at the membrane surface after palmitoylation by zDHHC4 enzyme and induced LIF through the Src/p53 pathway. LIF released from pericytes in turn suppressed angiogenic factors in endothelial cells as well as in pericytes themselves, leading to inhibition of angiogenesis. Interestingly, KAI1 had another mechanism to inhibit angiogenesis: It directly bound to VEGF and PDGF and inhibited activation of their receptors. In the two different in vivo cancer models, KAI1 supplementation significantly inhibited tumor angiogenesis and growth. A peptide derived from the large extracellular loop of KAI1 has been shown to have anti-angiogenic effects to block the progression of breast cancer and retinal neovascularization in vivo. CONCLUSIONS: KAI1 from PC is a novel molecular regulator that counterbalances the effect of angiogenic factors.


Asunto(s)
Proteína Kangai-1/metabolismo , Neovascularización Patológica/metabolismo , Neovascularización Fisiológica , Animales , Femenino , Proteína Kangai-1/genética , Masculino , Microdominios de Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Cancer Discov ; 10(8): 1194-1209, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32414908

RESUMEN

EGFR exon 20 insertion driver mutations (Exon20ins) in non-small cell lung cancer (NSCLC) are insensitive to EGFR tyrosine kinase inhibitors (TKI). Amivantamab (JNJ-61186372), a bispecific antibody targeting EGFR-MET, has shown preclinical activity in TKI-sensitive EGFR-mutated NSCLC models and in an ongoing first-in-human study in patients with advanced NSCLC. However, the activity of amivantamab in Exon20ins-driven tumors has not yet been described. Ba/F3 cells and patient-derived cells/organoids/xenograft models harboring diverse Exon20ins were used to characterize the antitumor mechanism of amivantamab. Amivantamab inhibited proliferation by effectively downmodulating EGFR-MET levels and inducing immune-directed antitumor activity with increased IFNγ secretion in various models. Importantly, in vivo efficacy of amivantamab was superior to cetuximab or poziotinib, an experimental Exon20ins-targeted TKI. Amivantamab produced robust tumor responses in two Exon20ins patients, highlighting the important translational nature of this preclinical work. These findings provide mechanistic insight into the activity of amivantamab and support its continued clinical development in Exon20ins patients, an area of high unmet medical need. SIGNIFICANCE: Currently, there are no approved targeted therapies for EGFR Exon20ins-driven NSCLC. Preclinical data shown here, together with promising clinical activity in an ongoing phase I study, strongly support further clinical investigation of amivantamab in EGFR Exon20ins-driven NSCLC.This article is highlighted in the In This Issue feature, p. 1079.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Animales , Anticuerpos Biespecíficos/farmacología , Antineoplásicos Inmunológicos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Exones , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Mutación , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/metabolismo
7.
Sci Rep ; 9(1): 19909, 2019 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-31882684

RESUMEN

Adequate preclinical model and model establishment procedure are required to accelerate translational research in lung cancer. We streamlined a protocol for establishing patient-derived cells (PDC) and identified effective targeted therapies and novel resistance mechanisms using PDCs. We generated 23 PDCs from 96 malignant effusions of 77 patients with advanced lung adenocarcinoma. Clinical and experimental factors were reviewed to identify determinants for PDC establishment. PDCs were characterized by driver mutations and in vitro sensitivity to targeted therapies. Seven PDCs were analyzed by whole-exome sequencing. PDCs were established at a success rate of 24.0%. Utilizing cytological diagnosis and tumor colony formation can improve the success rate upto 48.8%. In vitro response to a tyrosine kinase inhibitor (TKI) in PDC reflected patient treatment response and contributed to identifying effective therapies. Combination of dabrafenib and trametinib was potent against a rare BRAF K601E mutation. Afatinib was the most potent EGFR-TKI against uncommon EGFR mutations including L861Q, G719C/S768I, and D770_N771insG. Aurora kinase A (AURKA) was identified as a novel resistance mechanism to olmutinib, a mutant-selective, third-generation EGFR-TKI, and inhibition of AURKA overcame the resistance. We presented an efficient protocol for establishing PDCs. PDCs empowered precision medicine with promising translational values.


Asunto(s)
Adenocarcinoma del Pulmón/terapia , Neoplasias Pulmonares/terapia , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/metabolismo , Línea Celular , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Receptores ErbB/genética , Citometría de Flujo , Humanos , Immunoblotting , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Mutación/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Secuenciación del Exoma
8.
Clin Cancer Res ; 25(8): 2575-2587, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30670498

RESUMEN

PURPOSE: Given that osimertinib is the only approved third-generation EGFR-TKI against EGFR activating and resistant T790M mutated non-small cell lung cancer (NSCLC), additional mutant-selective inhibitors with a higher efficacy, especially for brain metastases, with favorable toxicity profile are still needed. In this study, we investigated the antitumor efficacy of YH25448, an oral, mutant-selective, irreversible third-generation EGFR-TKI in preclinical models. EXPERIMENTAL DESIGN: Antitumor activity of YH25448 was investigated in vitro using mutant EGFR-expressing Ba/F3 cells and various lung cancer cell lines. In vivo antitumor efficacy, ability to penetrate the blood-brain barrier (BBB), and skin toxicity of YH25448 were examined and compared with those of osimertinib using cell lines and PDX model. RESULTS: Compared with osimertinib, YH25448 showed a higher selectivity and potency in kinase assay and mutant EGFR-expressing Ba/F3 cells. In various cell line models harboring EGFR activating and T790M mutation, YH25448 effectively inhibited EGFR downstream signaling pathways, leading to cellular apoptosis. When compared in vivo at equimolar concentrations, YH25448 produced significantly better tumor regression than osimertinib. Importantly, YH25448 induced profound tumor regression in brain metastasis model with excellent brain/plasma and tumor/brain area under the concentration-time curve value. YH25448 rarely suppressed the levels of p-EGFR in hair follicles, leading to less keratosis than osimertinib in animal model. The potent systemic and intracranial activity of YH25448 has been shown in an ongoing phase I/II clinical trial for advanced EGFR T790M mutated NSCLC (NCT03046992). CONCLUSIONS: Our findings suggest that YH25448 is a promising third-generation EGFR inhibitor, which may be more effective and better tolerated than the currently approved osimertinib.


Asunto(s)
Neoplasias Encefálicas/secundario , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Inhibidores de Proteínas Quinasas/farmacología , Acrilamidas/química , Acrilamidas/farmacología , Acrilamidas/uso terapéutico , Adulto , Compuestos de Anilina/química , Compuestos de Anilina/farmacología , Compuestos de Anilina/uso terapéutico , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/mortalidad , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/química , Receptores ErbB/genética , Humanos , Neoplasias Pulmonares/diagnóstico por imagen , Masculino , Ratones , Modelos Moleculares , Mutación , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/uso terapéutico , Relación Estructura-Actividad , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Molecules ; 23(7)2018 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-29986512

RESUMEN

Ukgansan (UGS), a traditional herbal formula composing seven medicinal herbal plants, has been applied in Asian countries for treating neurosis, insomnia, and irritability. Here, the current study performed a simultaneous determination of the seven marker compounds (liquiritin apioside, liquiritin, ferulic acid, glycyrrhizin, decursin, decursinol angelate, and atractylenolide I) using high-performance liquid chromatography (HPLC), to establish quality control of UGS. A 70% ethanol extract of UGS and a mixture of the seven compounds were separated using a C-18 analytical column on a gradient solvent system of 1.0% (v/v) aqueous acetic acid and acetonitrile. Data were recorded at a UV wavelength of 250 nm for glycyrrhizin; 276 nm for liquiritin apioside, liquiritin, and atractylenolide I; and 325 nm for ferulic acid, decursin, and decursinol angelate. The results exhibited high linearity (correlation coefficient (r²) ≥ 0.9998) and proper precision (0.38⁻3.36%), accuracy (95.12⁻105.12%), and recovery (95.99⁻104.94%) for the seven marker compounds. The amount of the seven marker compounds at the concentrations from 0.190 to 16.431 mg/g. In addition, the current study evaluated the antioxidant effects of UGS by measuring their scavenging activities against the 2,2'-azinobis-(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) and 2,2'-diphenyl-1-picrylhydrazyl (DPPH) radicals using in vitro cell-free systems and observed its antioxidant activity. Among the seven components of the UGS extract, ferulic acid dramatically enhanced the scavenging of ABTS and DPPH radicals compared with other compounds. The concentrations of ferulic acid required for a 50% reduction (RC50) in ABTS and DPPH radicals were 16.22 µM and 41.21 µM, respectively. Furthermore, UGS extract exerted the neuroprotective effect and blocked the inflammatory response in neuronal hippocampal cells and microglia, respectively. Overall, the established method of HPLC will be valuable for improving the quality control of UGS extract, and ferulic acid may be useful as a potential antioxidant agent.


Asunto(s)
Antioxidantes/farmacología , Ácidos Cumáricos/farmacología , Medicamentos Herbarios Chinos/análisis , Neuronas/citología , Animales , Antioxidantes/química , Antioxidantes/aislamiento & purificación , Línea Celular , Ácidos Cumáricos/química , Ácidos Cumáricos/aislamiento & purificación , Técnicas In Vitro , Ratones , Estructura Molecular , Neuronas/efectos de los fármacos , Control de Calidad
10.
EMBO Rep ; 17(9): 1343-59, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27466323

RESUMEN

Epithelial to mesenchymal transition (EMT) and mesenchymal to epithelial transition (MET) are important interconnected events in tumorigenesis controlled by complex genetic networks. However, the cues that activate EMT-initiating factors and the mechanisms that reversibly connect EMT/MET are not well understood. Here, we show that cohesin-mediated chromatin organization coordinates EMT/MET by regulating mesenchymal genes. We report that RAD21, a subunit of the cohesin complex, is expressed in epithelial breast cancer cells, whereas its expression is decreased in mesenchymal cancer. Depletion of RAD21 in epithelial cancer cells causes transcriptional activation of TGFB1 and ITGA5, inducing EMT. Reduced binding of RAD21 changes intrachromosomal chromatin interactions within the TGFB1 and ITGA5 loci, creating an active transcriptional environment. Similarly, stem cell-like cancer cells also show an open chromatin structure at both genes, which correlates with high expression levels and mesenchymal fate characteristics. Conversely, overexpression of RAD21 in mesenchymal cancer cells induces MET-specific expression patterns. These findings indicate that dynamic cohesin-mediated chromatin structures are responsible for the initiation and regulation of essential EMT-related cell fate changes in cancer.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromatina/genética , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Transición Epitelial-Mesenquimal/genética , Neoplasias/genética , Neoplasias/metabolismo , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión al ADN , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Metástasis de la Neoplasia , Neoplasias/patología , Proteínas Nucleares/genética , Fosfoproteínas/genética , Regiones Promotoras Genéticas , Factor de Crecimiento Transformador beta1/genética , Cohesinas
11.
Cell Stem Cell ; 18(4): 508-21, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26996598

RESUMEN

Hematopoiesis is regulated by crosstalk between long-term repopulating hematopoietic stem cells (LT-HSCs) and supporting niche cells in the bone marrow (BM). Here, we examine the role of CD82/KAI1 in niche-mediated LT-HSC maintenance. We found that CD82/KAI1 is expressed predominantly on LT-HSCs and rarely on other hematopoietic stem-progenitor cells (HSPCs). In Cd82(-/-) mice, LT-HSCs were selectively lost as they exited from quiescence and differentiated. Mechanistically, CD82-based TGF-ß1/Smad3 signaling leads to induction of CDK inhibitors and cell-cycle inhibition. The CD82 binding partner DARC/CD234 is expressed on macrophages and stabilizes CD82 on LT-HSCs, promoting their quiescence. When DARC(+) BM macrophages were ablated, the level of surface CD82 on LT-HSCs decreased, leading to cell-cycle entry, proliferation, and differentiation. A similar interaction appears to be relevant for human HSPCs. Thus, CD82 is a functional surface marker of LT-HSCs that maintains quiescence through interaction with DARC-expressing macrophages in the BM stem cell niche.


Asunto(s)
Sistema del Grupo Sanguíneo Duffy , Células Madre Hematopoyéticas , Proteína Kangai-1 , Macrófagos , Receptores de Superficie Celular , Animales , Femenino , Humanos , Masculino , Ratones , Sistema del Grupo Sanguíneo Duffy/metabolismo , Células Madre Hematopoyéticas/metabolismo , Proteína Kangai-1/biosíntesis , Proteína Kangai-1/deficiencia , Proteína Kangai-1/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Superficie Celular/metabolismo
12.
Cancer Res Treat ; 48(2): 676-86, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26511811

RESUMEN

PURPOSE: Epigenetic alterations of specific genes have recently been identified as diagnostic biomarkers for human cancers. However, there are currently no standardized epigenetic biomarkers for drug sensitivity in human gastrointestinal cancer. Therefore, the aim of this study is to identify a novel epigenetic biomarker in gastrointestinal cancer. MATERIALS AND METHODS: Using bisulfite sequencing and pyrosequencing analysis, DNA methylation patterns of gastric, colon primary tissues and their cancer cells were analyzed, and histone modifications were analyzed using chromatin immunoprecipitation assay. In addition, cancer cells were exposed to cisplatin and treated with a DNA methyltransferase inhibitor. RESULTS: We report that in human gastric and colon cancers, latrophilin 2 (LPHN2) is silenced by epigenetic modifications, including CpG island methylation and aberrant histone modifications. We also confirmed that LPHN2 was silenced by DNA hypermethylation in primary gastric and colon tumor tissues compared to their normal counterparts. Interestingly, we found that cancer cells with methylated LPHN2 showed higher sensitivity to cisplatin. Also, 5-aza- 2'-deoxycytidine combined with cisplatin decreased the cytotoxicity of cisplatin in cancer cells with methylated LPHN2. In addition, LPHN2 knockdown in cancer cells with high LPHN2 expression sensitized these cells to the anti-proliferative effects of cisplatin. CONCLUSION: In human gastrointestinal cancer, we found that LPHN2 is regulated by epigenetic modifications, and that cancer cells with lower LPHN2 expression show higher sensitivity to cisplatin. Therefore, the methylation status of LPHN2 is a potential novel epigenetic biomarker for cisplatin treatment in human gastric and colon cancers.


Asunto(s)
Cisplatino/uso terapéutico , Epigénesis Genética , Neoplasias Gastrointestinales/tratamiento farmacológico , Neoplasias Gastrointestinales/genética , Receptores Acoplados a Proteínas G/genética , Biomarcadores , Metilación de ADN , Humanos , Regiones Promotoras Genéticas
13.
Nucleic Acids Res ; 44(2): 558-72, 2016 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-26420833

RESUMEN

Gene amplification is a hallmark of cancer with chromosomal instability although the underlying mechanism by which altered copy numbers are maintained is largely unclear. Cohesin, involved in sister chromatid cohesion, DNA repair, cell cycle progression and transcriptional regulation of key developmental genes, is frequently overexpressed in human cancer. Here we show that cohesin-dependent change in DNA replication controls the copy numbers of amplified genes in cancer cells with chromosomal instability. We found that the down-regulation of elevated cohesin leads to copy number-associated gene expression changes without disturbing chromosomal segregation. Highly amplified genes form typical long-range chromatin interactions, which are stabilized by enriched cohesin. The spatial proximities among cohesin binding sites within amplified genes are decreased by RAD21-knockdown, resulting in the rapid decline of amplified gene expression. After several passages, cohesin depletion inhibits DNA replication initiation by reducing the recruitment of pre-replication complexes such as minichromosome maintenance subunits 7 (MCM7), DNA polymerase α, and CDC45 at replication origins near the amplified regions, and as a result, decreases the DNA copy numbers of highly amplified genes. Collectively, our data demonstrate that cohesin-mediated chromatin organization and DNA replication are important for stabilizing gene amplification in cancer cells with chromosomal instability.


Asunto(s)
Inestabilidad Cromosómica , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/genética , Fosfoproteínas/genética , Neoplasias Gástricas/genética , Sitios de Unión , Factor de Unión a CCCTC , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Cromátides/química , Cromátides/metabolismo , Cromatina/química , Cromatina/metabolismo , Segregación Cromosómica , Hibridación Genómica Comparativa , ADN Polimerasa I/genética , ADN Polimerasa I/metabolismo , Replicación del ADN , Proteínas de Unión al ADN , Dosificación de Gen , Células HCT116 , Células Hep G2 , Humanos , Componente 7 del Complejo de Mantenimiento de Minicromosoma/genética , Componente 7 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Fosfoproteínas/metabolismo , Unión Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
14.
J Surg Oncol ; 112(1): 93-7, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26074337

RESUMEN

BACKGROUND: A Korean version of Toronto Extremity Salvage Score (TESS), a widely used disease-specific patient-reported questionnaire for assessing physical function of sarcoma patients, has not been developed. OBJECTIVES: 1) to translate and cross-culturally adapt the TESS into Korean, and 2) to examine its comprehensibility, reliability and validity. METHODS: TESS was translated into Korean, then translated back into English, and reviewed by a committee to develop the consensus version of the Korean TESS. The Korean TESS was administered to 126 patients to examine its comprehensibility, reliability, and validity. RESULTS: Comprehensibility was high, as the patients rated questions as "easy" or "very easy" in 96% for the TESS lower extremity (LE) and in 97% for the TESS upper extremity (UE). Test-retest reliability with intraclass coefficient (0.874 for LE and 0.979 for UE) and internal consistency with Cronbach's alpha (0.978 for LE and 0.989 for UE) were excellent. Korean TESS correlated with the MSTS score (r = 0.772 for LE and r = 0.635 for UE), and physical functioning domain of EORTC-CLQ C30 (r = 0.840 for LE and r = 0.630 for UE). CONCLUSION: Our study suggests that Korean version of the TESS is a comprehensible, reliable, and valid instrument to measure patient-reported functional outcome in patients with extremity sarcoma.


Asunto(s)
Comparación Transcultural , Evaluación de la Discapacidad , Extremidades/cirugía , Terapia Recuperativa , Sarcoma/cirugía , Traducción , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Psicometría , República de Corea , Encuestas y Cuestionarios , Adulto Joven
15.
Am J Pathol ; 185(3): 666-78, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25576785

RESUMEN

Recent genome-wide epigenomic and transcription profiling studies have demonstrated that epigenetic silencing can encompass multiple neighboring genes, termed as long-range epigenetic silencing (LRES). Herein, we identified a novel LRES region by comparing gene expression of human colon cancer HCT116 cells with their DNA methyltransferase 1 and DNA methyltransferase 3B double-knockout derivative double-knockout cells. Ten consecutive genes spanning 3 Mb of chromosome 15q25 were coordinately silenced, with eight genes showing promoter CpG island hypermethylation and enrichment of repressive histone marks, which were evaluated by bisulfite sequencing analysis and chromatin immunoprecipitation assay. Comparison of primary gastric tumor specimens with normal tissue confirmed that the long-range silencing of this region was tumor specific. Methylation of genes within the LRES region was evaluated in 190 gastric tumor tissues using the MethyLight assay, and their association with clinicopathological features, such as older age, high-grade differentiation, and diffuse or mixed-type histology, was determined. LRES-positive gastric cancer patients (six or more methylated genes) showed lower recurrence and better survival. Our findings emphasize the differential dynamics of DNA methylation and histone modification, indicating the importance of studying the relationship of each epigenetic modification in the context of chromatin domains. Patients with LRES showed lower recurrence and better prognosis, indicating that stratifying patients according to underlying molecular features, such as LRES regions, may better predict recurrence and survival.


Asunto(s)
Cromosomas Humanos Par 15 , Epigénesis Genética , Silenciador del Gen , Recurrencia Local de Neoplasia/genética , Neoplasias Gástricas/genética , Adulto , Anciano , Islas de CpG , Metilación de ADN , Femenino , Histonas , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/mortalidad , Recurrencia Local de Neoplasia/patología , Regiones Promotoras Genéticas , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Tasa de Supervivencia , Adulto Joven
16.
Cardiovasc Res ; 104(1): 171-82, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25082847

RESUMEN

AIMS: From our previous clinical trials, intracoronary infusion of granulocyte-colony stimulating factor (G-CSF)-mobilized peripheral blood mononuclear cells ((mob)PBMCs) proved to be effective in improving myocardial contractility and reducing infarct volume in acute myocardial infarction. We tested the effect of priming (mob)PBMCs with erythropoietin (EPO) to augment its therapeutic efficacy. METHODS AND RESULTS: (mob)PBMCs were obtained from healthy volunteers after a 3-day subcutaneous injection of G-CSF (10 µg/kg). About 40% of (mob)PBMCs were EPO receptor (EPOR) (+) and responded to 6 h EPO-priming (10 IU/mL) by increasing the expression of vasculogenic factors (i.e. IL8, IL10, bFGF, PDGF, MMP9) and adhesion molecules (i.e. integrin αV, ß1, ß2, ß8) through the JAK2 and Akt pathway. These responses were also observed in PBMCs from elderly patients with coronary disease. The conditioned media from EPO-primed (mob)PBMCs contained various cytokines such as IL8, IL10, TNFα, and PDGF, which enhanced the migration and tube formation capability of endothelial cells. EPO-primed (mob)PBMCs also showed increased adhesion on endothelial cells or fibronectin. Augmented vasculogenic potential of EPO-primed (mob)PBMCs was confirmed in a Matrigel plug assay, ischaemic hindlimb, and myocardial infarction models of athymic nude mice. There were two action mechanisms: (i) cellular effects confirmed by direct incorporation of human (mob)PBSCs into mouse vasculature and (ii) indirect humoral effects confirmed by the therapeutic effect of the supernatant of EPO-primed (mob)PBMCs. CONCLUSION: Brief ex vivo EPO-priming is a novel method to augment the vasculogenic potential of human (mob)PBMCs, which would help to achieve better results after intracoronary infusion in myocardial infarction patients.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Eritropoyetina/farmacología , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Movilización de Célula Madre Hematopoyética/métodos , Leucocitos Mononucleares/efectos de los fármacos , Músculo Esquelético/irrigación sanguínea , Neovascularización Fisiológica/efectos de los fármacos , Proteínas Angiogénicas/metabolismo , Animales , Células de la Médula Ósea/metabolismo , Estudios de Casos y Controles , Adhesión Celular/efectos de los fármacos , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Enfermedad de la Arteria Coronaria/sangre , Medios de Cultivo Condicionados/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Miembro Posterior , Humanos , Inyecciones Subcutáneas , Isquemia/metabolismo , Isquemia/fisiopatología , Isquemia/cirugía , Janus Quinasa 2/metabolismo , Leucocitos Mononucleares/metabolismo , Leucocitos Mononucleares/trasplante , Masculino , Ratones Desnudos , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/cirugía , Trasplante de Células Madre de Sangre Periférica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes/farmacología , Regeneración , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
17.
J Mol Cell Cardiol ; 75: 64-75, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25016235

RESUMEN

Platelets not only play a role in hemostasis, but they also promote angiogenesis and tissue recovery by releasing various cytokines and making an angiogenic milieu. Here, we examined autologous 'activated platelet supernatant (APS)' as a priming agent for stem cells; thereby enhance their pro-angiogenic potential and efficacy of stem cell-based therapy for ischemic diseases. The mobilized peripheral blood stem cells ((mob)PBSCs) were isolated from healthy volunteers after subcutaneous injection of granulocyte-colony stimulating factor. APS was collected separately from the platelet rich plasma after activation by thrombin. (mob)PBSCs were primed for 6h before analysis. Compared to naive platelet supernatants, APS had a higher level of various cytokines, such as IL8, IL17, PDGF and VEGF. APS-priming for 6h induced (mob)PBSCs to express key angiogenic factors, surface markers (i.e. CD34, CD31, and CXCR4) and integrins (integrins α5, ß1 and ß2). Also (mob)PBSCs were polarized toward CD14(++)/CD16(+) pro-angiogenic monocytes. The priming effect was reproduced by an in vitro reconstruction of APS. Through this phenotype, APS-priming increased cell-cell adhesion and cell-extracellular matrix adhesion. The culture supernatant of APS-primed (mob)PBSCs contained high levels of IL8, IL10, IL17 and TNFα, and augmented proliferation and capillary network formation of human umbilical vein endothelial cells. In vivo transplantation of APS-primed (mob)PBSCs into athymic mice ischemic hindlimbs and Matrigel plugs elicited vessel differentiation and tissue repair. In safety analysis, platelet activity increased after mixing with (mob)PBSCs regardless of priming, which was normalized by aspirin treatment. Collectively, our data identify that APS-priming can enhance the angiogenic potential of (mob)PBSCs, which can be used as an adjunctive strategy to improve the efficacy of cell therapy for ischemic diseases.


Asunto(s)
Plaquetas/metabolismo , Células de la Médula Ósea/citología , Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Neovascularización Fisiológica/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Animales , Plaquetas/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Citocinas/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Biológicos , Comunicación Paracrina/efectos de los fármacos , Fenotipo , Regeneración/efectos de los fármacos , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Trombosis/patología
18.
Mol Ther ; 22(8): 1518-1529, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24736277

RESUMEN

Emerging studies suggested that murine podoplanin-positive monocytes (PPMs) are involved in lymphangiogenesis. The goal of this study was to demonstrate the therapeutic lymphangiogenesis of human PPMs by the interaction with platelets. Aggregation culture of human peripheral blood mononuclear cells (PBMCs) resulted in cellular aggregates termed hematospheres. During 5-day culture, PPMs expanded exponentially and expressed several lymphatic endothelial cell-specific markers including vascular endothelial growth factor receptor (VEGFR)-3 and well-established lymphangiogenic transcription factors. Next, we investigated the potential interaction of PPMs with platelets that had C-type lectin-like receptor-2 (CLEC-2), a receptor of podoplanin. In vitro coculture of PPMs and platelets stimulated PPMs to strongly express lymphatic endothelial markers and upregulate lymphangiogenic cytokines. Recombinant human CLEC-2 also stimulated PPMs through Akt and Erk signaling. Likewise, platelets in coculture with PPMs augmented secretion of a lymphangiogenic cytokine, interleukin (IL)-1-ß, via podoplanin/CLEC-2 axis. The supernatant obtained from coculture was able to enhance the migration, viability, and proliferation of lymphatic endothelial cell. Local injection of hematospheres with platelets significantly increased lymphatic neovascularization and facilitated wound healing in the full-thickness skin wounds of nude mice. Cotreatment with PPMs and platelets augments lymphangiogenesis through podoplanin/CLEC-2 axis, which thus would be a promising novel strategy of cell therapy to treat human lymphatic vessel disease.


Asunto(s)
Plaquetas/metabolismo , Técnicas de Cocultivo/métodos , Lectinas Tipo C/metabolismo , Linfangiogénesis , Glicoproteínas de Membrana/metabolismo , Monocitos/metabolismo , Animales , Tratamiento Basado en Trasplante de Células y Tejidos , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Desnudos , Transducción de Señal , Piel/lesiones , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
19.
Biomaterials ; 34(8): 1929-41, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23267826

RESUMEN

Ex vivo expansion of human circulating angiogenic cells is a major challenge in autologous cell therapy for ischemic diseases. Here, we demonstrate that hematosphere-derived CXCR4(+)CD31(+) myeloid cells using peripheral blood possess robust proangiogenic capacity such as formation of vessel-like structures and tip cell-like morphology in Matrigel. We also found that CD31 positive myeloid cells are principal cellular component of hematospheres by magnetic cell sorting. Flow cytometry analysis showed that fresh peripheral blood contained 40.3 ± 15.2% of CXCR4(+)CD31(+) myeloid cells, but at day 5 of hematosphere culture, most of myeloid cells were CXCR4(+)CD31(+) by 86.9 ± 5.4%. Hematosphere culture significantly increased the production of angiogenic niche-supporting cytokines. Moreover, CD31-homophilic interaction and VEGF-VEGF receptor loop signaling were essential for sphere formation and acquisition of angiogenic capacity in hematospheres. Matrigel plug and ischemic hindlimb model provide in vivo evidence that hematosphere-derived myeloid cells have highly vasculogenic capacities, participate in new and mature vessel formation, and exert therapeutic effects on ischemic hindlimb. In conclusion, our strategy for ex vivo expansion of human CXCR4(+)CD31(+) angiogenic cells using hematospheres provides an autologous therapeutic cell source for ischemic diseases and a new model for investigating the microenvironment of angiogenesis.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Monocitos/citología , Monocitos/metabolismo , Neovascularización Fisiológica , Receptores CXCR4/metabolismo , Adulto , Animales , Células Cultivadas , Microambiente Celular/efectos de los fármacos , Colágeno/farmacología , Combinación de Medicamentos , Miembro Posterior/irrigación sanguínea , Miembro Posterior/patología , Humanos , Isquemia/patología , Isquemia/terapia , Laminina/farmacología , Receptores de Lipopolisacáridos/metabolismo , Ratones , Ratones Desnudos , Monocitos/efectos de los fármacos , Monocitos/trasplante , Neovascularización Fisiológica/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Proteoglicanos/farmacología , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Transducción de Señal/efectos de los fármacos , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
20.
Lab Invest ; 92(7): 1033-44, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22508389

RESUMEN

Epiregulin (EREG) induces cell growth by binding to the epidermal growth factor receptor (EGFR). Expression of EREG affects sensitivity to cetuximab a chimeric monoclonal antibody that inhibits the EGFR signaling pathway. The mechanism through which EREG is regulated is largely unknown, but a methyl-array study previously performed by our group revealed that EREG is methylated in gastric cancer cells. In this study, we found that EREG gene expression was low in 7 out of 11 gastric cancer cells and this downregulation was mediated by aberrant CpG methylation of the EREG promoter. Treatment with 5-aza-CdR restored EREG expression and demethylated CpG sites in the EREG promoter. Compared with DNA methyltransferase 1 (DNMT1), knock-down of DNA methyltransferase 3b (DNMT3b) significantly increased the expression of EREG and led to the demethylation of specific CpG sites in the EREG promoter, suggesting that DNMT3b primarily regulates CpG methylation and silencing of the EREG gene. EREG methylation was observed in 30% (4/13) of human primary gastric tumor tissues we evaluated. In addition to DNA methylation, results from a chromatin immunoprecipitation assay demonstrated that transcriptional levels of EREG were associated with the enrichment of active histone marks (H3K4me3 and AcH3) and of a repressive mark (H3K27me2). Treatment with 5-aza-CdR dynamically increased the low occupancy of H3K4me3 and AcH3, while decreasing the high enrichment of H3K27me2, indicating that dynamic histone modifications contribute to EREG regulation in addition to DNA methylation. Finally, the combination of 5-aza-CdR and cetuximab exerted a synergistic anti-proliferative effect on gastric cancer cells. Taken together, the results of our study showed for the first time that EREG is epigenetically silenced in gastric cancer cells by aberrant DNA methylation and histone modification.


Asunto(s)
Factor de Crecimiento Epidérmico/antagonistas & inhibidores , Factor de Crecimiento Epidérmico/genética , Epigénesis Genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Azacitidina/administración & dosificación , Azacitidina/análogos & derivados , Azacitidina/farmacología , Línea Celular Tumoral , Cetuximab , Islas de CpG , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , Decitabina , Epigénesis Genética/efectos de los fármacos , Epirregulina , Receptores ErbB/metabolismo , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Histonas/genética , Histonas/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Regiones Promotoras Genéticas , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , ARN Interferente Pequeño/genética , Neoplasias Gástricas/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto , ADN Metiltransferasa 3B
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA